WO2016131098A1 - Sulfonylureas and related compounds and use of same - Google Patents

Sulfonylureas and related compounds and use of same Download PDF

Info

Publication number
WO2016131098A1
WO2016131098A1 PCT/AU2016/050103 AU2016050103W WO2016131098A1 WO 2016131098 A1 WO2016131098 A1 WO 2016131098A1 AU 2016050103 W AU2016050103 W AU 2016050103W WO 2016131098 A1 WO2016131098 A1 WO 2016131098A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
disease
alkyl
mmol
ring
Prior art date
Application number
PCT/AU2016/050103
Other languages
French (fr)
Other versions
WO2016131098A8 (en
Inventor
Luke O'neill
Rebecca Coll
Matt Cooper
Avril Robertson
Kate SCHRODER
Original Assignee
The University Of Queensland
The Provost, Fellows, Foundation Scholars, And The Other Members Of Board, Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth Near Dublin
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=56691944&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2016131098(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from AU2015900507A external-priority patent/AU2015900507A0/en
Priority to EP19187141.7A priority Critical patent/EP3578547B1/en
Priority to MDE20180009T priority patent/MD3259253T2/en
Priority to LTEP16751821.6T priority patent/LT3259253T/en
Priority to EP21152421.0A priority patent/EP3888749A1/en
Priority to US15/551,264 priority patent/US10538487B2/en
Priority to EP16751821.6A priority patent/EP3259253B1/en
Priority to SI201630635T priority patent/SI3259253T1/en
Priority to SG11201706664QA priority patent/SG11201706664QA/en
Application filed by The University Of Queensland, The Provost, Fellows, Foundation Scholars, And The Other Members Of Board, Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth Near Dublin filed Critical The University Of Queensland
Priority to RS20200258A priority patent/RS60048B1/en
Priority to PL19187141T priority patent/PL3578547T3/en
Priority to PE2021001430A priority patent/PE20221627A1/en
Priority to KR1020177025608A priority patent/KR20170109678A/en
Priority to MX2017010528A priority patent/MX2017010528A/en
Priority to CA2975192A priority patent/CA2975192A1/en
Priority to NZ733948A priority patent/NZ733948A/en
Priority to ES16751821T priority patent/ES2777626T3/en
Priority to IL273065A priority patent/IL273065B2/en
Priority to MEP-2020-49A priority patent/ME03737B/en
Priority to CN201680010446.XA priority patent/CN107428696B/en
Priority to MA41553A priority patent/MA41553B1/en
Priority to DK16751821.6T priority patent/DK3259253T3/en
Priority to IL253661A priority patent/IL253661B2/en
Priority to MYPI2017001188A priority patent/MY193765A/en
Priority to JP2017560843A priority patent/JP6929792B2/en
Priority to AU2016222278A priority patent/AU2016222278B2/en
Priority to BR112017017610-6A priority patent/BR112017017610A2/en
Priority to RU2017128287A priority patent/RU2739356C2/en
Priority to PL16751821T priority patent/PL3259253T3/en
Publication of WO2016131098A1 publication Critical patent/WO2016131098A1/en
Publication of WO2016131098A8 publication Critical patent/WO2016131098A8/en
Priority to HK18107901.3A priority patent/HK1249501A1/en
Priority to US16/535,002 priority patent/US11130731B2/en
Priority to HRP20200214TT priority patent/HRP20200214T1/en
Priority to CY20201100252T priority patent/CY1122832T1/en
Priority to AU2020203464A priority patent/AU2020203464B2/en
Priority to US17/405,989 priority patent/US20220112159A1/en
Priority to AU2021258033A priority patent/AU2021258033A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/84Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/50Compounds containing any of the groups, X being a hetero atom, Y being any atom
    • C07C311/52Y being a hetero atom
    • C07C311/54Y being a hetero atom either X or Y, but not both, being nitrogen atoms, e.g. N-sulfonylurea
    • C07C311/56Y being a hetero atom either X or Y, but not both, being nitrogen atoms, e.g. N-sulfonylurea having sulfur atoms of the sulfonylurea groups bound to carbon atoms of rings other than six-membered aromatic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/18Sulfonamides
    • 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
    • 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
    • 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
    • A61K31/41921,2,3-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/64Sulfonylureas, e.g. glibenclamide, tolbutamide, chlorpropamide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/10Anti-acne agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/06Antigout agents, e.g. antihyperuricemic or uricosuric agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/24Antidepressants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • 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]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/10Anthelmintics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C311/00Amides of sulfonic acids, i.e. compounds having singly-bound oxygen atoms of sulfo groups replaced by nitrogen atoms, not being part of nitro or nitroso groups
    • C07C311/50Compounds containing any of the groups, X being a hetero atom, Y being any atom
    • C07C311/52Y being a hetero atom
    • C07C311/54Y being a hetero atom either X or Y, but not both, being nitrogen atoms, e.g. N-sulfonylurea
    • C07C311/57Y being a hetero atom either X or Y, but not both, being nitrogen atoms, e.g. N-sulfonylurea having sulfur atoms of the sulfonylurea groups bound to carbon atoms of six-membered aromatic rings
    • C07C311/60Y being a hetero atom either X or Y, but not both, being nitrogen atoms, e.g. N-sulfonylurea having sulfur atoms of the sulfonylurea groups bound to carbon atoms of six-membered aromatic rings having nitrogen atoms of the sulfonylurea groups bound to carbon atoms of six-membered aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/30Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members
    • C07D207/34Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/36Oxygen or sulfur atoms
    • C07D207/382-Pyrrolones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/70Sulfur atoms
    • C07D213/71Sulfur atoms to which a second hetero atom is attached
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/72Nitrogen atoms
    • C07D213/74Amino or imino radicals substituted by hydrocarbon or substituted hydrocarbon radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/36Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/02Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with only hydrogen atoms or radicals containing only carbon and hydrogen atoms, directly attached to carbon atoms of the nitrogen-containing ring; Alkylene-bis-isoquinolines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the nitrogen-containing ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D217/00Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems
    • C07D217/22Heterocyclic compounds containing isoquinoline or hydrogenated isoquinoline ring systems with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the nitrogen-containing ring
    • C07D217/24Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/04Ortho- or peri-condensed ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D221/00Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00
    • C07D221/02Heterocyclic compounds containing six-membered rings having one nitrogen atom as the only ring hetero atom, not provided for by groups C07D211/00 - C07D219/00 condensed with carbocyclic rings or ring systems
    • C07D221/04Ortho- or peri-condensed ring systems
    • C07D221/18Ring systems of four or more rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/18One oxygen or sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/10Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D241/14Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D241/24Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/36Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems
    • C07D241/38Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems with only hydrogen or carbon atoms directly attached to the ring nitrogen atoms
    • C07D241/40Benzopyrazines
    • C07D241/42Benzopyrazines with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/36Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems
    • C07D241/38Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings condensed with carbocyclic rings or ring systems with only hydrogen or carbon atoms directly attached to the ring nitrogen atoms
    • C07D241/40Benzopyrazines
    • C07D241/44Benzopyrazines with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/041,2,3-Triazoles; Hydrogenated 1,2,3-triazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/041,2,3-Triazoles; Hydrogenated 1,2,3-triazoles
    • C07D249/061,2,3-Triazoles; Hydrogenated 1,2,3-triazoles with aryl radicals directly attached to ring atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • C07D249/101,2,4-Triazoles; Hydrogenated 1,2,4-triazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D249/12Oxygen or sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D261/00Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings
    • C07D261/02Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings
    • C07D261/06Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members
    • C07D261/10Heterocyclic compounds containing 1,2-oxazole or hydrogenated 1,2-oxazole rings not condensed with other rings having two or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D261/18Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/12Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/36Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/04Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D307/18Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/64Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/34Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D307/56Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/68Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/77Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D307/78Benzo [b] furans; Hydrogenated benzo [b] furans
    • C07D307/82Benzo [b] furans; Hydrogenated benzo [b] furans with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D309/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings
    • C07D309/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D309/08Heterocyclic compounds containing six-membered rings having one oxygen atom as the only ring hetero atom, not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/06Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2
    • C07D311/08Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring
    • C07D311/16Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring substituted in position 7
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/06Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2
    • C07D311/08Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring
    • C07D311/18Benzo[b]pyrans, not hydrogenated in the carbocyclic ring with oxygen or sulfur atoms directly attached in position 2 not hydrogenated in the hetero ring substituted otherwise than in position 3 or 7
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D311/00Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings
    • C07D311/02Heterocyclic compounds containing six-membered rings having one oxygen atom as the only hetero atom, condensed with other rings ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D311/04Benzo[b]pyrans, not hydrogenated in the carbocyclic ring
    • C07D311/58Benzo[b]pyrans, not hydrogenated in the carbocyclic ring other than with oxygen or sulphur atoms in position 2 or 4
    • C07D311/60Benzo[b]pyrans, not hydrogenated in the carbocyclic ring other than with oxygen or sulphur atoms in position 2 or 4 with aryl radicals attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/44Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D317/46Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D317/48Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring
    • C07D317/62Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to atoms of the carbocyclic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/02Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings
    • C07D333/04Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom
    • C07D333/26Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom not condensed with other rings not substituted on the ring sulphur atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D333/30Hetero atoms other than halogen
    • C07D333/34Sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/50Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D333/52Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/50Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D333/52Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes
    • C07D333/54Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D333/00Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom
    • C07D333/50Heterocyclic compounds containing five-membered rings having one sulfur atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • C07D333/52Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes
    • C07D333/62Benzo[b]thiophenes; Hydrogenated benzo[b]thiophenes with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/12Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D493/00Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system
    • C07D493/02Heterocyclic compounds containing oxygen atoms as the only ring hetero atoms in the condensed system in which the condensed system contains two hetero rings
    • C07D493/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/06Peri-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/02Systems containing only non-condensed rings with a three-membered ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/08Systems containing only non-condensed rings with a five-membered ring the ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/14The ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/12Systems containing only non-condensed rings with a six-membered ring
    • C07C2601/16Systems containing only non-condensed rings with a six-membered ring the ring being unsaturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/04One of the condensed rings being a six-membered aromatic ring
    • C07C2602/08One of the condensed rings being a six-membered aromatic ring the other ring being five-membered, e.g. indane
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/04One of the condensed rings being a six-membered aromatic ring
    • C07C2602/10One of the condensed rings being a six-membered aromatic ring the other ring being six-membered, e.g. tetraline
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/02Ortho- or ortho- and peri-condensed systems
    • C07C2603/04Ortho- or ortho- and peri-condensed systems containing three rings
    • C07C2603/06Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members
    • C07C2603/10Ortho- or ortho- and peri-condensed systems containing three rings containing at least one ring with less than six ring members containing five-membered rings
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to the field of medical treatment and diagnosis of disease. More particularly, this invention relates to novel sulfonylurea and related compounds and their use in treating, or identifying a disease or condition responsive to modulation of NLRP3 or inhibition of the activation of NLRP3 or related components of the inflammatory process.
  • NLR NOD-like receptor
  • NLRP3 pyrin domain-containing protein 3
  • NLRP3 is an intracellular signalling molecule that senses many pathogen-derived, environmental and host-derived factors. Upon activation, NLRP3 binds to apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC). ASC then polymerises to form a large aggregate known as an ASC speck. Polymerised ASC in turn interacts with the cysteine protease caspase-1 to form a complex termed the inflammasome. This results in the activation of caspase-1 , which cleaves the proinflammatory cytokines IL-1 ⁇ and IL-18 to their active forms and mediates a type of inflammatory cell death known as pyroptosis. The ASC speck can also recruit and activate caspase-8, which can process pro-IL-1 ⁇ and pro-IL-18 and trigger apoptotic cell death.
  • ASC caspase activation and recruitment domain
  • Caspase-1 cleaves pro-IL-1 ⁇ and pro-IL-18 to their active forms, which are secreted from the cell. Active caspase-1 also cleaves gasdermin-D to trigger pyroptosis. Through its control of the pyroptotic cell death pathway, caspase-1 also mediates the release of alarm in molecules such as IL-33 and high mobility group box 1 protein (HMGB1 ). Caspase-1 also cleaves intracellular IL-1 R2 resulting in its degradation and allowing the release of IL- 1 a. In human cells caspase-1 may also control the processing and secretion of IL-37. A number of other caspase-1 substrates such as components of the cytoskeleton and glycolysis pathway may contribute to caspase-1 -dependent inflammation.
  • NLRP3-dependent ASC specks are released into the extracellular environment where they can activate caspase-1 , induce processing of caspase- 1 substrates and propagate inflammation.
  • Active cytokines derived from NLRP3 inflammasome activation are important drivers of inflammation and interact with other cytokine pathways to shape the immune response to infection and injury.
  • I L-1 ⁇ signalling induces the secretion of the pro-inflammatory cytokines IL-6 and TNF.
  • IL-1 ⁇ and I L- 8 synergise with IL-23 to induce I L- 7 production by memory CD4 Th17 cells and by ⁇ T cells in the absence of T cell receptor engagement.
  • IL- 18 and IL-12 also synergise to induce IFN- ⁇ production from memory T cells and NK cell driving a Th1 response.
  • PRRs intracellular pattern recognition receptors
  • NLRs nuclear-binding protein receptors
  • NLRP1 and NLRC4 are also capable of forming inflammasomes.
  • NLRP1 and NLRC4 are also capable of forming inflammasomes.
  • non-NLR PRRs such as the double- stranded DNA (dsDNA) sensors absent in melanoma 2 (AIM2) and interferon, gamma inducible protein 16 (IFI16).
  • dsDNA double- stranded DNA
  • AIM2 interferon, gamma inducible protein 16
  • IFI16 interferon, gamma inducible protein 16
  • NLRP3-dependent IL-1 ⁇ processing can also be activated by an indirect, non-canonical pathway downstream of caspase-1 1 .
  • NLRP3 The inherited CAPS diseases Muckle-Wells syndrome (MWS), familial cold autoinflammatory syndrome and neonatal-onset multisystem inflammatory disease are caused by gain-of-function mutations in NLRP3, thus defining NLRP3 as a critical component of the inflammatory process.
  • NLRP3 has also been implicated in the pathogenesis of a number of complex diseases, notably including metabolic disorders such as type 2 diabetes, atherosclerosis, obesity and gout.
  • NLRP3 A role for NLRP3 in diseases of the central nervous system is emerging, and lung diseases have also been shown to be influenced by NLRP3. Furthermore, NLRP3 has a role in the development of liver disease, kidney disease and aging. Many of these associations were defined using Nlrp3 mice, but there have also been insights into the specific activation of NLRP3 in these diseases. In type 2 diabetes, the deposition of islet amyloid polypeptide in the pancreas activates NLRP3 and I L- ⁇ signaling, resulting in cell death and inflammation.
  • NLRP3 inflammasome Several small molecules have been shown to inhibit the NLRP3 inflammasome. Glyburide inhibits IL-1 ⁇ production at micromolar concentrations in response to the activation of NLRP3 but not NLRC4 or NLRP1 .
  • Other previously characterised NLRP3 inhibitors include parthenolide, 3,4-methylenedioxy-P-nitrostyrene and dimethyl sulfoxide (DMSO), although these agents have limited potency and are nonspecific
  • NLRP3-related diseases include biologic agents that target IL-1 . These are the recombinant IL-1 receptor antagonist anakinra, the neutralizing IL-1 ⁇ antibody canakinumab and the soluble decoy IL-1 receptor rilonacept. These approaches have proven successful in the treatment of CAPS, and these biologic agents have been used in clinical trials for other IL- ⁇ -associated diseases.
  • NLRP3 inflammasome Several small molecules have been shown to inhibit the NLRP3 inflammasome. Glyburide inhibits IL- ⁇ production at micromolar concentrations in response to the activation of NLRP3 but not NLRC4 or NLRP1 .Other previously characterised NLRP3 inhibitors include parthenolide, 3,4-methylenedioxy-P-nitrostyrene and dimethyl sulfoxide (DMSO), although these agents have limited potency and are nonspecific.
  • DMSO dimethyl sulfoxide
  • cytokine release inhibitory drugs CRIDs
  • CRIDs are a class of diarylsulfonylurea containing compounds that inhibit the post-translational processing of IL-1 ⁇ . Post-translational processing of IL-1 ⁇ is accompanied by activation of caspase-1 and cell death. CRIDs arrest activated monocytes so that caspase-1 remains inactive and plasma membrane latency is preserved.
  • Ri is selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted;
  • R 2 is selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted; and both Ri is directly bonded to J and R 2 is directly bonded to the adjacent nitrogen, via a carbon atom .
  • a pharmaceutical composition comprising a compound of the first aspect, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier, diluent and/or excipient.
  • a third aspect of the invention resides in a method of treatment or prevention of a disease, disorder or condition including the step of administering an effective amount of a compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof, or the pharmaceutical composition of the second aspect to thereby treat or prevent the disease disorder or condition.
  • a fourth aspect of the invention provides for a compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof, or the pharmaceutical composition of the second aspect for use in the treatment or prevention of a disease, disorder or condition.
  • a fifth aspect of the invention provides for use of a compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof, in the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition.
  • the disease, disorder or condition is responsive to inhibition of activation of the NLRP3 inflammasome.
  • the disease, disorder or condition is a disease, disorder or condition of the immune system, the cardiovascular system, the endocrine system, the gastrointestinal tract, the renal system, the respiratory system, the central nervous system, is a cancer or other malignancy and/or is caused by or associated with a pathogen.
  • a method of diagnosing a disease, disorder or condition in a mammal including the step of administering a labelled compound of formula (I), (la), (lb), (Ic) or (I I), or a pharmaceutically effective salt, solvate or prodrug thereof, to the mammal or to a biological sample obtained from the mammal to facilitate diagnosis of the disease disorder or condition in the mammal.
  • a seventh aspect of the invention resides in a method of modulating the activity of a biological target comprising the step of exposing the biological target to a compound of the first aspect, or a pharmaceutically acceptable salt thereof.
  • the biological target may be selected from the group consisting of the NLRP3 inflammasome, ⁇ _-1 ⁇ , IL-17, IL-18, IL-1 a, IL-37, IL-33 and Th17 cells.
  • FIG 1 A to 1 C is a series of graphical representations of the plasma concentrations of a known sulfonylurea (MCC950) following different dosing levels in mice; and
  • FIG 2A to 2C is a series of graphical representations of the plasma concentrations of a sulfonylurea of the present invention (MCC7840) following different dosing levels in mice.
  • the present invention is predicated, at least in part, on the finding that certain sulfonyl ureas and related compounds have advantageous properties and show useful activity in the inhibition of activation of the NLRP3 inflammasome and/or inhibition of IL-1 ⁇ and/or IL-17 and/or IL-18, and/or IL-1 , and/or IL-37, and/or IL-33 as well as interfere with or modulate the activity of T helper cells such as Th17.
  • the compounds of the invention are useful in the treatment of a wide range of disorders in which the inflammation process, or the NLRP3 inflammasome and/or IL-1 ⁇ and/or IL-17 and/or IL-18, and/or IL-1 a, and/or IL-37, and/or IL-33 and/or Th17 cells play a part.
  • NLRP3 inhibition may block all processes downstream of NLRP3, including ASC speck formation and caspase-8 and caspase-1 activation. Consequently, NLRP3 inhibition will block all caspase-1 dependent processes such as IL-1 ⁇ , IL-18 and IL-37 processing and secretion, gasdermin D cleavage, pyroptosis, and release of IL-1 a, IL-33 and HMGB. Furthermore, NLRP3-dependent extracellular release of the ASC speck will be blocked, and caspase-8-dependent pro-IL-1 ⁇ and pro-IL-18 cleavage and apoptotic cell death will be prevented. Thus, specific inhibition of NLRP3 by compounds of the first aspect will prevent multiple downstream inflammatory signals and should therefore prove more effective anti-inflammatory therapy than IL-1 blockade alone.
  • Anti-IL-1 biologies block IL-1 derived from NLRP3-independent sources, such IL-1 produced by other inflammasomes (e.g. NLRC4, NLRP1 , NLRP6, AIM2) and IL-1 generated by the latter pathways may be important for host defence against pathogens.
  • IL-1 /IL-1 R antagonists exhibit increased incidence of upper airway infections.
  • Specific inhibition of NLRP3 by the present compounds may thus exert less generalised immunosuppression compared to anti-IL-1 biologies.
  • IL-1 ⁇ and IL-18, generated by the Nlrp3/caspase-1 axis play critical roles in driving IL-17 production by CD4 Th17 cells and ⁇ T cells.
  • IL-1 ⁇ and IL- 18 synergise with IL-23 to induce IL-17 production by memory CD4 Th17 cells and by ⁇ T cells in the absence of TCR engagement.
  • IL-1 -driven IL-17 has also been implicated in psoriasis, type I diabetes, rheumatoid arthritis, type 2 diabetes mellitus, atherosclerosis, obesity, gout, and recently, asthma.
  • each of these diseases has been shown to involve the activation of tissue macrophages, dendritic cells, or brain microglia, driven by either soluble alarmins, or the frustrated phagocytosis of metabolites that accumulate extracellularly.
  • NLRP3 senses these events, leading to IL-1 release, triggering inflammation to clear the offensive material. Disease will result if this process becomes chronic or over-activated, which explains why so many diseases have been shown to involve NLRP3.
  • Inhibitors that act to prevent NLRP3 activation hence can have utility in IL-17 driven, as well as IL-1 driven diseases.
  • pharmaceutically acceptable salt refers to salts which are toxicologically safe for systemic or localised administration such as salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids.
  • the pharmaceutically acceptable salts may be selected from the group including alkali and alkali earth, ammonium, aluminium, iron, amine, glucosamine, chloride, sulphate, sulphonate, bisulphate, nitrate, citrate, tartrate, bitarate, phosphate, carbonate, bicarbonate, malate, maleate, napsylate, fumarate, succinate, acetate, benzoate, terephthalate, palmoate, piperazine, pectinate and S-methyl methionine salts and the like.
  • alkyl refers to a straight-chain or branched alkyl substituent containing from, for example, 1 to about 12 carbon atoms, preferably 1 to about 9 carbon atoms, more preferably 1 to about 6 carbon atoms, even more preferably from 1 to about 4 carbon atoms, still yet more preferably from 1 to 2 carbon atoms.
  • substituents may be selected from the group consisting of methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, terf-butyl, pentyl, isoamyl, 2-methylbutyl, 3-methylbutyl, hexyl, heptyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2-ethylbutyl, 3- ethylbutyl, octyl, nonyl, decyl, undecyl, dodecyl and the like.
  • the number of carbons referred to relates to the carbon backbone and carbon branching but does not include carbon atoms belonging to any substituents, for example the carbon atoms of an alkoxy substituent branching off the main carbon chain.
  • Substituted alkyl includes alkyl substituted with one or more moieties selected from the group consisting of halo ⁇ e.g., CI, F, Br, and I); halogenated alkyl ⁇ e.g., CF 3 , 2-Br-ethyl, CH 2 F, CH 2 CI, CH 2 CF 3 , or CF 2 CF 3 ); hydroxyl; amino; carboxylate; carboxamido; alkylamino; arylamino; alkoxy; aryloxy; nitro; azido; cyano; thio; sulfonic acid; sulfate; phosphonic acid; phosphate; and phosphonate as well as those described under the definition of Optionally substituted'.
  • alkenyf refers to optionally substituted unsaturated linear or branched hydrocarbon groups, having 2 to 12 carbon atoms, preferably 2 to 9 carbon atoms, more preferably 2 to 6 carbon atoms and having at least one carbon-carbon double bond.
  • the alkenyl group may have a specified number of carbon atoms, for example, C2-C6 alkenyl which includes alkenyl groups having 2, 3, 4, 5 or 6 carbon atoms in linear or branched arrangements.
  • the number of carbons referred to relates to the carbon backbone and carbon branching but does not include carbon atoms belonging to any substituents.
  • substituents may be selected from the group consisting of ethenyl, propenyl, isopropenyl, butenyl, s- and t-butenyl, pentenyl, hexenyl, hept-l,3-diene, hex-l,3-diene, non-l,3,5-triene and the like.
  • Substituted alkenyl includes alkenyl substituted with one or more moieties selected from the group consisting of halo ⁇ e.g., CI, F, Br, and I); halogenated alkyl ⁇ e.g., CF 3 , 2-Br-ethyl, CH 2 F, CH 2 CI, CH 2 CF 3 , or CF 2 CF 3 ); hydroxyl; amino; carboxylate; carboxamido; alkylamino; arylamino; alkoxy; aryloxy; nitro; azido; cyano; thio; sulfonic acid; sulfate; phosphonic acid; phosphate; and phosphonate as well as those described under the definition of Optionally substituted'.
  • halo ⁇ e.g., CI, F, Br, and I
  • halogenated alkyl ⁇ e.g., CF 3 , 2-Br-ethyl, CH 2 F, CH 2
  • alkoxy as used herein means straight or branched chain alkyl groups linked by an oxygen atom (i.e., -O-alkyl), wherein alkyl is as described above.
  • alkoxy refers to oxygen-linked groups comprising 1 to 10 carbon atoms ("C1 -10 alkoxy”).
  • alkoxy refers to oxygen-linked groups comprising 1 to 8 carbon atoms ("C1 -8 alkoxy"), 1 to 6 carbon atoms (“C 1 -6 alkoxy”), 1 to 4 carbon atoms (“C1 -4 alkoxy") or 1 to 3 carbon atoms ("C1 -3 alkoxy').
  • cycloalkyi and cycloalkenyl refers to optionally substituted saturated and unsaturated mono-cyclic, bicyclic or tricyclic carbon groups.
  • the cycloalkyi or cycloalkenyl group may have a specified number of carbon atoms, for example, C3-C6 cycloalkyi or cycloalkenyl includes within its scope a carbocyclic group having 3, 4, 5 or 6 carbon atoms.
  • substituents may be selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl and the like.
  • Substituted cycloalkyl or cycloalkenyl includes substitutions with one or more moieties selected from the group consisting of halo (e.g. , CI, F, Br, and I); halogenated alkyl (e.g. , CF 3 , 2-Br-ethyl, CH 2 F,
  • alkylthio as used herein means a thio group with one or more alkyl substituents, where alkyl is defined as above.
  • amino as used herein means a moiety represented by the structure NR 23 , and includes primary amines, and secondary and tertiary amines substituted by alkyl (i.e., alkylamino).
  • R 23 may represent, for example, two hydrogen atoms, two alkyl moieties, or one hydrogen atom and one alkyl moiety.
  • 'aryi refers to a stable monocyclic, bicyclic, or tricyclic carbon ring of up to 8 members in each ring, wherein at least one ring is aromatic as defined by the HCickel 4n+2 rule.
  • the term includes polycyclic systems comprising saturated carbon rings or heteroaryl or heterocyclic groups so long as at least one ring is aryl, as described.
  • aralkyl and "arylalkyl” as used herein mean an aryl group as defined above linked to the molecule through an alkyl group as defined above.
  • heteroaryl refers to an aryl group containing from one or more (particularly one to four) non-carbon atom(s) (particularly N, O or S) or a combination thereof, which heteroaryl group is optionally substituted at one or more carbon or nitrogen atom(s).
  • Heteroaryl rings may also be fused with one or more cyclic hydrocarbon, heterocyclic, aryl, or heteroaryl rings.
  • Heteroaryl includes, but is not limited to, 5-membered heteroaryls having one hetero atom (e.g.
  • heteroaryls having two heteroatoms in 1 ,2 or 1 ,3 positions (e.g., oxazoles, pyrazoles, imidazoles, thiazoles, purines); 5-membered heteroaryls having three heteroatoms (e.g., triazoles, thiadiazoles); 5-membered heteroaryls having four heteroatoms (e.g., tetrazoles); 6-membered heteroaryls with one heteroatom (e.g., pyridine, quinoline, isoquinoline, phenanthrine, 5,6-cycloheptenopyridine); 6-membered heteroaryls with two heteroatoms (e.g., pyridazines, cinnolines, phthalazines, pyrazines, pyrimidines, quinazolines); 6-membered heretoaryls with three heteroatoms (e.g.,
  • Heterocyclyl refers to a non-aromatic ring having 5 to 8 atoms in the ring and of those atoms 1 to 4 are heteroatoms. Heterocyclic rings may also be fused with one or more cyclic hydrocarbon, heterocyclic, aryl, or heteroaryl rings. Heterocyclic includes partially and fully saturated heterocyclic groups. Heterocyclic systems may be attached to another moiety via any number of carbon atoms or heteroatoms of the radical and may be both saturated and unsaturated.
  • heterocyclic include C 4 -C 6 selenocycles, pyrrolidinyl, pyrrolinyl, pyranyl, piperidinyl, piperazinyl, morpholinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrazolinyl, dithiolyl, oxathiolyl, dioxanyl, dioxinyl, oxazinyl, azepinyl, diazepinyl, thiazepinyl, oxepinyl and thiapinyl, imidazolinyl, thiomorpholinyl, and the like.
  • Optionally substituted in reference to a substituent group refers to substituent groups optionally substituted with one or more moieties, for example, those selected from the group consisting of optionally substituted C1 - 10 alkyl (e.g., optionally substituted C1 -6 alkyl); optionally substituted C3-6 cycloalkyl (e.g., optionally substituted cyclopropyl); optionally substituted hydroxyalkyl; optionally substituted C1 -10 alkoxy (e.g., optionally substituted C1 -6 alkoxy); optionally substituted C2-10 alkenyl; optionally substituted C2-10 alkynyl; optionally substituted C6-C12 aryl; aryloxy; optionally substituted heteroaryl; optionally substituted heterocyclyl; halo (e.g., CI, F, Br, and I); hydroxyl; halogenated alkyl (e.g., CF 3 , 2-Br-ethy
  • a range of the number of atoms in a structure is indicated (e.g. , a Ci -Ci 2 , C-i-C-io, C1-C9, C1-C6, Ci-C 4 , or C 2 -C 2 o, C 2 -C-
  • d-C-i 2 1 -9 carbon atoms (e.g. , C1 -C9), 1 -6 carbon atoms (e.g. , C-1 -C6), 1 -4 carbon atoms (e.g. , Ci-C 4 ), 1 -3 carbon atoms (e.g., Ci-C 3 ), or 2-8 carbon atoms (e.g., C 2 -C 8 ) as used with respect to any chemical group (e.g.
  • alkyl, etc. referenced herein encompasses and specifically describes 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , and/or 12 carbon atoms, as appropriate, as well as any sub-range thereof (e.g., 1 -2 carbon atoms, 1 -3 carbon atoms, 1 -4 carbon atoms, 1 -5 carbon atoms, 1 -6 carbon atoms, 1 -7 carbon atoms, 1 -8 carbon atoms, 1 -9 carbon atoms, 1 -10 carbon atoms, 1 -1 1 carbon atoms, 1 -12 carbon atoms, 2-3 carbon atoms, 2-4 carbon atoms, 2-5 carbon atoms, 2-6 carbon atoms, 2-7 carbon atoms, 2-8 carbon atoms, 2-9 carbon atoms, 2-10 carbon atoms, 2-1 1 carbon atoms, 2-12 carbon atoms, 3-4 carbon atoms, 3-5 carbon atoms, 3-6 carbon atoms, 3-7 carbon atoms, 3-8 carbon atoms, 3-5
  • R-i is selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted;
  • R 2 is selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted; and both R-i is directly bonded to J and R 2 is directly bonded to the adjacent nitrogen, via a carbon atom.
  • W is O.
  • J is S.
  • Ri is selected from the group consisting of C5 or C 6 cycloalkyl, 5-membered or 6-membered heteroaryl, bicyclic heteroaryl wherein at least one ring is heteroaryl, phenyl, biphenyl, phenylheterocyclyl, 5- membered or 6-membered heterocyclyl, and heterocyclylcycloalkyl, all of which may be optionally substituted.
  • W is O
  • J is S
  • R-i is selected from the group consisting of pyrazole, furan, tetrahydrofuran, tetrahydropyran, pyran, pyrrolidine, pyrrole, triazole, tetrazole, imidazole, pyridine, morpholine, piperazine, piperidine, substituted phenyl, phenylheteroaryl, phenylheterocyclyl, biphenyl, quinoline, isoquinoline, naphthyl, pyrazine and pyrimidine, all of which may be optionally substituted as appropriate.
  • W is O
  • J is S
  • R-i 2-furan or 2- thiophene it is selected from unsubstituted 2-furan or 2,5-substituted furan and unsubstituted 2-thiophene or 2,5-substituted thiophene.
  • R-i is an unsubstituted furan then it has the ability to cross the blood brain barrier at levels about 10 times greater than CRID3, a prior art sulfonylurea.
  • R-i is 5-membered heterocyclyl or heteroaryl, each of which may be optionally substituted, comprising at least one, preferably at least two ring heteroatoms selected from N, 0 and S.
  • Ri is a nitrogen heterocyclyl or nitrogen heteroaryl, each of which may be optionally substituted.
  • Ri is 5-membered nitrogen heterocyclyl or 5- membered nitrogen heteroaryl, each of which may be optionally substituted.
  • Ri is 5-membered heterocyclyl or 5-membered heteroaryl, each comprising at least two ring nitrogen atoms and each of which rings may be optionally substituted.
  • W is O
  • J is S
  • R-i is selected from the group consisting of quinoline, isoquinoline, naphthyl, pyrazine, tetrazole, imidazole, pyrrolidine, pyrrole, tetrahydropyran, pyran, piperidine, piperazine, pyrazole, pyridine, pyrimidine and triazole, each of which may be optionally substituted.
  • R-i and/or R 2 may comprise a selenocycle.
  • R 2 may be selected from bicyclic and tricyclic hydrocarbons, 5-, 6- and 7-membered heterocycle or heteroaryl, each of which rings may be optionally substituted, and substituted phenyl.
  • the tricyclic hydrocarbon may be an indacene.
  • R 2 may be selected from 5-, 6- or 7- membered nitrogen heterocycles, 6-membered nitrogen heteroaryl and aryl with fused cycloalkyl ring.
  • W is 0, J is S and Ri may be selected from the group consisting of:
  • R 2 may be selected from:
  • each incidence of Y is independently selected from C, N, S and O, and which may be optionally substituted, as appropriate;
  • 5 are independently selected from the group consisting of hydrogen, halo, cyano, amide, sulphonamide, acyl, hydroxyl, Ci- Ce alkyl, d-C6 haloalkyi, Cs-Cs cyloalkyl, and C-1 -C6 alkoxy, all of which groups may be optionally substituted, as appropriate, with halo, cyano or C C 6 alkoxy; and wherein R-n and R 2 may combine to form phenyl, a 5- or 6-membered oxygen heterocycle or a 5- or 6-membered nitrogen heteroaryl, each of which may be optionally substituted;
  • R-12 and R-13 may combine to form a 5- or 6-membered nitrogen heteroaryl, which may be optionally substituted;
  • RH and R15 may combine to form a 5- or 6-membered cycloalkyl ring, phenyl, a 5- or 6-membered oxygen heterocycle or a 5- or 6-membered nitrogen heteroaryl, each of which may be optionally substituted.
  • each incidence of Y is a carbon and R 5 is hydrogen or halo.
  • R 2 and R 4 are hydrogen
  • R-n and R 5 are C C 6 alkyl
  • 3 is hydrogen or halo.
  • R 2 is selected from a substituted or hydrogenated indacene, a 2,6-dialkylphenyl, a 2,6-dialkyl-4-halophenyl, 2,6-dicycloalkylphenyl, and a 2,6-dicycloalkyl-4-halophenyl.
  • R 2 is selected from hexahydroindacene, 2,6-diisopropylphenyl 2,6-diisopropyl-4-chlorophenyl, 2,6- dicyclopropylphenyl and 2,6-dicyclopropyl-4-chlorophenyl.
  • W is O and J is S, Ri is heteroaryl and R 2 is
  • each Y is CH and R 5 is H or halogen, preferably R 5 is H.
  • W is O and J is S, Ri is heteroaryl and R 2
  • R-11 and Ri 5 are Ci -6 alkyl, preferably isopropyl
  • Ri 3 is H or halogen, preferably H or CI.
  • W is 0 and J is S, Ri is heteroaryl and R 2
  • R-,-, and R 5 are isopropyl, R 2 and R are H, and R 3 is H or CI.
  • the compound of formula (I) may be selected from a compound of formula (la), (lb) and (Ic), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
  • Ri is selected from the group consisting of pyrazole, furan, tetrahydrofuran, tetrahydropyran, pyran, pyrrolidine, pyrrole, triazole, tetrazole, imidazole, pyridine, morpholine, piperazine, piperidine, substituted phenyl, phenylheteroaryl, phenylheterocyclyl, biphenyl, quinoline, isoquinoline, naphthyl, pyrazine and pyrimidine, all of which may be optionally substituted as appropriate.
  • R-i is selected from the group consisting of:
  • the compound of formula (I) may be selected from a compound of formula (II), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
  • R 2 is as previously defined for any embodiment of formula (I), (la), (lb) or (lc), or may be a fluorescent group; each incidence of R 6 is independently selected from the group consisting of hydrogen, halo, cyano, CrC 6 alkyl, C C 6 alkylamino, CrC 6 alkylhydroxy, C 3 - Ce cycloalkyi, alkylphenyl, phenyl, benzyl, C-i-Ce ester, C2-C6 alkenyl, C-i-Ce t fluoroalkyl and C C 6 alkoxy, each of which may be optionally substituted, or R 6 may be a fluorescent group.
  • at least one of A, B, D and E is N (i.e. nitrogen).
  • At least two of A, B, D and E are N.
  • A, B, D and E are selected from N and C.
  • A is C and at least two of B, D and E are N.
  • A, B, D and E form a ring selected from a pyrazole, an imidazole, a triazole, and a tetrazole.
  • A, B, D, and E form a ring selected from a pyrazole or an imidazole ring, most preferably a pyrazole ring.
  • A, B, D and E and/or R 2 may comprise a selenocycle.
  • the compound of formula (I) may be selected from a compound of formula (lla), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
  • A, B, D and E are selected from N and C and at least two of A, B, D, and E are N; each incidence of R 6 is independently selected from the group consisting of hydrogen, halide, cyano, C1-C6 alkyl, C1 -C6 alkylamino, C1 -C6 alkylhydroxy, C 3 -C 6 cycloalkyl, alkylphenyl, phenyl, benzyl, C C 6 ester, C 2 -C 6 alkenyl, C C 6 trifluoroalkyl and C1-C6 alkoxy, each of which may be optionally substituted.
  • the compound of formula (I) may be selected from a compound of formula (lib), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
  • A, B, D and E are selected from N and C and at least two of A, B, D, and E are N;
  • each incidence of R 6 is independently selected from the group consisting of hydrogen, halide, cyano, C C 6 alkyl, Ci -C 6 alkylamino, Ci -C 6 alkylhydroxy, C3-C6 cycloalkyl, alkylphenyl, phenyl, benzyl, C1-C6 ester, C2-C6 alkenyl, C1-C6 trifluoroalkyl and C C 6 alkoxy, each of which may be optionally substituted.
  • the compound of formula (II), is selected from:
  • R 40 is selected from H, alkyl and halo
  • R 4 i is selected from H and alkyl
  • each incidence of P is independently selected from C, 0 or S; and wherein each incidence of R 6 , when present, is independently selected from those groups defined for formula (II).
  • R 6 moiety extending from the centre of each ring may represent a group bonded to the ring carbons or ring heteroatoms, as appropriate taking valency into consideration, or may not be present.
  • R 6 is d-C6 alkyl or C-1 -C6 alkylhydroxy.
  • R 6 may not be a tertiary alcohol substituent.
  • the compound of the first aspect may be selected from a compound of formula (Ilia), (lllb) or (lllc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
  • R 2 i is selected from H, alkyl, perhaloalkyl or hydroxylalkyl
  • R22 is selected from H, alkyl, perhaloalkyl, C 3 -C 6 cycloalkyl, phenyl or benzyl
  • R-I 8 is H or halogen
  • R-I6 and Ri 7 are H or alkyl; or R-
  • R-I 9 and R20 are H or alkyl; or R19 and R20, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
  • R21 and R22 are not both H.
  • R 6 , Ri 7 , R18, R19 and R 20 are not all H.
  • R21 is selected from H, alkyl, perhaloalkyl or hydroxylalkyl; preferably C1-6 perhaloalkyl or hydroxylalkyl;
  • R22 is selected from H, alkyl, perhaloalkyl, C3-C6 cycloalkyl, phenyl or benzyl;
  • 7 together with the atoms to which they are attached, form a cyclopentyl ring;
  • R-19 and R 2 o together with the atoms to which they are attached, form a cyclopentyl ring
  • R-I 8 is H or halogen, preferably R-
  • R 2 i and R22 are not both H.
  • R21 is selected from H, alkyl, perhaloalkyi or hydroxylalkyl; preferably C1-6 perhaloalkyi or hydroxylalkyl;
  • R22 is selected from H, alkyl, perhaloalkyi, C3-C6 cycloalkyi, phenyl and benzyl;
  • R-I6 and R 2 o are C1-6 alkyl, preferably isopropyl
  • R-17 and R 9 are H
  • R-I 8 is H or halogen; preferably R-
  • R 2 i and R22 are not both H.
  • the compound of the first aspect may be selected from a compound of formula (IVa), (IVb) or (IVc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
  • R 2 i and R22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyl, C3-C6 cycloalkyi, phenyl and benzyl or R21 and R22, together with the carbon atoms to which they are attached, may form a cyclopentyl or a cyclohexyl ring;
  • R-18 is H or halogen;
  • R-16 and R 7 are H or alkyl; or R 6 and R 7 , together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
  • R-I 9 and R 20 are H or alkyl; or R 9 and R 2 o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
  • R 2 i and R 22 are not both H;
  • 6 , R17, R18, R19 and R 20 are not all H.
  • R 2 and R 22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyi, C 3 -C 6 cycloalkyi, phenyl and benzyl; preferably the perhaloalkyi and hydroxylalkyi are Ci -6 perhaloalkyi and hydroxylalkyi;
  • 7 together with the atoms to which they are attached, form a cyclopentyl ring
  • R-19 and R 20 together with the atoms to which they are attached, form a cyclopentyl ring;
  • R-18 is H or halogen; preferably R-
  • R 2 and R 22 are not both H.
  • R 2 i and R 22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyi, C3-C6 cycloalkyi, phenyl and benzyl; preferably the perhaloalkyi and hydroxylalkyi are C1 -6 perhaloalkyi and hydroxylalkyi;
  • R-16 and R 20 are Ci -6 alkyl, preferably isopropyl; R-17 and R-
  • R 8 is H or halogen; preferably R 8 is H or CI;
  • R 2 i and R22 are not both H.
  • the compound of the first aspect may be selected from a compound of formula (Va), (Vb) or (Vc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
  • R 2 i and R22 are selected from H, alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl;
  • R-I 8 is H or halogen
  • R-I 6 and Ri 7 are H or alkyl; or R-
  • R-19 and R 2 o are H or alkyl; or R-19 and R 2 o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
  • R 2 i and R 22 are not both H;
  • 6 , R17, R18, R19 and R 2 o are not all H.
  • R21 and R22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyi, C 3 -C 6 cycloalkyi, phenyl and benzyl; preferably the perhaloalkyi and hydroxylalkyi are Ci-6 perhaloalkyi and hydroxylalkyi;
  • R-19 and R 2 o together with the atoms to which they are attached, form a cyclopentyl ring
  • R-I 8 is H or halogen; preferably R-
  • R 2 i and R 22 are not both H.
  • R 2 i and R 22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyi, C3-C6 cycloalkyi, phenyl and benzyl; preferably the perhaloalkyi and hydroxylalkyi are C1-6 perhaloalkyi and hydroxylalkyi;
  • R-I 6 and R 20 are Ci -6 alkyl, preferably isopropyl;
  • R-I 8 is H or halogen; preferably R 8 is H or CI; and
  • R 2 i and R 22 are not both H.
  • the compound of the first aspect may be selected from a compound of formula (Via) or (VIb), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
  • Formula (Via) Formula (Vlb) wherein, R22 is selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl;
  • R-18 is H or halogen
  • R-16 and R 7 are H or alkyl; or R 6 and R 7 , together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
  • R-19 and R 20 are H or alkyl; or R 9 and R 2 o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S; and
  • R 6 , Ri 7 , R18, R19 and R 20 are not all H.
  • R22 is selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl; preferably the perhaloalkyl and hydroxylalkyl are Ci -6 perhaloalkyl and hydroxylalkyl;
  • R 6 and R 7 together with the atoms to which they are attached, form a cyclopentyl ring
  • R-19 and R 2 o together with the atoms to which they are attached, form a cyclopentyl ring; and R-18 is H or halogen; preferably R-
  • R22 is selected from alkyl, perhaloalkyl, hydroxylalkyl, C 3 -C 6 cycloalkyl, phenyl and benzyl; preferably the perhaloalkyl and hydroxylalkyl are C1-6 perhaloalkyl and hydroxylalkyl;
  • R-I 6 and R 2 o are C1-6 alkyl, preferably isopropyl
  • R-17 and R 9 are H;
  • R-18 is H or halogen; preferably R-
  • the compound of the first aspect may be selected from a compound of formula (VII), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
  • each incidence of R 30 is independently selected from alkyl, perhaloalkyl, hydroxylalkyl, C 3 -C 6 cycloalkyl, and alkylam ino;
  • R-18 is H or halogen
  • R-I 6 and R 7 are H or alkyl; or R 6 and R 7 , together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
  • R-19 and R 20 are H or alkyl; or R 9 and R 2 o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
  • R 6 , R-17, R18, R19 and R 20 are not all H; and provided that when Q is 0 and R-
  • each incidence of R 30 is independently selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, and alkylamino; preferably C1-6 alkyl, perhaloalkyl, hydroxylalkyl, and alkylamino;
  • R-I 8 is H or halogen; preferably R-
  • each incidence of R 30 is independently selected from alkyl, perhaloalkyl, hydroxylalkyl, C 3 -C 6 cycloalkyl, and alkylamino; preferably Ci -6 alkyl, perhaloalkyl, hydroxylalkyl, and alkylamino;
  • R-I 6 and R 20 are Ci -6 alkyl, preferably isopropyl;
  • R-17 and R-19 are H; and R-18 is H or halogen; preferably R-
  • the compounds of the first aspect, and particularly those of formulae (II) to (VI), provide a range of unexpected benefits over those sulfonylureas of the prior art, which benefits may be selected from: Improved microsomal stability; Improved permeability; Reduced Pgp liability; Reduced plasma protein binding; Increased half-life; Improved oral bioavailability; Improved AUC; Improved Cmax; Reduced Cyp inhibition; Improved inhibition of activation of the NLRP3 inflammasome; and Improved solubility.
  • the solubility, and certain other, improvements may be seen particularly in an aqueous environment.
  • the compounds of the first aspect offer improved pharmacokinetic characteristics.
  • CRID3 a known sulfonylurea, has a half life of 3.2 hours (mouse) which may lead to substantial trough levels from QD or BD dosing when the t1/2 is extrapolated to man.
  • the compounds of the first aspect may differ in, for example, their protein binding, metabolism and oral availability.
  • the compounds of of the first aspect have a tPSA of less than 90 A 2 .
  • the compounds of the first aspect have a tPSA of less than 90 A 2 and a molecular weight of less than 405.
  • the absence of a tertiary alcohol group in some embodiments, increases plasma concentration and aids in decreasing both MW and polar surface area thereby giving an overall improvement in blood brain barrier penetration.
  • one or more hydrogens of the substituents or optional substitutions thereupon may be deuterated.
  • Deuterated analogues of the compounds of the invention may exhibit increased metabolic stability due to the kinetic isotope effect.
  • the compound of the first aspect is selected from the group consisting of:
  • the compounds of the first aspect may exhibit improved properties compared to known anti-diabetes drugs.
  • examples of such compounds may include those below:
  • the compound is an inhibitor of activation of the NLRP3 inflammasome.
  • the present invention provides for sulfonyl urea and related drugs exhibiting significantly lower NLRP3 IC50 values in cell based assay using HMDM (see experimental section for protocols) than the above comparator compounds.
  • HMDM cell based assay using HMDM
  • one or more of the compounds of the first aspect may be useful as photoswitchable compounds which may be applied in a range of uses including but not limited to insulin release.
  • Such compounds may, in one embodiment, be selected from the group consisting of:
  • R 2 is as defined in any one or more of the embodiments of compounds of formula (I) to (VII) described previously.
  • one or more compounds of the first aspect may be appropriate for use as probes, such as photoaffinity probes, or as reactive intermediates which can be modified either directly or by means of a linking moiety to give biotinylated, fluorescent or photoaffinity probes including, but not limited to, those shown below:
  • R 2 is as defined in any one or more of the embodiments described for formula (I) to (VII).
  • probes or reactive intermediates may be selected from those below:
  • the compounds of the first aspect may be modified or derivatised by means well understood in the art to allow linkage to a molecule such as biotin, or a fluorescent group or photoaffinity label, as shown with certain of the compounds above.
  • the compound of formula (I) or (II) does not comprise a structure selected from the groups below shown attached to the sulfonyl moiety (i.e. as an Ri group):
  • the compound of the first aspect including any compound of formula (I) to (VII)
  • Ri is not one of 2,4-disubstituted furan, 2,4-disubstituted thiophene, 2,5-disubstituted furan and 2,5-disubstituted thiophene.
  • the compound of the first aspect including any compound of formula (I) to (VII)
  • has J as S, W as O and R-i is selected from substituted triazole, thiadiazole, 4-substituted pyridine and 1 ,2- disubstituted imidazole then R 2 is not unsubstituted phenyl, 2- or 4-chlorophenyl or 3,4- substituted phenyl, substituted with one or more of halo, trifluoromethyl, nitro or thiomethyl.
  • R-i is selected from substituted triazole, thiadiazole, benzothiazole and substituted pyrimidine then R 2 is not thiophene, 3-chlorophenyl, 4-ethoxyphenyl, substituted benzimidazole or substituted benzothiazole.
  • R 2 is not 2,6-diisopropylphenyl.
  • R 2 is not 3- or 3,4- halo, methyl, ethyl or trifluoromethyl substituted phenyl.
  • the carbon atom of R 2 which is directly bonded to the urea nitrogen is an aryl, heteroaryl or heterocyclic ring carbon.
  • the compound of the first aspect including any compound of formula (I) to (VII)
  • R 2 is a substituted phenyl and Ri is a pyrazole then the Ri pyrazole is not substituted with an aryl or heteroaryl group.
  • the compound of the first aspect including any compound of formula (I) to (VII)
  • the pyrazole is not fused in positions 1 and 5 with a 6-membered heterocycle to form a pyrazolopyrimidine derivative.
  • the compound of the first aspect including any compound of formula (I) to (VII) is not a compound selected from the group consisting of:
  • prodrugs are compounds which, when administered to a mammal, are converted in whole or in part to a compound of the invention.
  • the prodrugs are pharmacologically inert chemical derivatives that can be converted in vivo to the active drug molecules to exert a therapeutic effect. Any of the compounds described herein can be administered as a prodrug to increase the activity, bioavailability, or stability of the compound or to otherwise alter the properties of the compound.
  • Typical examples of prodrugs include compounds that have biologically labile protecting groups on a functional moiety of the active compound.
  • Prodrugs include, but are not limited to, compounds that can be oxidized, reduced, aminated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, dehydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated, and/or dephosphorylated to produce the active compound.
  • prodrug ligands are known.
  • alkylation, acylation, or other lipophilic modification of one or more heteroatoms of the compound, such as a free amine or carboxylic acid residue may reduce polarity and allow for the compound's passage into cells.
  • substituent groups that can replace one or more hydrogen atoms on a free amine and/or carboxylic acid moiety include, but are not limited to, the following: aryl; steroids; carbohydrates (including sugars); 1 ,2-diacylglycerol; alcohols; acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester (including alkyl or arylalkyl sulfonyl, such as methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as provided in the definition of an aryl given herein); optionally substituted arylsulfonyl; lipids (including phospholipids); phosphotidylcholine; phosphocholine; amino acid residues or derivatives; amino acid acyl residues or derivatives; peptides; cholesterols; or other pharmaceutically acceptable leaving groups which, when administered in vivo, provide the free amine
  • any of these moieties can be used in combination with the disclosed active agents to achieve a desired effect.
  • compounds with one or more chiral centers are provided. While racemic mixtures of compounds of the invention may be active, selective, and bioavailable, isolated isomers may be of interest as well.
  • the compounds of the first aspect may contain chiral centers, which may be either of the (R) or (S) configuration, or which may comprise a mixture thereof. Accordingly, the present invention also includes stereoisomers of the compounds described herein, where applicable, either individually or admixed in any proportions. Stereoisomers may include, but are not limited to, enantiomers, diastereomers, racemic mixtures, and combinations thereof. Such stereoisomers can be prepared and separated using conventional techniques, either by reacting enantiomeric starting materials, or by separating isomers of compounds and prodrugs of the present invention. Isomers may include geometric isomers.
  • geometric isomers include, but are not limited to, cis isomers or trans isomers across a double bond.
  • Other isomers are contemplated among the compounds of the present invention.
  • the isomers may be used either in pure form or in admixture with other isomers of the compounds described herein.
  • optical isomers of the compounds according to the present invention include the following: i) physical separation of crystals whereby macroscopic crystals of the individual enantiomers are manually separated. This technique may particularly be used when crystals of the separate enantiomers exist (i.e., the material is a conglomerate), and the crystals are visually distinct;
  • enzymatic asymmetric synthesis a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer;
  • kinetic resolutions comprising partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions; ix) enantiospecific synthesis from non-racemic precursors whereby the desired enantiomer is obtained from non-chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis;
  • x) chiral liquid chromatography whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase.
  • the stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions;
  • xiii) transport across chiral membranes whereby a racemate is placed in contact with a thin membrane barrier.
  • the barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane barrier. Separation occurs as a result of the non-racemic chiral nature of the membrane which allows only one enantiomer of the racemate to pass through.
  • the compound optionally may be provided in a composition that is enantiomerically enriched, such as a mixture of enantiomers in which one enantiomer is present in excess, in particular, to the extent of 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more, including 100%.
  • a composition that is enantiomerically enriched, such as a mixture of enantiomers in which one enantiomer is present in excess, in particular, to the extent of 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more, including 100%.
  • the terms (R), (S), (R,R), (S,S), (R,S) and (S,R) as used herein mean that the composition contains a greater proportion of the named isomer of the compound in relation to other isomers.
  • these terms indicate that the composition contains at least 90% by weight of the named isomer and 10% by weight or less of the one or more other isomers; or more preferably about 95% by weight of the named isomer and 5% or less of the one or more other isomers.
  • the composition may contain at least 99% by weight of the named isomer and 1 % or less by weight of the one or more other isomers, or may contain 100% by weight of the named isomer and 0% by weight of the one of more other isomers. These percentages are based on the total amount of the compound of the present invention present in the composition.
  • the compounds of the first aspect may be utilized per se or in the form of a pharmaceutically acceptable ester, amide, salt, solvate, prodrug, or isomer, as appropriate.
  • the compound may be provided as a pharmaceutically acceptable salt.
  • a salt of the drug compound should be both pharmacologically and pharmaceutically acceptable, but non- pharmaceutically acceptable salts may conveniently be used to prepare the free active compound or pharmaceutically acceptable salts thereof and are not excluded from the scope of this invention.
  • Such pharmacologically and pharmaceutically acceptable salts can be prepared by reaction of the drug with an organic or inorganic acid, using standard methods detailed in the literature.
  • Examples of pharmaceutically acceptable salts of the compounds useful according to the invention include acid addition salts. Salts of non- pharmaceutically acceptable acids, however, may be useful, for example, in the preparation and purification of the compounds.
  • Suitable acid addition salts according to the present invention include organic and inorganic acids. Preferred salts include those formed from hydrochloric, hydrobromic, sulfuric, phosphoric, citric, tartaric, lactic, pyruvic, acetic, succinic, fumaric, maleic, oxaloacetic, methanesulfonic, ethanesulfonic, p-toluenesulfonic, benzenesulfonic, and isethionic acids.
  • compositions include propionic acid, glycolic acid, oxalic acid, malic acid, malonic acid, benzoic acid, cinnamic acid, mandelic acid, salicylic acid, and the like.
  • pharmaceutically acceptable salts include, but are not limited to, sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1 ,4-dioates, hexyne-1 ,6-dioates
  • An acid addition salt may be reconverted to the free base by treatment with a suitable base.
  • Preparation of basic salts of acid moieties which may be present on a compound or prodrug useful according to the present invention may be prepared in a similar manner using a pharmaceutically acceptable base, such as sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, triethylamine, or the like.
  • Esters of the active agent compounds according to the present invention may be prepared through functionalization of hydroxyl and/or carboxyl groups that may be present within the molecular structure of the compound.
  • Amides and prodrugs may also be prepared using techniques known to those skilled in the art. For example, amides may be prepared from esters, using suitable amine reactants, or they may be prepared from an anhydride or an acid chloride by reaction with ammonia or a lower alkyl amine.
  • esters and amides of compounds of the invention can be made by reaction with a carbonylating agent ⁇ e.g., ethyl formate, acetic anhydride, methoxyacetyl chloride, benzoyl chloride, methyl isocyanate, ethyl chloroformate, methanesulfonyl chloride) and a suitable base ⁇ e.g., 4-dimethylaminopyridine, pyridine, triethylamine, potassium carbonate) in a suitable organic solvent (e.g., tetrahydrofuran, acetone, methanol, pyridine, ⁇ , ⁇ -dimethylformamide) at a temperature of 0 °C to 60 °C.
  • a carbonylating agent e.g., ethyl formate, acetic anhydride, methoxyacetyl chloride, benzoyl chloride, methyl isocyanate, ethyl chloroformate, me
  • Prodrugs are typically prepared by covalent attachment of a moiety, which results in a compound that is therapeutically inactive until modified by an individual's metabolic system.
  • Examples of pharmaceutically acceptable solvates include, but are not limited to, compounds according to the invention in combination with water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, or ethanolamine.
  • the compounds used in the methods of the invention may exist in different forms.
  • the compounds may exist in stable and metastable crystalline forms and isotropic and amorphous forms, all of which are intended to be within the scope of the present invention.
  • a compound useful as an active agent according to the invention is a base
  • the desired salt may be prepared by any suitable method known to the art, including treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, pyranosidyl acids such as glucuronic acid and galacturonic acid, alpha-hydroxy acids such as citric acid and tartaric acid, amino acids such as aspartic acid and glutamic acid, aromatic acids such as benzoic acid and cinnamic acid, sulfonic acids such a p-toluenesulfonic acid or ethanesulfonic acid, or the like.
  • an inorganic acid such as hydrochlor
  • the desired salt may be prepared by any suitable method known in the art, including treatment of the free acid with an inorganic or organic base, such as an am ine (primary, secondary or tertiary), an alkali metal or alkaline earth metal hydroxide or the like.
  • suitable salts include organic salts derived from amino acids such as glycine and arginine, ammonia, primary, secondary and tertiary amines, and cyclic amines such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium , calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
  • a pharmaceutical composition comprising a compound of formula (I) to (VII), or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier, diluent and/or excipient.
  • the pharmaceutically acceptable carrier, diluent and/or excipient may be or include one or more of diluents, solvents, pH buffers, binders, fillers, emulsifiers, disintegrants, polymers, lubricants, oils, fats, waxes, coatings, viscosity-modifying agents, glidants and the like.
  • the salt forms of the compounds of the invention may be especially useful due to their improved solubility.
  • the pharmaceutical composition includes a cyclodextrin.
  • the cyclodextrin may be selected from alpha, beta or gamma cyclodextrins.
  • the cyclodextrin is selected from a methyl cyclodextrin, a hydroxypropyl cyclodextrin and a sulfobutylether cyclodextrin.
  • Cyclodextrin formulations such as for example, one or more compounds of the invention with hydroxypropyl beta cyclodextrin or methyl beta cyclodextrin, may have uses in cholesterol sequestration/cholesterol lowering or via NLRP3 inhibition for Non-alcoholic steatohepatitis (NASH), alcoholic liver disease, atherosclerosis and also in Alzheimer's Disease (AD).
  • NASH Non-alcoholic steatohepatitis
  • AD Alzheimer's Disease
  • Diluents may include one or more of microcrystalline cellulose, lactose, mannitoi, calcium phosphate, calcium sulfate, kaolin, dry starch, powdered sugar, and the like.
  • Binders may include one or more of povidone, starch, stearic acid, gums, hydroxypropylmethyl cellulose and the like.
  • Disintegrants may include one or more of starch, croscarmellose sodium, crospovidone, sodium starch glycolate and the like.
  • Solvents may include one or more of ethanoi, methanol, isopropanoi, chloroform, acetone, methylethyl ketone, methylene chloride, water and the like.
  • Lubricants may include one or more of magnesium stearate, zinc stearate, calcium stearate, stearic acid, sodium stearyl fumarate, hydrogenated vegetable oil, glyceryl behenate and the like.
  • a glidant may be one or more of colloidal silicon dioxide, talc or cornstarch and the like.
  • Buffers may include phosphate buffers, borate buffers and carbonate buffers, although without limitation thereto.
  • Fillers may include one or more gels inclusive of gelatin, starch and synthetic polymer gels, although without limitation thereto.
  • Coatings may comprise one or more of film formers, solvents, plasticizers and the like.
  • Suitable film formers may be one or more of hydroxypropyl methyl cellulose, methyl hydroxyethyl cellulose, ethyl cellulose, hydroxypropyl cellulose, povidone, sodium carboxymethyl cellulose, polyethylene glycol, acrylates and the like.
  • Suitable solvents may be one or more of water, ethanoi, methanol, isopropanoi, chloroform, acetone, methylethyl ketone, methylene chloride and the like.
  • Plasticizers may be one or more of propylene glycol, castor oil, glycerin, polyethylene glycol, poiysorbates, and the like.
  • composition may be in the form of a tablet, capsule, caplet, powder, an injectable liquid, a suppository, a slow release formulation, an osmotic pump formulation or any other form that is effective and safe for administration.
  • the pharmaceutical composition is for the treatment or prevention of a disease, disorder or condition in a mammal.
  • a third aspect of the invention resides in a method of treatment or prevention of a disease, disorder or condition including the step of administering an effective amount of a compound of formula (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof, or the pharmaceutical composition of the second aspect to thereby treat or prevent the disease disorder or condition.
  • a fourth aspect of the invention provides for a compound of formula (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof, or the pharmaceutical composition of the second aspect for use in the treatment or prevention of a disease, disorder or condition.
  • a fifth aspect of the invention provides for use of a compound of formula (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof, in the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition.
  • administering or “administration”, and the like, describe the introduction of the compound or composition to a mammal such as by a particular route or vehicle.
  • Routes of administration may include topical, parenteral and enteral which include oral, buccal, sub-lingual, nasal, anal, gastrointestinal, subcutaneous, intramuscular and intradermal routes of administration, although without limitation thereto.
  • treat administration of the compound or composition to a subject to at least ameliorate, reduce or suppress existing signs or symptoms of the disease, disorder or condition experienced by the subject.
  • prevent prophylactically administering the formulation to a subject who does not exhibit signs or symptoms of a disease disorder or condition, but who is expected or anticipated to likely exhibit such signs or symptoms in the absence of prevention.
  • Preventative treatment may at least lessen or partly ameliorate expected symptoms or signs.
  • ' effective amount refers to the administration of an amount of the relevant compound or composition sufficient to prevent the occurrence of symptoms of the condition being treated, or to bring about a halt in the worsening of symptoms or to treat and alleviate or at least reduce the severity of the symptoms.
  • the effective amount will vary in a manner which would be understood by a person of skill in the art with patient age, sex, weight etc. An appropriate dosage or dosage regime can be ascertained through routine trial.
  • the terms "subject” or “individual” or “patient” may refer to any subject, particularly a vertebrate subject, and even more particularly a mammalian subject, for whom therapy is desired.
  • Suitable vertebrate animals include, but are not restricted to, primates, avians, livestock animals (e.g., sheep, cows, horses, donkeys, pigs), laboratory test animals (e.g., rabbits, mice, rats, guinea pigs, hamsters), companion animals (e.g., cats, dogs) and captive wild animals (e.g., foxes, deer, dingoes).
  • a preferred subject is a human in need of treatment for a disease, disorder or condition as described herein. However, it will be understood that the aforementioned terms do not imply that symptoms are necessarily present.
  • the disease, disorder or condition is one which is responsive to inhibition of activation of the NLRP3 inflammasome.
  • the compound of the first aspect, or pharmaceutically effective salt, solvate or prodrug thereof is a specific inhibitor of NLRP3.
  • the disease, disorder or condition is responsive to modulation of one or more of IL- ⁇ ⁇ , IL-17, IL-18, IL-1 , IL-37, IL- 33 and Th17 cells.
  • the modulation is inhibition of one or more of IL- 1 ⁇ , IL-17, IL-18, IL-1 a, IL-37, and IL-33.
  • the modulation of Th17 cells is by inhibition of production and/or secretion of IL-17.
  • the disease, disorder or condition is a disease, disorder or condition of the immune system, the cardiovascular system, the endocrine system, the gastrintestinal tract, the renal system, the respiratory system, the central nervous system, is a cancer or other malignancy and/or is caused by or associated with a pathogen.
  • any particular disease, disorder or condition may be categorized according to more than one of the above general embodiments.
  • a non-limiting example is Type I diabetes which is an autoimmune disease and a disease of the endocrine system.
  • the disease, disorder or condition is of the immune system.
  • the disease disorder or condition is an inflammatory disease disorder or condition or an autoimmune disease disorder or condition.
  • the disease, disorder or condition is of the skin.
  • the disease, disorder or condition is of the cardiovascular system.
  • the disease, disorder or condition is a cancer, tumour or other malignancy.
  • cancers tumours and malignancies refer to diseases disorders or conditions, or to cells or tissues associated with the diseases, disorders or conditions, characterized by aberrant or abnormal cell proliferation, differentiation and/or migration often accompanied by an aberrant or abnormal molecular phenotype that includes one or more genetic mutations or other genetic changes associated with oncogenesis, expression of tumour markers, loss of tumour suppressor expression or activity and/or aberrant or abnormal cell surface marker expression.
  • cancers, tumours and malignancies may include sarcomas, lymphomas, leukemias, solid tumours, blastomas, gliomas, carcinomas, melanomas and metastatic cancers, although without limitation thereto.
  • sarcomas lymphomas, leukemias, solid tumours, blastomas, gliomas, carcinomas, melanomas and metastatic cancers, although without limitation thereto.
  • a more comprehensive listing of cancers tumours and malignancies may be found at the National Cancer Institutes website http://www.cancer.gov/cancertopics/types/alphalist.
  • the disease, disorder or condition is of the renal system .
  • the disease, disorder or condition is of the gastro-intestinal tract. [00190] In one embodiment, the disease, disorder or condition is of the respiratory system.
  • the disease, disorder or condition is of the endocrine system.
  • the disease, disorder or condition is of the central nervous system (CNS).
  • CNS central nervous system
  • the disease, disorder or condition is caused by, or is associated with, a pathogen.
  • the pathogen may be a virus, a bacterium, a protist, a worm or a fungus or any other organism capable of infecting a mammal, although without limitation thereto.
  • Non-limiting examples of viruses include influenza virus, cytomegalovirus, Epstein Barr Virus, human immunodeficiency virus (HIV), alphavirus such as Chikungunya and Ross River virus, flaviviruses such as Dengue virus, Zika virus and papillomavirus, although without limitation thereto.
  • HIV human immunodeficiency virus
  • alphavirus such as Chikungunya and Ross River virus
  • flaviviruses such as Dengue virus, Zika virus and papillomavirus, although without limitation thereto.
  • Non-limiting examples of pathogenic bacteria include
  • Staphylococcus aureus Helicobacter pylori, Bacillus anthracis, Bordatella pertussis, Corynebacterium diptheriae, Clostridium tetani, Clostridium botulinum, Streptococcus pneumoniae, Streptococcus pyogenes, Listeria monocytogenes, Hemophilus influenzae, Pasteureiia multicida, Shigella dysenteriae, Mycobacterium tuberculosis, Mycobacterium leprae, Mycoplasma pneumoniae, Mycoplasma hominis, Neisseria meningitidis, Neisseria gonorrhoeae, Rickettsia rickettsii, Legionella pneumophila, Klebsiella pneumoniae, Pseudomonas aeruginosa, Propionibacterium acnes, Treponema pallidum, Chlamydia trachomatis, Vibrio
  • Non-limiting examples of worms include helminths inclusive of schistisimes, roundworms, tapeworms and flukes, although without limitation thereto.
  • Non-limiting examples of fungi include Candida and Aspergillus species, although without limitation thereto.
  • the disease, disorder or condition is selected from the group consisting of constitutive inflammation including the cryopyrin-associated periodic syndromes (CAPS): Muckle-Wells syndrome (MWS), familial cold autoinflammatory syndrome (FCAS) and neonatal-onset multisystem inflammatory disease (NOMID); including autoinflammatory diseases: familial Mediterranean fever (FMF), TNF receptor associated periodic syndrome (TRAPS), mevalonate kinase deficiency (MKD), hyperimmunoglobulinemia D and periodic fever syndrome (H IDS), deficiency of interleukin 1 receptor (DIRA) antagonist, Majeed syndrome, pyogenic arthritis, pyoderma gangrenosum and acne (PAPA), haploinsufficiency of A20 (HA20), pediatric granulomatous arthritis (PGA), PLCG2 -associated antibody deficiency and immune dysregulation (PLAID), PLCG2-associated autoinflammation, antibody deficiency and immune dysregulation (APLAID), sideroblastic anemia with
  • the disease, disorder or condition being treated is NASH.
  • NLRP3 inflammasome activation is central to inflammatory recruitment in NASH, and inhibition of NLRP3 may both prevent and reverse liver fibrosis.
  • Compounds of the present invention by interrupting the function of NLRP3 inflammasomes in liver tissue, can cause histological reductions in liver inflammation, decreased recruitment of macrophages and neutrophils, and suppression of NF- ⁇ activation. Inhibition of the NLRP3 can reduce hepatic expression of pro-IL-1 ⁇ and normalized hepatic and circulating I L- p , IL-6 and MCP-1 levels thereby assisting in treatment of the disease.
  • the disease, disorder or condition being treated is severe steroid resistant (SSR) asthma.
  • SSR severe steroid resistant
  • Respiratory infections induce an NLRP3 inflammasome/caspase-l/IL- ⁇ signaling axis in the lungs that promotes SSR asthma.
  • the NLRP3 inflammasome recruits, and activates, pro-caspase-1 to induce IL-1 ⁇ responses.
  • NLRP3 inflammasome-induced IL-1 ⁇ responses are therefore important in the control of infections, however, excessive activation results in aberrant inflammation and has been associated with the pathogenesis of SSR asthma and COPD.
  • the administration of compounds of the first aspect that target specific disease processes, are more therapeutically attractive than non- specifically inhibiting inflammatory responses with steroids or IL-1 ⁇ .
  • Targeting the NLRP3 inflammasome/caspase-1/IL-1 ⁇ signaling axis with the compounds of the first aspect may therefore be useful in the treatment of SSR asthma and other steroid-resistant inflammatory conditions.
  • the disease, disorder or condition being treated is Parkinson's disease.
  • Parkinson's is the most common neurodegenerative movement disorder and is characterized by a selective loss of dopaminergic neurons, accompanied by the accumulation of mis-folded a-synuclein (Syn) into Lewy bodies that are pathological hallmarks of the disease.
  • Syn mis-folded a-synuclein
  • Chronic microglial neuroinflammation is evident early in the disease, and has been proposed to drive pathology.
  • a central role for microglial NLRP3 is postulated in Parkinson's progression.
  • the NLRP3 inflammasome is activated by fibrillar Syn via a Syk kinase dependent mechanism, and also occurs in the absence of Syn pathology at the early stages of dopaminergic degeneration, and drives neuronal loss.
  • the compounds of the first aspect may block NLRP3 inflammasome activation by fibrillar Syn or mitochondrial dysfunction and thereby confer effective neuroprotection of the nigrostriatal dopaminergic system and assist with treatment of Parkinson's.
  • a method of diagnosing a disease, disorder or condition in a mammal including the step of administering a labelled compound of formula (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof, to the mammal or to a biological sample obtained from the mammal to facilitate diagnosis of the disease disorder or condition in the mammal.
  • Inflammasome activation in particular that of the NLRP3 inflammasome, is known to drive initiation, progression and chronic development of a vast number of inflammatory diseases.
  • the sulfonylureas and related compounds of the first aspect are potent and specific direct inhibitors of NLRP3. Accordingly, a chemical probe specific for NLRP3, which is present in immune cells during inflammation has potential utility in diagnosing inflammatory and other related diseases.
  • An NRLP3 activation probe comprising a compound of the first aspect could act as an effective surrogate biomarker of inflammatory diease for ex vivo (blood) or in vivo (MRI, PET etc.) diagnostics.
  • the use of the compounds of the first aspect in diagnosing inflammatory and other related diseases may be achieved by near infrared fluorescent imaging and ex vivo characterisation of immune cells by degree of inhibition of IL-1 beta, pro-caspase 1 cleavage and IL-18 levels.
  • peripheral blood monocytes PMBCs
  • macrophages macrophages
  • dendritic cells CD4 + T cells
  • Th17 cells Th1 cells
  • Th2 cells are relevant.
  • PET positron emission tomography
  • Typical isotopes include 11 C, 13 N, 15 0, 18 F, 64 Cu, 62 Cu, 124 l, 76 Br, 82 Rb and 68 Ga, with 18 F being the most clinically utilized.
  • diagnostic probe for radioimaging, PET and the like whereby the intensity, location and temporal accretion of the diagnostic probe is able to identify the degree and/or the location of immune cells with activated NLRP3 as a surrogate biomarker of the patient's inflammatory state, and site of inflammation within the body. They will also be useful for application to biological samples removed from the body i.e. in vitro diagnosis.
  • a seventh aspect of the invention resides in a method of modulating the activity of a biological target comprising the step of exposing the biological target to a compound of formula (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof.
  • the biological target may be selected from the group consisting of NLRP3 inflammasome, IL-1 ⁇ , IL-17, IL-18, IL-1 a, IL-37, IL-33 and Th17 cells.
  • a biological sample may include cells, tissues, fluids, molecules or other biological materials obtained, or obtainable, from a mammal.
  • Non-limiting examples include urine, blood and fractions thereof such as serum, plasma, lymphocytes and erythrocytes, cerebrospinal fluid, PAP smears, nasal and ocular secretions, amniotic fluid, faeces, semen, tissue and/or organ biopsies and nucleic acid (e.g. DNA, RNA) or protein samples, although without limitation thereto.
  • the R2 acid chloride intermediate was dissolved in acetone and added drop- wise to a solution of sodium azide (1.5 eq) in a watenacetone (50:50) solution at 0 °C. Iced water was added to precipitate the resulting R2 acylazide intermediate which was dissolved in toluene and dried (MgS04) prior to adding the solution in a drop-wise fashion to anhydrous toluene at reflux while maintaining a constant flow of inert gas. The reaction was heated until completion, typically 2 h, to give the R2 isocyanate.
  • R1 sulfonamide intermediate (1 eq.) was dissolved in anhydrous THF or anhydrous methanol and treated with NaH (1 eq.) under reduced pressure. Once effervescence ceased the R2 isocyanate intermediate was added and the reaction mixture was stirred at ambient temperature overnight.
  • Ethyl furan-2-carboxylate (9.0 g, 64.3 mmol) was dissolved in dichloromethane (200 mL) and chlorosulfonic acid (7.5 g, 64.3 mmol) added. The reaction was stirred at ambient temperature for 6 hours, or until completion, then pyridine (5.6 g, 70.7 mmol) and PCI5 (14.7 g, 70.7 mmol) were added portionwise. The reaction mixture was stirred at ambient temperature for 16 hours then quenched using ice-water and stirred for 30 mins. The mixture was extracted using DCM and the combined organics washed with water, brine, dried (Na 2 S0 4 ) and concentrated in vacuo.
  • Methyl 2-methyl-5-sulfamoylfuran-3-carboxylate can be prepared by modification of procedures used to synthesise ethyl 2-methyl-5-sulfamoylfuran- 3-carboxylate but using methyl 2-methylfuran-3-carboxylate as starting material in place of ethyl 2-methylfuran-3-carboxylate.
  • Methyl 2-methyl-5-sulfamoylfuran-3-carboxylate (0.7 g, 3.2 mmol) in anhydrous THF (20 mL) at -10 °C was treated with c/ 3 -methyl magnesium iodide solution (1.0 M in Et 2 0, 26 mL) drop-wise over 10 minutes with vigorous stirring. The solution was then stirred at ambient temperature for 12 h then cooled to 0 °C and treated drop-wise with a solution of sat. ammonium chloride. The aqueous solution was extracted using EtOAc (2 x 25 mL), the combined organics washed with brine (25 mL), dried (Na 2 S0 4 ) and concentrated in vacuo.
  • reaction mixture was diluted with water and extracted with ethyl acetate (2 x 25 mL). The combined organics were washed with water (50 mL), brine (50 mL), dried (Na 2 S0 4 ) and concentrated in vacuo. The residue obtained was triturated with diethyl ether and n-pentane to give 1 - isopropyl-5-(trifluoromethyl)-1 /-/-pyrazole-3-sulfonamide as a white solid (75 mg, 61 %).
  • 2,3-dihydrobenzo[b]thiophene 1 , 1 -dioxide (0.75 g, 4.45 mmol) was heated in chlorosulfonic acid (1 .5 mL, 22.2 mmol ) at 80 °C for 4 h. Reaction mixture was poured onto crushed ice and stirred for 5 minutes. The aqueous solution was extracted with dichloromethane (2x50 mL) and the combined organics dried (MgS0 4 ) and concentrated in vacuo to give 2,3-dihydrobenzo[£>]thiophene-6- sulfonyl chloride 1 , 1 -dioxide (0.45 g, 38%) as a light brown oil.
  • Benzene-1 ,3-disulfonyl dichloride (0.50 g, 0.726 mmol) was dissolved in tetrahydrofuran (4 mL) and the solution was cooled to 0 °C.
  • aqueous ammonia (0.4 mL) was added at 0 °C and the mixture was stirred at ambient temperature for 1 h.
  • the mixture was poured into chilled water and extracted with ethyl acetate.
  • the combined organic extracts were washed with brine, dried (Na 2 S0 4 ) and concentrated in vacuo.
  • the resulting solid was triturated with pentane to afford the titled compound as a light brown solid (0.16 g, 87%).
  • 4-(trifluoromethyl)pyridine-2-sulfonamide was synthesized according to the procedures used to synthesise pyridine-2-sulfonamide but using 4- (trifluoromethyl)pyridine-2-thiol in place of pyridine-2-thiol.
  • the product 4- (trifluoromethyl)pyridine-2-sulfonamide was given as a solid (0.7 g, 56%).
  • Methyl 4-chlorobutanimidate hydrochloride (25 g, 146.1 mmol) was dissolved in DCM (250 mL) treated with Et 3 N (44.3 g, 4.38 mmol) and resulting solution was cooled to 0 °C.
  • 2,2-Dimethoxyethan-1 -amine (12.2 g, 1 16.9 mmol) was added dropwise to the above mixture over a period of 5 min.
  • the resulting reaction mixture was warmed to 60 °C and stirred for 3 h.
  • the reaction mixture was concentrated in vacuo and residue obtained was treated with in formic acid (150 mL) and heated at 80 °C for 24 h.
  • Xantphos (74mg, 0.128mmol) and Pd 2 (dba) 3 (60 mg, 0.064 mmol) were sequentially added to the aforementioned solution and the vessel purged with nitrogen gas for 5 minutes. The resulting mixture was stirred at 1 10 °C for 12h. Upon completion, the mixture was cooled to RT, diluted with EtOAc (25 mL) and filtered through celite. The filtrate was dried (Na 2 S0 4 ) and concentrated in vacuo.
  • 3-Nitro-1 H-pyrazole (5 g, 44 mmol) was dissolved in A/,A/-dimethylformamide (100 ml_), cooled to -5 °C and NaH (3.8 g, 93.6 mmol) added portionwise. The reaction mixture was stirred for 15 mins before adding dibromodifluoromethane (8.6 g, 44 mmol) and allowing to warm to ambient temperature overnight. The reaction mixture was quenched using ice-water and extracted using ethyl acetate.
  • reaction mixture was diluted using DCM, washed using water, brine, dried (Na 2 S0 4 ) and concentrated in vacuo.
  • the crude product was purified by column chromatography on silica gel using 10% EtOAc-hexanes eluent to give 1 -cyclopropyl-3-(2,5-dimethyl-1 H-pyrrol-1 -yl)-1 H-pyrazole as a yellow liquid (0.2 g, 32%).
  • 3-nitro-1 H-pyrazole (1 g, 8.85 mmol) was dissolved in N,N-dimethylformamide (20 mL) and treated with potassium carbonate (1.47 g, 10.62 mmol) and bromocyclohexane (1 .8 g, 10.62 mmol). The mixture was heated to 100 °C for 16 hours (or until completion) then cooled to ambient temperature diluted using water (100 mL) and extracted using ethyl acetate (2 x 75 mL). The combined organics were washed using water (100 mL), brine (100 mL), dried (Na 2 S0 4 ) and concentrated in vacuo.
  • Ethyl 1 -methyl-3-nitro-1 --pyrazole-5-carboxylate (0.65 g, 3.3 mmol) was dissolved in THF (20 mL) and MeOH (5 mL) at 0 °C.
  • Zinc powder (1 .0 g, 16.3 mmol) and aqueous NH 4 CI (0.87 g, 16.3 mmol) were added sequentially.
  • the resulting reaction mixture was stirred at ambient temperature for 4h, then heated to 70 C for 1 hour. The solvents were removed in vacuo. The residue obtained was dissolved in EtOAc (30 mL) and filtered through a bed of Celite.
  • Ethyl 1 -benzyl-3-nitro-1 --pyrazole-5-carboxylate (1.2 g, 4.36 mmol) was dissolved in THF (20 mL) and MeOH (5 mL) at 0 °C.
  • Zinc powder (1.4 g, 21 .8 mmol) and aqueous NH 4 CI (1.16g, 21 .8 mmol) were added sequentially.
  • the resulting reaction mixture was stirred at ambient temperature for 4h, then concentrated in vacuo.
  • the residue obtained was dissolved in EtOAc (30 mL) and filtered through a bed of Celite.
  • A/-Bromosuccinimide (1.02 g, 5.78 mmol) was added portion-wise to a solution of 1 ,2,3,5,6,7-hexahydro-s-indacen-4-amine (1 g, 5.78 mmol) in DCM (20 mL) at 0 °C.
  • the solution was gradually warmed to ambient temperature and stirred for 12 h.
  • the reaction mixture was diluted with sat. aqueous Na 2 S 2 0 3 (50 mL) and extracted with DCM (2 x 25 mL). The combined organic extracts were washed with water (25 mL), brine (25 mL), dried (Na 2 S0 4 ) and concentrated in vacuo.
  • A/-Chlorosuccinimide (0.46 g, 3.46 mmol) was added portion-wise to a solution of 1 ,2,3,5,6,7-hexahydro-s-indacen-4-amine, 1 (0.6 g, 3.46 mmol) in CHCI 3 (10 mL) at 0 °C. The solution was gradually warmed to ambient temperature and stirred for 10 h. The reaction mixture was diluted with sat. aqueous Na 2 S 2 0 3 (50 mL) and extracted with DCM (2 x 25 mL). The combined organic extracts were washed with water (25 mL), brine (25 mL), dried (Na 2 S0 4 ) and concentrated in vacuo.
  • aluminium trichloride (2 g, 15 mmol) was added to anhydrous 1 ,2-dichloroethane (40 mL) at 0 °C followed by the acid chloride solution (10 mL) drop-wise over 5 min and the resulting solution was stirred for 30 min at 0 °C.
  • a further portion of aluminium trichloride (3 g, 22.5 mmol) was added followed by drop-wise addition of the remaining acid chloride solution (20 mL) at 0 °C.
  • the reaction mixture was stirred at room temperature for 1 h or until completion, diluted with water and extracted using EtOAc (2 x 50 mL).
  • N-(3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-yl)pivalamide (0.55 g, 2.12 mmol) in acetic acid (10 mL) was treated drop-wise with a solution of bromine (0.4 g, 2.55 mmol) in acetic acid (2.0 mL) and the reaction stirred at ambient temperature for 3 h. Ice cold water was added to the reaction mixture and stirred for 10 min.
  • N-(4-bromo-3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-yl)pivalamide (0.6 g, 1 .78 mmol) in EtOH (10 mL) and cHCI (15 mL) was heated at 90 °C for 36 h. The solution was concentrated in vacuo then basified using aq NH 4 OH solution.
  • 2,4,6-Trichloropyrimidine (2.7 g, 14.7 mmol) was dissolved in anhydrous THF (30 mL) at 0 °C under nitrogen atmosphere.
  • Cul (280 mg, 1.47 mmol) was added to the aforementioned solution and subsequently treated with 2M tert- butylmagnesium chloride in THF (3.78 g, 16.15 mL, 32.3 mmol) at 0 °C under nitrogen atmosphere.
  • the resulting mixture was stirred at RT for 3 h.
  • the reaction mixture was diluted with saturated NH 4 CI solution and extracted with EtOAc (2 x 50 mL).
  • A/-(2,3-dihydro-1 /-/-inden-4-yl)acetamide 200 mg, 1 .1 1 mmol was dissolved in AcOH (5 mL) and cooled to 0 °C.
  • A/-Chlorosuccinimide (220 mg, 1 .69 mmol) was added then the reaction mixture was warmed to RT and strirred overnight.
  • the aldehyde (0.68 g, 3.58 mmol) was oxidized using silver (I) oxide (1 .5 eq.) in 5% aqueous sodium hydroxide at rt for 20 days.
  • the crude reaction mixture was filtered through celite, extracted using diethyl ether (2 x 50 mL) to remove unreacted aldehyde then the aqueous phase was acidified to pH 1 using 3.0 M aqueous HCI drop-wise at 0 °C.
  • the aldehyde (0.5 g, 2.77 mmol) in acetone (5.0 mL) was treated with sulfamic acid (0.4 g, 4.17 mmol) in two portions at 0 °C.
  • a solution of sodium chlorite (0.32 g, 3.6 mmol) in water (1 .0 mL) was added drop-wise and stirring continued at 0 °C for 4 h.
  • the reaction mixture was diluted with water (20 mL) and extracted using 10% IPA/chloroform (2 x 20 mL). The combined organics were washed with water (25 mL), brine (25 mL), dried (Na 2 S0 4 ) and concentrated in vacuo.
  • Methyl 2,3-dihydroxybenzoate (1.0 g, 5.95 mmol) in DMF (16 mL) was treated with KF (1 .79 g, 30.9 mmol) and stirred at ambient temperature for 30 minutes. Diiodomethane (1 .79 g, 6.7 mmol) was added and the reaction heated at 100 °C for 5 hours. The reaction mixture was cooled to rt, poured onto water (100 mL) and extracted using diethyl ether (2 x 50 mL). The combined organics were washed with water (50 mL), brine(50 mL), dried (MgS0 4 ) and concentrated in vacuo.
  • Phenyl chloroform ate (1 .5 eq) was added slowly to a solution of quinolin-8- amine (1 g, 6.9 mmol) in THF (10 mL) and triethylamine (2 eq.) to 0 °C. The solution was stirred at room temperature for 2 h or until completion. The solution was diluted using sat.aq. NaHC03 solution, extracted using ethyl acetate (2 x 50 mL), washed with water, brine then dried (Na 2 S0 4 ) and concentrated in vacuo.

Abstract

ABSTRACT The present invention provides for certain sulfonyl ureas and related compounds which have advantageous properties and show useful activity in the inhibition of activation of the NLRP3 inflammasome. Such compounds are useful in the treatment of a wide range of disorders in which the inflammation process, or more specifically the NLRP3 inflammasome, have been implicated as being a key factor.

Description

SULFONYLUREAS AND RELATED COMPOUNDS AND USE OF SAME
FIELD OF THE INVENTION
[0001 ] The invention relates to the field of medical treatment and diagnosis of disease. More particularly, this invention relates to novel sulfonylurea and related compounds and their use in treating, or identifying a disease or condition responsive to modulation of NLRP3 or inhibition of the activation of NLRP3 or related components of the inflammatory process.
BACKGROUND TO THE INVENTION
[0002] Any reference to background art herein is not to be construed as an admission that such art constitutes common general knowledge in Australia or elsewhere.
[0003] The NOD-like receptor (NLR) family, pyrin domain-containing protein 3 (NLRP3) inflammasome is a component of the inflammatory process, and its aberrant activation is pathogenic in inherited disorders such as cryopyrin- associated periodic syndromes (CAPS) and complex diseases such as multiple sclerosis, type 2 diabetes, Alzheimer's disease and atherosclerosis.
[0004] NLRP3 is an intracellular signalling molecule that senses many pathogen-derived, environmental and host-derived factors. Upon activation, NLRP3 binds to apoptosis-associated speck-like protein containing a caspase activation and recruitment domain (ASC). ASC then polymerises to form a large aggregate known as an ASC speck. Polymerised ASC in turn interacts with the cysteine protease caspase-1 to form a complex termed the inflammasome. This results in the activation of caspase-1 , which cleaves the proinflammatory cytokines IL-1 β and IL-18 to their active forms and mediates a type of inflammatory cell death known as pyroptosis. The ASC speck can also recruit and activate caspase-8, which can process pro-IL-1 β and pro-IL-18 and trigger apoptotic cell death.
[0005] Caspase-1 cleaves pro-IL-1 β and pro-IL-18 to their active forms, which are secreted from the cell. Active caspase-1 also cleaves gasdermin-D to trigger pyroptosis. Through its control of the pyroptotic cell death pathway, caspase-1 also mediates the release of alarm in molecules such as IL-33 and high mobility group box 1 protein (HMGB1 ). Caspase-1 also cleaves intracellular IL-1 R2 resulting in its degradation and allowing the release of IL- 1 a. In human cells caspase-1 may also control the processing and secretion of IL-37. A number of other caspase-1 substrates such as components of the cytoskeleton and glycolysis pathway may contribute to caspase-1 -dependent inflammation.
[0006] NLRP3-dependent ASC specks are released into the extracellular environment where they can activate caspase-1 , induce processing of caspase- 1 substrates and propagate inflammation.
[0007] Active cytokines derived from NLRP3 inflammasome activation are important drivers of inflammation and interact with other cytokine pathways to shape the immune response to infection and injury. For example, I L-1 β signalling induces the secretion of the pro-inflammatory cytokines IL-6 and TNF. IL-1 β and I L- 8 synergise with IL-23 to induce I L- 7 production by memory CD4 Th17 cells and by γδ T cells in the absence of T cell receptor engagement. IL- 18 and IL-12 also synergise to induce IFN-γ production from memory T cells and NK cell driving a Th1 response.
[0008] Other intracellular pattern recognition receptors (PRRs) are also capable of forming inflammasomes. These include other NLR family members such as NLRP1 and NLRC4, as well as non-NLR PRRs such as the double- stranded DNA (dsDNA) sensors absent in melanoma 2 (AIM2) and interferon, gamma inducible protein 16 (IFI16). NLRP3-dependent IL-1 β processing can also be activated by an indirect, non-canonical pathway downstream of caspase-1 1 .
[0009] The inherited CAPS diseases Muckle-Wells syndrome (MWS), familial cold autoinflammatory syndrome and neonatal-onset multisystem inflammatory disease are caused by gain-of-function mutations in NLRP3, thus defining NLRP3 as a critical component of the inflammatory process. NLRP3 has also been implicated in the pathogenesis of a number of complex diseases, notably including metabolic disorders such as type 2 diabetes, atherosclerosis, obesity and gout.
[0010] A role for NLRP3 in diseases of the central nervous system is emerging, and lung diseases have also been shown to be influenced by NLRP3. Furthermore, NLRP3 has a role in the development of liver disease, kidney disease and aging. Many of these associations were defined using Nlrp3 mice, but there have also been insights into the specific activation of NLRP3 in these diseases. In type 2 diabetes, the deposition of islet amyloid polypeptide in the pancreas activates NLRP3 and I L- β signaling, resulting in cell death and inflammation.
[001 1 ] Several small molecules have been shown to inhibit the NLRP3 inflammasome. Glyburide inhibits IL-1 β production at micromolar concentrations in response to the activation of NLRP3 but not NLRC4 or NLRP1 . Other previously characterised NLRP3 inhibitors include parthenolide, 3,4-methylenedioxy-P-nitrostyrene and dimethyl sulfoxide (DMSO), although these agents have limited potency and are nonspecific
[0012] Current treatments for NLRP3-related diseases include biologic agents that target IL-1 . These are the recombinant IL-1 receptor antagonist anakinra, the neutralizing IL-1 β antibody canakinumab and the soluble decoy IL-1 receptor rilonacept. These approaches have proven successful in the treatment of CAPS, and these biologic agents have been used in clinical trials for other IL-^-associated diseases.
[0013] Several small molecules have been shown to inhibit the NLRP3 inflammasome. Glyburide inhibits IL- β production at micromolar concentrations in response to the activation of NLRP3 but not NLRC4 or NLRP1 .Other previously characterised NLRP3 inhibitors include parthenolide, 3,4-methylenedioxy-P-nitrostyrene and dimethyl sulfoxide (DMSO), although these agents have limited potency and are nonspecific.
[0014] Certain diarylsulfonylurea-containing compounds have been identified as cytokine release inhibitory drugs (CRIDs) (Perregaux et al.; J. Pharmacol. Exp. Ther. 299, 187-197, 2001 ). CRIDs are a class of diarylsulfonylurea containing compounds that inhibit the post-translational processing of IL-1 β. Post-translational processing of IL-1 β is accompanied by activation of caspase-1 and cell death. CRIDs arrest activated monocytes so that caspase-1 remains inactive and plasma membrane latency is preserved.
[0015] There is a need to provide compounds with improved pharmacological and/or physiological and or physicochemical properties and/or those that provide a useful alternative to known compounds.
SUMMARY OF INVENTION
[0016] According to a first aspect of the invention, there is provided a compound of formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000006_0001
Formula (I) wherein, W is selected from 0, S and Se; J is selected from S and Se;
Ri is selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted;
R2 is selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted; and both Ri is directly bonded to J and R2 is directly bonded to the adjacent nitrogen, via a carbon atom .
[0017] According to a second aspect of the invention there is provided a pharmaceutical composition comprising a compound of the first aspect, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier, diluent and/or excipient.
[0018] A third aspect of the invention resides in a method of treatment or prevention of a disease, disorder or condition including the step of administering an effective amount of a compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof, or the pharmaceutical composition of the second aspect to thereby treat or prevent the disease disorder or condition.
[0019] A fourth aspect of the invention provides for a compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof, or the pharmaceutical composition of the second aspect for use in the treatment or prevention of a disease, disorder or condition.
[0020] A fifth aspect of the invention provides for use of a compound of the first aspect, or a pharmaceutically effective salt, solvate or prodrug thereof, in the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition. [0021 ] In one embodiment, the disease, disorder or condition is responsive to inhibition of activation of the NLRP3 inflammasome.
[0022] In particular non-limiting embodiments of the above aspects, the disease, disorder or condition is a disease, disorder or condition of the immune system, the cardiovascular system, the endocrine system, the gastrointestinal tract, the renal system, the respiratory system, the central nervous system, is a cancer or other malignancy and/or is caused by or associated with a pathogen.
[0023] In a sixth aspect of the invention there is provided a method of diagnosing a disease, disorder or condition in a mammal including the step of administering a labelled compound of formula (I), (la), (lb), (Ic) or (I I), or a pharmaceutically effective salt, solvate or prodrug thereof, to the mammal or to a biological sample obtained from the mammal to facilitate diagnosis of the disease disorder or condition in the mammal.
[0024] A seventh aspect of the invention resides in a method of modulating the activity of a biological target comprising the step of exposing the biological target to a compound of the first aspect, or a pharmaceutically acceptable salt thereof.
[0025] The biological target may be selected from the group consisting of the NLRP3 inflammasome, ΙΙ_-1 β, IL-17, IL-18, IL-1 a, IL-37, IL-33 and Th17 cells.
[0026] The various features and embodiments of the present invention, referred to in individual sections above apply, as appropriate, to other sections, mutatis mutandis. Consequently features specified in one section may be combined with features specified in other sections as appropriate.
[0027] Further features and advantages of the present invention will become apparent from the following detailed description. BRIEF DESCRIPTION OF THE DRAWINGS
[0028] In order that the invention may be readily understood and put into practical effect, preferred embodiments will now be described by way of example with reference to the accompanying figures wherein:
[0029] FIG 1 A to 1 C is a series of graphical representations of the plasma concentrations of a known sulfonylurea (MCC950) following different dosing levels in mice; and
[0030] FIG 2A to 2C is a series of graphical representations of the plasma concentrations of a sulfonylurea of the present invention (MCC7840) following different dosing levels in mice.
DETAILED DESCRIPTION
[0031 ] The present invention is predicated, at least in part, on the finding that certain sulfonyl ureas and related compounds have advantageous properties and show useful activity in the inhibition of activation of the NLRP3 inflammasome and/or inhibition of IL-1 β and/or IL-17 and/or IL-18, and/or IL-1 , and/or IL-37, and/or IL-33 as well as interfere with or modulate the activity of T helper cells such as Th17. Particularly, the compounds of the invention are useful in the treatment of a wide range of disorders in which the inflammation process, or the NLRP3 inflammasome and/or IL-1 β and/or IL-17 and/or IL-18, and/or IL-1 a, and/or IL-37, and/or IL-33 and/or Th17 cells play a part.
[0032] Evidence from human CAPS patients and mouse models of CAPS has lead the present inventors to believe that NLRP3 inhibition will be a superior treatment over IL-1 biologies, as inhibition of all NLRP3-dependent processes will be more effective than inhibition of a single NLRP3-dependent process, such as IL-1 signalling.
[0033] Individuals with CAPS display dysregulated secretion of both IL-1 β and IL-18, and CAPS patients treated with anti-IL-1 biologies have residual disease. Symptoms such as bony overgrowth and joint deformity are not prevented by IL-1 biologies. In addition, symptoms involving the central nervous system such as hearing loss are difficult to control using IL-1 biologies, which appear to poorly penetrate the central nervous system. Studies in mouse models of CAPS indicate that deficiency in either IL-1 signalling or IL-18 alone is insufficient to block systemic inflammation, particularly in older animals. In a severe model of CAPS, only a complete loss of caspase-1 signalling fully rescued the disease.
[0034] Specific inhibition of NLRP3 by sulfonyurea-containing compounds, such as those of the first aspect, may block all processes downstream of NLRP3, including ASC speck formation and caspase-8 and caspase-1 activation. Consequently, NLRP3 inhibition will block all caspase-1 dependent processes such as IL-1 β , IL-18 and IL-37 processing and secretion, gasdermin D cleavage, pyroptosis, and release of IL-1 a, IL-33 and HMGB. Furthermore, NLRP3-dependent extracellular release of the ASC speck will be blocked, and caspase-8-dependent pro-IL-1 β and pro-IL-18 cleavage and apoptotic cell death will be prevented. Thus, specific inhibition of NLRP3 by compounds of the first aspect will prevent multiple downstream inflammatory signals and should therefore prove more effective anti-inflammatory therapy than IL-1 blockade alone.
[0035] Anti-IL-1 biologies block IL-1 derived from NLRP3-independent sources, such IL-1 produced by other inflammasomes (e.g. NLRC4, NLRP1 , NLRP6, AIM2) and IL-1 generated by the latter pathways may be important for host defence against pathogens. For example, patients receiving IL-1 /IL-1 R antagonists exhibit increased incidence of upper airway infections. Specific inhibition of NLRP3 by the present compounds may thus exert less generalised immunosuppression compared to anti-IL-1 biologies. [0036] IL-1 β and IL-18, generated by the Nlrp3/caspase-1 axis, play critical roles in driving IL-17 production by CD4 Th17 cells and γδ T cells. IL-1 β and IL- 18 synergise with IL-23 to induce IL-17 production by memory CD4 Th17 cells and by γδ T cells in the absence of TCR engagement. IL-1 -driven IL-17 has also been implicated in psoriasis, type I diabetes, rheumatoid arthritis, type 2 diabetes mellitus, atherosclerosis, obesity, gout, and recently, asthma.
[0037] In essence, each of these diseases has been shown to involve the activation of tissue macrophages, dendritic cells, or brain microglia, driven by either soluble alarmins, or the frustrated phagocytosis of metabolites that accumulate extracellularly. NLRP3 senses these events, leading to IL-1 release, triggering inflammation to clear the offensive material. Disease will result if this process becomes chronic or over-activated, which explains why so many diseases have been shown to involve NLRP3. Inhibitors that act to prevent NLRP3 activation hence can have utility in IL-17 driven, as well as IL-1 driven diseases.
[0038] In this patent specification, the terms 'comprises', 'comprising', 'includes', 'including', or similar terms are intended to mean a non-exclusive inclusion, such that a method or composition that comprises a list of elements does not include those elements solely, but may well include other elements not listed.
[0039] Unless defined otherwise, all technical and scientific terms used herein have the same meaning as would be commonly understood by those of ordinary skill in the art to which this invention belongs.
[0040] The term "pharmaceutically acceptable salt", as used herein, refers to salts which are toxicologically safe for systemic or localised administration such as salts prepared from pharmaceutically acceptable non-toxic bases or acids including inorganic or organic bases and inorganic or organic acids. The pharmaceutically acceptable salts may be selected from the group including alkali and alkali earth, ammonium, aluminium, iron, amine, glucosamine, chloride, sulphate, sulphonate, bisulphate, nitrate, citrate, tartrate, bitarate, phosphate, carbonate, bicarbonate, malate, maleate, napsylate, fumarate, succinate, acetate, benzoate, terephthalate, palmoate, piperazine, pectinate and S-methyl methionine salts and the like.
[0041 ] The term "alkyl' refers to a straight-chain or branched alkyl substituent containing from, for example, 1 to about 12 carbon atoms, preferably 1 to about 9 carbon atoms, more preferably 1 to about 6 carbon atoms, even more preferably from 1 to about 4 carbon atoms, still yet more preferably from 1 to 2 carbon atoms. Examples of such substituents may be selected from the group consisting of methyl, ethyl, propyl, isopropyl, n-butyl, sec-butyl, isobutyl, terf-butyl, pentyl, isoamyl, 2-methylbutyl, 3-methylbutyl, hexyl, heptyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2-ethylbutyl, 3- ethylbutyl, octyl, nonyl, decyl, undecyl, dodecyl and the like. The number of carbons referred to relates to the carbon backbone and carbon branching but does not include carbon atoms belonging to any substituents, for example the carbon atoms of an alkoxy substituent branching off the main carbon chain. Substituted alkyl includes alkyl substituted with one or more moieties selected from the group consisting of halo {e.g., CI, F, Br, and I); halogenated alkyl {e.g., CF3, 2-Br-ethyl, CH2F, CH2CI, CH2CF3, or CF2CF3); hydroxyl; amino; carboxylate; carboxamido; alkylamino; arylamino; alkoxy; aryloxy; nitro; azido; cyano; thio; sulfonic acid; sulfate; phosphonic acid; phosphate; and phosphonate as well as those described under the definition of Optionally substituted'.
[0042] The term "alkenyf refers to optionally substituted unsaturated linear or branched hydrocarbon groups, having 2 to 12 carbon atoms, preferably 2 to 9 carbon atoms, more preferably 2 to 6 carbon atoms and having at least one carbon-carbon double bond. Where appropriate, the alkenyl group may have a specified number of carbon atoms, for example, C2-C6 alkenyl which includes alkenyl groups having 2, 3, 4, 5 or 6 carbon atoms in linear or branched arrangements. The number of carbons referred to relates to the carbon backbone and carbon branching but does not include carbon atoms belonging to any substituents. Examples of such substituents may be selected from the group consisting of ethenyl, propenyl, isopropenyl, butenyl, s- and t-butenyl, pentenyl, hexenyl, hept-l,3-diene, hex-l,3-diene, non-l,3,5-triene and the like. Substituted alkenyl includes alkenyl substituted with one or more moieties selected from the group consisting of halo {e.g., CI, F, Br, and I); halogenated alkyl {e.g., CF3, 2-Br-ethyl, CH2F, CH2CI, CH2CF3, or CF2CF3); hydroxyl; amino; carboxylate; carboxamido; alkylamino; arylamino; alkoxy; aryloxy; nitro; azido; cyano; thio; sulfonic acid; sulfate; phosphonic acid; phosphate; and phosphonate as well as those described under the definition of Optionally substituted'.
[0043] The term "alkoxy" as used herein means straight or branched chain alkyl groups linked by an oxygen atom (i.e., -O-alkyl), wherein alkyl is as described above. In particular embodiments, alkoxy refers to oxygen-linked groups comprising 1 to 10 carbon atoms ("C1 -10 alkoxy"). In further embodiments, alkoxy refers to oxygen-linked groups comprising 1 to 8 carbon atoms ("C1 -8 alkoxy"), 1 to 6 carbon atoms ("C 1 -6 alkoxy"), 1 to 4 carbon atoms ("C1 -4 alkoxy") or 1 to 3 carbon atoms ("C1 -3 alkoxy').
[0044] The terms "cycloalkyi" and "cycloalkenyl" refers to optionally substituted saturated and unsaturated mono-cyclic, bicyclic or tricyclic carbon groups. Where appropriate, the cycloalkyi or cycloalkenyl group may have a specified number of carbon atoms, for example, C3-C6 cycloalkyi or cycloalkenyl includes within its scope a carbocyclic group having 3, 4, 5 or 6 carbon atoms. Examples of such substituents may be selected from the group consisting of cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl and the like. Substituted cycloalkyl or cycloalkenyl includes substitutions with one or more moieties selected from the group consisting of halo (e.g. , CI, F, Br, and I); halogenated alkyl (e.g. , CF3, 2-Br-ethyl, CH2F,
CH2CI, CH2CF3, or CF2CF3); hydroxyl; amino; carboxylate; carboxamido; alkylamino; arylamino; alkoxy; aryloxy; nitro; azido; cyano; thio; sulfonic acid; sulfate; phosphonic acid; phosphate; and phosphonate as well as those described under the definition of Optionally substituted' .
[0045] The term "alkylthio" as used herein means a thio group with one or more alkyl substituents, where alkyl is defined as above.
[0046] The term "amino" as used herein means a moiety represented by the structure NR23, and includes primary amines, and secondary and tertiary amines substituted by alkyl (i.e., alkylamino). Thus, R23 may represent, for example, two hydrogen atoms, two alkyl moieties, or one hydrogen atom and one alkyl moiety.
[0047] The term :'aryi" refers to a stable monocyclic, bicyclic, or tricyclic carbon ring of up to 8 members in each ring, wherein at least one ring is aromatic as defined by the HCickel 4n+2 rule. The term includes polycyclic systems comprising saturated carbon rings or heteroaryl or heterocyclic groups so long as at least one ring is aryl, as described.
[0048] The terms "aralkyl" and "arylalkyl" as used herein mean an aryl group as defined above linked to the molecule through an alkyl group as defined above.
[0049] The term "heteroaryl" refers to an aryl group containing from one or more (particularly one to four) non-carbon atom(s) (particularly N, O or S) or a combination thereof, which heteroaryl group is optionally substituted at one or more carbon or nitrogen atom(s). Heteroaryl rings may also be fused with one or more cyclic hydrocarbon, heterocyclic, aryl, or heteroaryl rings. Heteroaryl includes, but is not limited to, 5-membered heteroaryls having one hetero atom (e.g. , thiophenes, pyrroles, furans); 5 membered heteroaryls having two heteroatoms in 1 ,2 or 1 ,3 positions (e.g., oxazoles, pyrazoles, imidazoles, thiazoles, purines); 5-membered heteroaryls having three heteroatoms (e.g., triazoles, thiadiazoles); 5-membered heteroaryls having four heteroatoms (e.g., tetrazoles); 6-membered heteroaryls with one heteroatom (e.g., pyridine, quinoline, isoquinoline, phenanthrine, 5,6-cycloheptenopyridine); 6-membered heteroaryls with two heteroatoms (e.g., pyridazines, cinnolines, phthalazines, pyrazines, pyrimidines, quinazolines); 6-membered heretoaryls with three heteroatoms (e.g., 1 ,3,5- triazine); and 6-membered heteroaryls with four heteroatoms. "Substituted heteroaryl" means a heteroaryl having one or more non-interfering groups as substituents and including those defined under Optionally substituted'.
[0050] "Heterocyclyl" as used herein refers to a non-aromatic ring having 5 to 8 atoms in the ring and of those atoms 1 to 4 are heteroatoms. Heterocyclic rings may also be fused with one or more cyclic hydrocarbon, heterocyclic, aryl, or heteroaryl rings. Heterocyclic includes partially and fully saturated heterocyclic groups. Heterocyclic systems may be attached to another moiety via any number of carbon atoms or heteroatoms of the radical and may be both saturated and unsaturated. Non-limiting examples of heterocyclic include C4-C6 selenocycles, pyrrolidinyl, pyrrolinyl, pyranyl, piperidinyl, piperazinyl, morpholinyl, tetrahydrofuranyl, tetrahydrothiophenyl, pyrazolinyl, dithiolyl, oxathiolyl, dioxanyl, dioxinyl, oxazinyl, azepinyl, diazepinyl, thiazepinyl, oxepinyl and thiapinyl, imidazolinyl, thiomorpholinyl, and the like.
[0051 ] "Optionally substituted" in reference to a substituent group refers to substituent groups optionally substituted with one or more moieties, for example, those selected from the group consisting of optionally substituted C1 - 10 alkyl (e.g., optionally substituted C1 -6 alkyl); optionally substituted C3-6 cycloalkyl (e.g., optionally substituted cyclopropyl); optionally substituted hydroxyalkyl; optionally substituted C1 -10 alkoxy (e.g., optionally substituted C1 -6 alkoxy); optionally substituted C2-10 alkenyl; optionally substituted C2-10 alkynyl; optionally substituted C6-C12 aryl; aryloxy; optionally substituted heteroaryl; optionally substituted heterocyclyl; halo (e.g., CI, F, Br, and I); hydroxyl; halogenated alkyl (e.g., CF3, 2-Br-ethyl, CH2F, CH2CF3, and CF2CF3); amino (e.g., NH2, NR-|2H, and NR-|2R-|3); alkylamino; arylamino; acyl; amido; CN; N02; N3; CH2OH; CONH2; CONR24R25; C02R24; CH2OR24; NHCOR24; NHCO2R24; C1 -3 alkylthio; sulfate; sulfonic acid; sulfonate esters such as alkyl or aralkyl sulfonyl, including methanesulfonyl; phosphonic acid; phosphate; phosphonate; mono-, di-, or triphosphate esters; trityl or monomethoxytrityl; R2 SO; R24S02; CF3S; and CF3S02; trialkylsilyl such as dimethyl-t-butylsilyl or diphenylmethylsilyl; and R24 and R25 are each independently selected from H or optionally substituted C1 -10 alkyl, C1 -6 alkyl or C1 -4 alkyl.
[0052] Whenever a range of the number of atoms in a structure is indicated (e.g. , a Ci -Ci2, C-i-C-io, C1-C9, C1-C6, Ci-C4, or C2-C2o, C2-C-|2, C2-Cio, C2-C9, C2-C8, C2-C6, C2-C4 alkyl, alkenyl, etc.), it is specifically contemplated that any sub-range or individual number of carbon atoms falling within the indicated range also can be used. Thus, for instance, the recitation of a range of 1 -12 carbon atoms (e.g. , d-C-i2), 1 -9 carbon atoms (e.g. , C1 -C9), 1 -6 carbon atoms (e.g. , C-1 -C6), 1 -4 carbon atoms (e.g. , Ci-C4), 1 -3 carbon atoms (e.g., Ci-C3), or 2-8 carbon atoms (e.g., C2-C8) as used with respect to any chemical group (e.g. , alkyl, etc.) referenced herein encompasses and specifically describes 1 , 2, 3, 4, 5, 6, 7, 8, 9, 10, 1 1 , and/or 12 carbon atoms, as appropriate, as well as any sub-range thereof (e.g., 1 -2 carbon atoms, 1 -3 carbon atoms, 1 -4 carbon atoms, 1 -5 carbon atoms, 1 -6 carbon atoms, 1 -7 carbon atoms, 1 -8 carbon atoms, 1 -9 carbon atoms, 1 -10 carbon atoms, 1 -1 1 carbon atoms, 1 -12 carbon atoms, 2-3 carbon atoms, 2-4 carbon atoms, 2-5 carbon atoms, 2-6 carbon atoms, 2-7 carbon atoms, 2-8 carbon atoms, 2-9 carbon atoms, 2-10 carbon atoms, 2-1 1 carbon atoms, 2-12 carbon atoms, 3-4 carbon atoms, 3-5 carbon atoms, 3-6 carbon atoms, 3-7 carbon atoms, 3-8 carbon atoms, 3-9 carbon atoms, 3-10 carbon atoms, 3-1 1 carbon atoms, 3-12 carbon atoms, 4-5 carbon atoms, 4-6 carbon atoms, 4-7 carbon atoms, 4-8 carbon atoms, 4-9 carbon atoms, 4-10 carbon atoms, 4-1 1 carbon atoms, and/or 4-12 carbon atoms, etc., as appropriate).
[0053] According to a first aspect of the invention, there is provided a compound of formula (I), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000017_0001
Formula (I) wherein, W is selected from O, S and Se; J is selected from S and Se;
R-i is selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted;
R2 is selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted; and both R-i is directly bonded to J and R2 is directly bonded to the adjacent nitrogen, via a carbon atom.
[0054] In one preferred embodiment, W is O.
[0055] In one preferred embodiment, J is S.
[0056] In a particularly preferred embodiment, W is O and J is S. [0057] In one embodiment, Ri is selected from the group consisting of C5 or C6 cycloalkyl, 5-membered or 6-membered heteroaryl, bicyclic heteroaryl wherein at least one ring is heteroaryl, phenyl, biphenyl, phenylheterocyclyl, 5- membered or 6-membered heterocyclyl, and heterocyclylcycloalkyl, all of which may be optionally substituted.
[0058] In certain embodiments, W is O, J is S and R-i is selected from the group consisting of pyrazole, furan, tetrahydrofuran, tetrahydropyran, pyran, pyrrolidine, pyrrole, triazole, tetrazole, imidazole, pyridine, morpholine, piperazine, piperidine, substituted phenyl, phenylheteroaryl, phenylheterocyclyl, biphenyl, quinoline, isoquinoline, naphthyl, pyrazine and pyrimidine, all of which may be optionally substituted as appropriate.
[0059] In one embodiment, when W is O, J is S and R-i is 2-furan or 2- thiophene it is selected from unsubstituted 2-furan or 2,5-substituted furan and unsubstituted 2-thiophene or 2,5-substituted thiophene.
[0060] In one embodiment, when W is O, J is S, and Ri is 2,5-substituted furan or 2,5-substituted thiophene then the 2,5-substituted furan or 2,5- substituted thiophene is not substituted with a tertiary alcohol group.
[0061 ] In certain embodiments it has been found that when R-i is an unsubstituted furan then it has the ability to cross the blood brain barrier at levels about 10 times greater than CRID3, a prior art sulfonylurea.
[0062] In the above embodiments, reference to 2,5-substituted does not preclude the presence of further substitutions on the ring but merely indicates that the numbered substitutions must be present. For example, 2,4,5- substitutions are considered within the scope of such terms.
[0063] Reference to 2-furan and 2-thiophene means that the ring is connected to the sulfonyl sulphur at the 2-ring position, as shown below:
Figure imgf000019_0001
[0064] In one embodiment, R-i is 5-membered heterocyclyl or heteroaryl, each of which may be optionally substituted, comprising at least one, preferably at least two ring heteroatoms selected from N, 0 and S.
[0065] In certain embodiments, Ri is a nitrogen heterocyclyl or nitrogen heteroaryl, each of which may be optionally substituted.
[0066] In one embodiment, Ri is 5-membered nitrogen heterocyclyl or 5- membered nitrogen heteroaryl, each of which may be optionally substituted.
[0067] In an embodiment, Ri is 5-membered heterocyclyl or 5-membered heteroaryl, each comprising at least two ring nitrogen atoms and each of which rings may be optionally substituted.
[0068] In one embodiment, W is O, J is S and R-i is selected from the group consisting of quinoline, isoquinoline, naphthyl, pyrazine, tetrazole, imidazole, pyrrolidine, pyrrole, tetrahydropyran, pyran, piperidine, piperazine, pyrazole, pyridine, pyrimidine and triazole, each of which may be optionally substituted.
[0069] In one embodiment R-i and/or R2 may comprise a selenocycle.
[0070] In one embodiment, R2 may be selected from bicyclic and tricyclic hydrocarbons, 5-, 6- and 7-membered heterocycle or heteroaryl, each of which rings may be optionally substituted, and substituted phenyl.
[0071 ] Suitably, the tricyclic hydrocarbon may be an indacene.
[0072] In one embodiment, R2 may be selected from 5-, 6- or 7- membered nitrogen heterocycles, 6-membered nitrogen heteroaryl and aryl with fused cycloalkyl ring. [0073] In one embodiment of the compound of formula (I), W is 0, J is S and Ri may be selected from the group consisting of:
Figure imgf000020_0001
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
[0074] In any embodiment of the first aspect, when J is S and W is 0, and in combination with any of the R-i groups listed above, R2 may be selected from:
Figure imgf000023_0002
wherein, each incidence of Y is independently selected from C, N, S and O, and which may be optionally substituted, as appropriate;
R5, Rii, R-I2, R-I3, R-I4 and R-|5 are independently selected from the group consisting of hydrogen, halo, cyano, amide, sulphonamide, acyl, hydroxyl, Ci- Ce alkyl, d-C6 haloalkyi, Cs-Cs cyloalkyl, and C-1 -C6 alkoxy, all of which groups may be optionally substituted, as appropriate, with halo, cyano or C C6 alkoxy; and wherein R-n and R 2 may combine to form phenyl, a 5- or 6-membered oxygen heterocycle or a 5- or 6-membered nitrogen heteroaryl, each of which may be optionally substituted;
R-12 and R-13 may combine to form a 5- or 6-membered nitrogen heteroaryl, which may be optionally substituted; and
RH and R15 may combine to form a 5- or 6-membered cycloalkyl ring, phenyl, a 5- or 6-membered oxygen heterocycle or a 5- or 6-membered nitrogen heteroaryl, each of which may be optionally substituted. [0075] Suitably, each incidence of Y is a carbon and R5 is hydrogen or halo.
[0076] In one embodiment, R 2 and R 4 are hydrogen, R-n and R 5 are C C6 alkyl and R-|3 is hydrogen or halo.
[0077] Preferably, R2 is selected from a substituted or hydrogenated indacene, a 2,6-dialkylphenyl, a 2,6-dialkyl-4-halophenyl, 2,6-dicycloalkylphenyl, and a 2,6-dicycloalkyl-4-halophenyl.
[0078] In certain preferred embodiments, and in combination with any Ri group described for any of the formulae of the first aspect, R2 is selected from hexahydroindacene, 2,6-diisopropylphenyl 2,6-diisopropyl-4-chlorophenyl, 2,6- dicyclopropylphenyl and 2,6-dicyclopropyl-4-chlorophenyl.
[0079] In one embodiment, W is O and J is S, Ri is heteroaryl and R2 is
Figure imgf000024_0001
wherein each Y is CH and R5 is H or halogen, preferably R5 is H.
In one embodiment, W is O and J is S, Ri is heteroaryl and R2
Figure imgf000025_0001
wherein
R-11 and Ri5 are Ci-6 alkyl, preferably isopropyl;
Figure imgf000025_0002
Ri 3 is H or halogen, preferably H or CI.
] In one embodiment W is 0 and J is S, Ri is heteroaryl and R2
Figure imgf000025_0003
wherein R-,-, and R 5 are isopropyl, R 2 and R are H, and R 3 is H or CI.
[0082] In particular embodiments, the compound of formula (I) may be selected from a compound of formula (la), (lb) and (Ic), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000026_0001
formula (la) formula (lb) formula (lc) wherein, R-i is as previously described for any embodiment of formula (I).
[0083] In one embodiment of the compound of formula (la), (lb) and (lc), Ri is selected from the group consisting of pyrazole, furan, tetrahydrofuran, tetrahydropyran, pyran, pyrrolidine, pyrrole, triazole, tetrazole, imidazole, pyridine, morpholine, piperazine, piperidine, substituted phenyl, phenylheteroaryl, phenylheterocyclyl, biphenyl, quinoline, isoquinoline, naphthyl, pyrazine and pyrimidine, all of which may be optionally substituted as appropriate.
[0084] In one embodiment of the compound of formula (la), (lb) and (lc), R-i is selected from the group consisting of:
Figure imgf000026_0002
Figure imgf000027_0001
Figure imgf000028_0001
[0085] In one embodiment, the compound of formula (I) may be selected from a compound of formula (II), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000028_0002
Formula (II) wherein, A, B, D and E are independently selected from C, N, 0, S and Se but at least one thereof is C; each dashed line may represent a bond;
R2 is as previously defined for any embodiment of formula (I), (la), (lb) or (lc), or may be a fluorescent group; each incidence of R6 is independently selected from the group consisting of hydrogen, halo, cyano, CrC6 alkyl, C C6 alkylamino, CrC6 alkylhydroxy, C3- Ce cycloalkyi, alkylphenyl, phenyl, benzyl, C-i-Ce ester, C2-C6 alkenyl, C-i-Ce t fluoroalkyl and C C6 alkoxy, each of which may be optionally substituted, or R6 may be a fluorescent group. [0086] In one preferred embodiment of the compound of formula (II), at least one of A, B, D and E is N (i.e. nitrogen).
[0087] In a further preferred embodiment of the compound of formula (II), at least two of A, B, D and E are N.
[0088] In one embodiment of the compound of formula (II), A, B, D and E are selected from N and C.
[0089] In a further embodiment of the compound of formula (II), A is C and at least two of B, D and E are N.
[0090] In one embodiment, A, B, D and E form a ring selected from a pyrazole, an imidazole, a triazole, and a tetrazole.
[0091 ] Preferably, A, B, D, and E form a ring selected from a pyrazole or an imidazole ring, most preferably a pyrazole ring.
[0092] In one embodiment A, B, D and E and/or R2 may comprise a selenocycle.
[0093] In one embodiment, the compound of formula (I) may be selected from a compound of formula (lla), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000029_0001
Formula (lla) wherein R-n R-|2 R13 Ru and R-15 are as previously defined;
A, B, D and E are selected from N and C and at least two of A, B, D, and E are N; each incidence of R6 is independently selected from the group consisting of hydrogen, halide, cyano, C1-C6 alkyl, C1 -C6 alkylamino, C1 -C6 alkylhydroxy, C3-C6 cycloalkyl, alkylphenyl, phenyl, benzyl, C C6 ester, C2-C6 alkenyl, C C6 trifluoroalkyl and C1-C6 alkoxy, each of which may be optionally substituted.
[0094] In one embodiment, the compound of formula (I) may be selected from a compound of formula (lib), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000030_0001
Formula (lib) wherein Y and R5 are as previously defined;
A, B, D and E are selected from N and C and at least two of A, B, D, and E are N;
each incidence of R6 is independently selected from the group consisting of hydrogen, halide, cyano, C C6 alkyl, Ci -C6 alkylamino, Ci -C6 alkylhydroxy, C3-C6 cycloalkyl, alkylphenyl, phenyl, benzyl, C1-C6 ester, C2-C6 alkenyl, C1-C6 trifluoroalkyl and C C6 alkoxy, each of which may be optionally substituted.
[0095] In one embodiment the compound of formula (II), is selected from:
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000032_0002
wherein, R40 is selected from H, alkyl and halo;
R4i is selected from H and alkyl;
each incidence of P is independently selected from C, 0 or S; and wherein each incidence of R6, when present, is independently selected from those groups defined for formula (II).
[0096] It will be understood that the R6 moiety extending from the centre of each ring may represent a group bonded to the ring carbons or ring heteroatoms, as appropriate taking valency into consideration, or may not be present.
[0097] In one embodiment of formula (II), R6 is d-C6 alkyl or C-1 -C6 alkylhydroxy.
[0098] In certain embodiments of the compound of formula (II), for example when R2 is hexahydraindacene and R-i is furan, R6 may not be a tertiary alcohol substituent.
[0099] In one embodiment, the compound of the first aspect may be selected from a compound of formula (Ilia), (lllb) or (lllc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000033_0001
Formula (lllb)
Figure imgf000033_0002
Formula (lllc) wherein, R2i is selected from H, alkyl, perhaloalkyl or hydroxylalkyl; R22 is selected from H, alkyl, perhaloalkyl, C3-C6 cycloalkyl, phenyl or benzyl;
R-I 8 is H or halogen;
R-I6 and Ri7 are H or alkyl; or R-|6 and Ri7, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
R-I 9 and R20 are H or alkyl; or R19 and R20, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
provided that R21 and R22 are not both H; and
provided that R 6, Ri7, R18, R19 and R20 are not all H.
[00100] In a preferred embodiment of compounds of formulae (Ilia), (1Mb) and (IMc):
R21 is selected from H, alkyl, perhaloalkyl or hydroxylalkyl; preferably C1-6 perhaloalkyl or hydroxylalkyl;
R22 is selected from H, alkyl, perhaloalkyl, C3-C6 cycloalkyl, phenyl or benzyl;
R-I6 and R-|7, together with the atoms to which they are attached, form a cyclopentyl ring;
R-19 and R2o, together with the atoms to which they are attached, form a cyclopentyl ring;
R-I 8 is H or halogen, preferably R-|8 is H; and
provided that R2i and R22 are not both H.
[00101 ] In another preferred embodiment of the compounds of formulae (Ilia), (1Mb) and (lllc): R21 is selected from H, alkyl, perhaloalkyi or hydroxylalkyl; preferably C1-6 perhaloalkyi or hydroxylalkyl;
R22 is selected from H, alkyl, perhaloalkyi, C3-C6 cycloalkyi, phenyl and benzyl;
R-I6 and R2o are C1-6 alkyl, preferably isopropyl;
R-17 and R 9 are H,
R-I 8 is H or halogen; preferably R-|8 is H or CI; and
provided that R2i and R22 are not both H.
[00102] In one embodiment, the compound of the first aspect may be selected from a compound of formula (IVa), (IVb) or (IVc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000035_0001
Formula (IVc) wherein, R2i and R22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyl, C3-C6 cycloalkyi, phenyl and benzyl or R21 and R22, together with the carbon atoms to which they are attached, may form a cyclopentyl or a cyclohexyl ring; R-18 is H or halogen;
R-16 and R 7 are H or alkyl; or R 6 and R 7, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
R-I 9 and R20 are H or alkyl; or R 9 and R2o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
provided that R2i and R22 are not both H; and
provided that R-|6, R17, R18, R19 and R20 are not all H.
[00103] In a preferred embodiment of compounds of formulae (IVa), (IVb) and (IVc):
R2 and R22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyi, C3-C6 cycloalkyi, phenyl and benzyl; preferably the perhaloalkyi and hydroxylalkyi are Ci -6 perhaloalkyi and hydroxylalkyi;
R-16 and R-|7, together with the atoms to which they are attached, form a cyclopentyl ring;
R-19 and R20, together with the atoms to which they are attached, form a cyclopentyl ring;
R-18 is H or halogen; preferably R-|8 is H; and
provided that R2 and R22 are not both H.
[00104] In another preferred embodiment of compounds of formulae (IVa), (IVb) and (IVc):
R2i and R22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyi, C3-C6 cycloalkyi, phenyl and benzyl; preferably the perhaloalkyi and hydroxylalkyi are C1 -6 perhaloalkyi and hydroxylalkyi;
R-16 and R20 are Ci-6 alkyl, preferably isopropyl; R-17 and R-|9 are H,
R 8 is H or halogen; preferably R 8 is H or CI;
provided that R2i and R22 are not both H.
[00105] In one embodiment, the compound of the first aspect may be selected from a compound of formula (Va), (Vb) or (Vc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000037_0001
wherein, R2i and R22 are selected from H, alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl;
R-I 8 is H or halogen;
R-I 6 and Ri7 are H or alkyl; or R-|6 and Ri7, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, O and S;
R-19 and R2o are H or alkyl; or R-19 and R2o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
provided that R2i and R22 are not both H; and
provided that R-|6, R17, R18, R19 and R2o are not all H.
[00106] In a preferred embodiment of compounds of formulae (Va), (Vb) and (Vc):
R21 and R22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyi, C3-C6 cycloalkyi, phenyl and benzyl; preferably the perhaloalkyi and hydroxylalkyi are Ci-6 perhaloalkyi and hydroxylalkyi;
R-I 6 and R-17, together with the atoms to which they are attached, form a cyclopentyl ring;
R-19 and R2o, together with the atoms to which they are attached, form a cyclopentyl ring;
R-I 8 is H or halogen; preferably R-|8 is H; and
provided that R2i and R22 are not both H.
[00107] In another preferred embodiment of compounds of formulae (Va), (Vb) and (Vc):
R2i and R22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyi, C3-C6 cycloalkyi, phenyl and benzyl; preferably the perhaloalkyi and hydroxylalkyi are C1-6 perhaloalkyi and hydroxylalkyi;
R-I 6 and R20 are Ci-6 alkyl, preferably isopropyl;
Figure imgf000038_0001
R-I 8 is H or halogen; preferably R 8 is H or CI; and
provided that R2i and R22 are not both H.
[00108] In one embodiment, the compound of the first aspect may be selected from a compound of formula (Via) or (VIb), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000039_0001
Formula (Via) Formula (Vlb) wherein, R22 is selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl;
R-18 is H or halogen;
R-16 and R 7 are H or alkyl; or R 6 and R 7, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
R-19 and R20 are H or alkyl; or R 9 and R2o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S; and
provided that R 6, Ri7, R18, R19 and R20 are not all H.
[00109] In a preferred embodiment of compounds of formulae (Via) and (Vlb):
R22 is selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl; preferably the perhaloalkyl and hydroxylalkyl are Ci-6 perhaloalkyl and hydroxylalkyl;
R 6 and R 7, together with the atoms to which they are attached, form a cyclopentyl ring;
R-19 and R2o, together with the atoms to which they are attached, form a cyclopentyl ring; and R-18 is H or halogen; preferably R-|8 is H.
[00110] In another preferred embodiment of compounds of formulae (Via) and (Vlb):
R22 is selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl; preferably the perhaloalkyl and hydroxylalkyl are C1-6 perhaloalkyl and hydroxylalkyl;
R-I 6 and R2o are C1-6 alkyl, preferably isopropyl;
R-17 and R 9 are H; and
R-18 is H or halogen; preferably R-|8 is H or CI.
[00111] In one embodiment, the compound of the first aspect may be selected from a compound of formula (VII), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000040_0001
Formula (VII) wherein, Q is O or S; each incidence of R30 is independently selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, and alkylam ino;
R-18 is H or halogen;
R-I 6 and R 7 are H or alkyl; or R 6 and R 7, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
R-19 and R20 are H or alkyl; or R 9 and R2o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
provided that R 6, R-17, R18, R19 and R20 are not all H; and provided that when Q is 0 and R-|6 and R-1 7, and separately R-19 and R2o, together with the respective carbon atoms to which they are attached, form a cyclopentyl ring then R30 is not C-3 hydroxylalkyl.
[001 12] In a preferred embodiment of compounds of formulae (VII):
Q is 0 or S; each incidence of R30 is independently selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, and alkylamino; preferably C1-6 alkyl, perhaloalkyl, hydroxylalkyl, and alkylamino;
R-I 6 and R-17, together with the atoms to which they are attached, form a cyclopentyl ring;
R-19 and R20, together with the atoms to which they are attached, form a cyclopentyl ring; and
R-I 8 is H or halogen; preferably R-|8 is H and provided that when Q is 0 then R30 is not C-3 hydroxylalkyl.
[001 13] In another preferred embodiment of compounds of formulae (VII):
Q is O or S; each incidence of R30 is independently selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, and alkylamino; preferably Ci-6 alkyl, perhaloalkyl, hydroxylalkyl, and alkylamino;
R-I 6 and R20 are Ci-6 alkyl, preferably isopropyl;
R-17 and R-19 are H; and R-18 is H or halogen; preferably R-|8 is H or CI.
[001 14] The compounds of the first aspect, and particularly those of formulae (II) to (VI), provide a range of unexpected benefits over those sulfonylureas of the prior art, which benefits may be selected from: Improved microsomal stability; Improved permeability; Reduced Pgp liability; Reduced plasma protein binding; Increased half-life; Improved oral bioavailability; Improved AUC; Improved Cmax; Reduced Cyp inhibition; Improved inhibition of activation of the NLRP3 inflammasome; and Improved solubility. The solubility, and certain other, improvements may be seen particularly in an aqueous environment.
[001 15] In one embodiment, the compounds of the first aspect offer improved pharmacokinetic characteristics. CRID3, a known sulfonylurea, has a half life of 3.2 hours (mouse) which may lead to substantial trough levels from QD or BD dosing when the t1/2 is extrapolated to man. The compounds of the first aspect may differ in, for example, their protein binding, metabolism and oral availability.
[001 16] Particularly it has been found that compounds of the first aspect, especially those wherein A, B, D and E form a 5-membered nitrogen heteroaryl, for example a pyrazole ring, are less metabolically labile and/or have improved pharmacokinetic properties over otherwise structurally similar furans and thiophenes seen in the prior art.
[001 17] In one embodiment, the compounds of of the first aspect have a tPSA of less than 90 A2.
[001 18] It is one advantage of the present compounds of the first aspect that they may demonstrate a significantly lowered polar surface area in comparison to prior art sulfonylureas, such as CRID3.
[001 19] In one further embodiment, the compounds of the first aspect have a tPSA of less than 90 A2 and a molecular weight of less than 405. [00120] The absence of a tertiary alcohol group, in some embodiments, increases plasma concentration and aids in decreasing both MW and polar surface area thereby giving an overall improvement in blood brain barrier penetration.
[00121 ] In any of the embodiments described for the compound of the first aspect, including the compounds of formula (I) to (VII), one or more hydrogens of the substituents or optional substitutions thereupon may be deuterated.
[00122] Deuterated analogues of the compounds of the invention may exhibit increased metabolic stability due to the kinetic isotope effect.
[00123] In one embodiment, the compound of the first aspect is selected from the group consisting of:
Figure imgf000043_0001
Figure imgf000044_0001
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0001
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
[00124] In certain embodiments, the compounds of the first aspect may exhibit improved properties compared to known anti-diabetes drugs. Examples of such compounds may include those below:
Figure imgf000059_0002
Figure imgf000059_0003
Figure imgf000060_0001
[00125] These four compounds of formula (I) may be viewed as very potent versions of current sulfonylurea anti-diabetes drugs. The IC50 data presented in the experimental section reflects this view. It is believed that known drugs do not target NLRP3 to any therapeutically significant extent and so it would be necessary to use very high doses to have any significant effect on the NLRP3 inflammasome. The four compounds shown above, and others of the first aspect, show advantageously improved properties in a significant decrease in IC50 versus the NLRP3 inflammasome and additionally have the benefits, not realised by existing diabetes and other drugs, associated with NLRP3 inhibition such as improved wound healing and other advantages described herein.
[00126] In any one or more embodiments of the first aspect and in relation to any one or more of the compounds of formula (I) to (VII), the compound is an inhibitor of activation of the NLRP3 inflammasome.
[00127] Therefore it will be appreciated that the present invention provides for sulfonyl urea and related drugs exhibiting significantly lower NLRP3 IC50 values in cell based assay using HMDM (see experimental section for protocols) than the above comparator compounds. Currently known diabetes drugs are not potent inhibitors of the NLRP3 inflammasome at therapeutic doses and to achieve any such inhibition would require dosing outside of recommended levels. The present compounds allow lower doses to be used and therefore limit the risk of toxic effects.
[00128] In a further embodiment, one or more of the compounds of the first aspect may be useful as photoswitchable compounds which may be applied in a range of uses including but not limited to insulin release. Such compounds may, in one embodiment, be selected from the group consisting of:
Figure imgf000061_0001
wherein, R2 is as defined in any one or more of the embodiments of compounds of formula (I) to (VII) described previously.
[00129] In certain embodiments of the invention one or more compounds of the first aspect may be appropriate for use as probes, such as photoaffinity probes, or as reactive intermediates which can be modified either directly or by means of a linking moiety to give biotinylated, fluorescent or photoaffinity probes including, but not limited to, those shown below:
Figure imgf000062_0001
Figure imgf000063_0001
wherein, R2 is as defined in any one or more of the embodiments described for formula (I) to (VII).
[00130] Particularly, such compounds as probes or reactive intermediates may be selected from those below:
Figure imgf000064_0001
[00131 ] It will be appreciated that the compounds of the first aspect may be modified or derivatised by means well understood in the art to allow linkage to a molecule such as biotin, or a fluorescent group or photoaffinity label, as shown with certain of the compounds above.
[00132] In one embodiment, the compound of formula (I) or (II) does not comprise a structure selected from the groups below shown attached to the sulfonyl moiety (i.e. as an Ri group):
Figure imgf000065_0001
[00133] In one embodiment, wherein the compound of the first aspect, including any compound of formula (I) to (VII), has J as S, W as O and R2 is selected from hexahydroindacene, 2,6-diisopropylphenyl and 2,6-diisopropyl-4- chlorophenyl then Ri is not one of 2,4-disubstituted furan, 2,4-disubstituted thiophene, 2,5-disubstituted furan and 2,5-disubstituted thiophene.
[00134] In one embodiment, wherein the compound of the first aspect, including any compound of formula (I) to (VII), has J as S, W as O and R-i is selected from substituted triazole, thiadiazole, 4-substituted pyridine and 1 ,2- disubstituted imidazole then R2 is not unsubstituted phenyl, 2- or 4-chlorophenyl or 3,4- substituted phenyl, substituted with one or more of halo, trifluoromethyl, nitro or thiomethyl.
[00135] In one embodiment, wherein the compound of the first aspect, including any compound of formula (I) to (VII), has J as S, W as 0 and R-i is selected from substituted triazole, thiadiazole, benzothiazole and substituted pyrimidine then R2 is not thiophene, 3-chlorophenyl, 4-ethoxyphenyl, substituted benzimidazole or substituted benzothiazole.
[00136] In one embodiment, wherein the compound of the first aspect, including any compound of formula (I) to (VII), has J as S, W as O and Ri is ethoxy substituted benzothiazole, then R2 is not 2,6-diisopropylphenyl.
[00137] In one embodiment, wherein the compound of the first aspect, including any compound of formula (I) to (VII), has J as S, W as O and R-i is selected from benzofuran, benzothiophene and indole then R2 is not 3- or 3,4- halo, methyl, ethyl or trifluoromethyl substituted phenyl.
[00138] In one embodiment, wherein the compound of the first aspect, including any compound of formula (I) to (VII), has J as S, W as O and R2 is substituted pyrimidine, then Ri is not pyrazole substituted with ester or carboxy.
[00139] In one embodiment, wherein the compound of the first aspect, including any compound of formula (I) to (VII), has J as S and W as O then the carbon atom of R2 which is directly bonded to the urea nitrogen is not a carbonyl carbon.
[00140] In one embodiment, wherein the compound of the first aspect, including any compound of formula (I) to (VII), has J as S and W as O, then the carbon atom of R2 which is directly bonded to the urea nitrogen is an aryl, heteroaryl or heterocyclic ring carbon.
[00141 ] In one embodiment, wherein the compound of the first aspect, including any compound of formula (I) to (VII), has J as S and W as O, R2 is a substituted phenyl and Ri is a pyrazole then the Ri pyrazole is not substituted with an aryl or heteroaryl group.
[00142] In one embodiment, wherein the compound of the first aspect, including any compound of formula (I) to (VII), has J as S, W as 0, and R-i is a pyrazole and the sulfonylurea linker is branched in position 4 thereof, the pyrazole is not fused in positions 1 and 5 with a 6-membered heterocycle to form a pyrazolopyrimidine derivative.
[00143] In one embodiment, the compound of the first aspect, including any compound of formula (I) to (VII) is not a compound selected from the group consisting of:
1 . 1 -(4-Chloro-2,6-diisopropyl-phenyl)-3-[3-(1 -hydroxy-1 -methyl-ethyl)- benzenesulfonyl]-urea;
2. 1 -(1 ,2,3,5,6,7-Hexahydro-s-indacen-4-yl)-3-[4-(1 -hydroxy-1 -methyl- ethyl)-furan-2-sulfonyl]-urea;
3. 1 -(1 ,2,3,5,6,7-Hexahydro-4-aza-s-indacen-8-yl)-3-[4-(1 -hydroxy-1 - methyl-ethyl)-furan-2-sulfonyl]-urea;
4. 1 -(1 ,2,3,5,6,7-Hexahydro-s-indacen-4-yl)-3-[4-(1 -hydroxy-1 -methyl- ethyl)-thiophene-2-sulfonyl]-urea;
5. 1 -(4-[1 ,3]Dioxolan-2-yl-furan-2-sulfonyl)-3-(1 ,2,3,5,6,7-hexahydro-s- indacen-4-yl)-urea;
6. 1 -(2,6-Diisopropyl-phenyl)-3-[4-(1 -hydroxy-1 -methyl-ethyl)-furan-2- sulfonyl]-urea;
7. 1 -(2,6-Diisopropyl-phenyl)-3-[4-(1 -hydroxy-1 -methyl-ethyl)-thiophene-2- sulfonyl]-urea;
8. 1 -(4-Acetyl-thiophene-2-sulfonyl)-3-(1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)-urea;
9. 1 -(1 H-Benzoimidazole-5-sulfonyl)-3-(1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)-urea; 10. 1 -(1 ,2,3,5,6,7-Hexahydro-s-indacen-4-yl)-3-[4-(1 -hydroxy-1 -methyl- ethyl)-thiophene-2-sulfonyl]-urea;
1 1. 1 -(8-Chloro-1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)-3-[4-(1 -hydroxy-1 - methyl-ethyl)-furan-2-sulfonyl]-urea;
12. 1 -(4-Acetyl-furan-2-sulfonyl)-3-(1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)- urea;
13. 1 -(8-Fluoro-1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)-3-[4-(1 -hydroxy-1 - methyl-ethyl)-furan-2-sulfonylj-urea;
14. 1 -(4-Fluoro-2,6-diisopropyl-phenyl)-3-[3-(1 -hydroxy-1 -methyl-ethyl)- benzenesulfonyl]-urea; and
15. 1 -(6-Fluoro-1 H-benzoimidazole-5-sulfonyl)-3-(1 ,2,3,5,6,7-hexahydro-s- indacen-4-yl)-urea;
16. 1 -(4-Chloro-2,6-diisopropyl-phenyl)-3-(1 H-indole-6-sulfonyl)-urea; 17. 1 -(4-Chloro-2,6-diisopropyl-phenyl)-3-(5-fluoro-1 H-indole-6-sulfonyl)- urea;
18. 1 -[1 ,2,3,5,6,7-Hexahydro-s-indacen-u-yl)-3-(1 H-indole-6-sulfonyl)-urea; 19. 1 -(5-Fluoro-1 H-indole-6-sulfonyl)-3-(1 ,2,3,5,6,7-hexanhydro-5-indacen-4- yl)-urea;
20. 1 -[4-Chloro-2,6-diisopropyl-phenyl]-3-[2-fluoro-5-(2-methyl-(1 ,3)dioxolan-
2-yl)-benzenesulfonyl]-urea;
21. 3-[3-[4-Chloro-2,6-diisopropyl-phenyl]-ureidosulfonyl]-N-methyl- benzenesulfonam ide;
22. 1 -[2-Fluoro-5-(2-methyl-(1 ,3)dioxolan-2-yl)benzenesulfonyl]-3-
1 ,2,3,5,6,7-hexahydro-indacen-4-yl)-urea;
23. 3- [3-(1 ,2,3,5,6,7-Hexahydro-S-indacen-4-yl)-ureidosulfonyl]-N-methyl- benzenesulfonam ide;
24.4- (1 -hydroxy-1 -methyl-ethyl)-furan-2-sulfonam ide.
[00144] In some embodiments of the present invention, therapeutically inactive prodrugs of the compounds of the first aspect are provided. Prodrugs are compounds which, when administered to a mammal, are converted in whole or in part to a compound of the invention. In most embodiments, the prodrugs are pharmacologically inert chemical derivatives that can be converted in vivo to the active drug molecules to exert a therapeutic effect. Any of the compounds described herein can be administered as a prodrug to increase the activity, bioavailability, or stability of the compound or to otherwise alter the properties of the compound. Typical examples of prodrugs include compounds that have biologically labile protecting groups on a functional moiety of the active compound. Prodrugs include, but are not limited to, compounds that can be oxidized, reduced, aminated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, dehydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated, and/or dephosphorylated to produce the active compound.
[00145] A number of prodrug ligands are known. In general, alkylation, acylation, or other lipophilic modification of one or more heteroatoms of the compound, such as a free amine or carboxylic acid residue, may reduce polarity and allow for the compound's passage into cells. Examples of substituent groups that can replace one or more hydrogen atoms on a free amine and/or carboxylic acid moiety include, but are not limited to, the following: aryl; steroids; carbohydrates (including sugars); 1 ,2-diacylglycerol; alcohols; acyl (including lower acyl); alkyl (including lower alkyl); sulfonate ester (including alkyl or arylalkyl sulfonyl, such as methanesulfonyl and benzyl, wherein the phenyl group is optionally substituted with one or more substituents as provided in the definition of an aryl given herein); optionally substituted arylsulfonyl; lipids (including phospholipids); phosphotidylcholine; phosphocholine; amino acid residues or derivatives; amino acid acyl residues or derivatives; peptides; cholesterols; or other pharmaceutically acceptable leaving groups which, when administered in vivo, provide the free amine. Any of these moieties can be used in combination with the disclosed active agents to achieve a desired effect. [00146] In some embodiments, compounds with one or more chiral centers are provided. While racemic mixtures of compounds of the invention may be active, selective, and bioavailable, isolated isomers may be of interest as well.
[00147] The compounds of the first aspect may contain chiral centers, which may be either of the (R) or (S) configuration, or which may comprise a mixture thereof. Accordingly, the present invention also includes stereoisomers of the compounds described herein, where applicable, either individually or admixed in any proportions. Stereoisomers may include, but are not limited to, enantiomers, diastereomers, racemic mixtures, and combinations thereof. Such stereoisomers can be prepared and separated using conventional techniques, either by reacting enantiomeric starting materials, or by separating isomers of compounds and prodrugs of the present invention. Isomers may include geometric isomers. Examples of geometric isomers include, but are not limited to, cis isomers or trans isomers across a double bond. Other isomers are contemplated among the compounds of the present invention. The isomers may be used either in pure form or in admixture with other isomers of the compounds described herein.
[00148] Various methods are known in the art for preparing optically active forms and determining activity. Such methods include standard tests described herein and other similar tests which are well known in the art. Examples of methods that can be used to obtain optical isomers of the compounds according to the present invention include the following: i) physical separation of crystals whereby macroscopic crystals of the individual enantiomers are manually separated. This technique may particularly be used when crystals of the separate enantiomers exist (i.e., the material is a conglomerate), and the crystals are visually distinct;
ii) simultaneous crystallization whereby the individual enantiomers are separately crystallized from a solution of the racemate, possible only if the latter is a conglomerate in the solid state;
iii) enzymatic resolutions whereby partial or complete separation of a racemate by virtue of differing rates of reaction for the enantiomers with an enzyme;
iv) enzymatic asymmetric synthesis, a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer;
v) chemical asymmetric synthesis whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce asymmetry (i.e., chirality) in the product, which may be achieved using chiral catalysts or chiral auxiliaries;
vi) diastereomer separations whereby a racemic compound is reacted with an enantiomerically pure reagent (the chiral auxiliary) that converts the individual enantiomers to diastereomers. The resulting diastereomers are then separated by chromatography or crystallization by virtue of their now more distinct structural differences and the chiral auxiliary later removed to obtain the desired enantiomer;
vii) first- and second-order asymmetric transformations whereby diastereomers from the racemate equilibrate to yield a preponderance in solution of the diastereomerfrom the desired enantiomer or where preferential crystallization of the diastereomer from the desired enantiomer perturbs the equilibrium such that eventually in principle all the material is converted to the crystalline diastereomer from the desired enantiomer. The desired enantiomer is then released from the diastereomers;
viii) kinetic resolutions comprising partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions; ix) enantiospecific synthesis from non-racemic precursors whereby the desired enantiomer is obtained from non-chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis;
x) chiral liquid chromatography whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase. The stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions;
xi) chiral gas chromatography whereby the racemate is volatilized and enantiomers are separated by virtue of their differing interactions in the gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase;
xii) extraction with chiral solvents whereby the enantiomers are separated by virtue of preferential dissolution of one enantiomer into a particular chiral solvent; and
xiii) transport across chiral membranes whereby a racemate is placed in contact with a thin membrane barrier. The barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane barrier. Separation occurs as a result of the non-racemic chiral nature of the membrane which allows only one enantiomer of the racemate to pass through.
[00149] The compound optionally may be provided in a composition that is enantiomerically enriched, such as a mixture of enantiomers in which one enantiomer is present in excess, in particular, to the extent of 95% or more, 96% or more, 97% or more, 98% or more, or 99% or more, including 100%. [00150] The terms (R), (S), (R,R), (S,S), (R,S) and (S,R) as used herein mean that the composition contains a greater proportion of the named isomer of the compound in relation to other isomers. In a preferred embodiment, these terms indicate that the composition contains at least 90% by weight of the named isomer and 10% by weight or less of the one or more other isomers; or more preferably about 95% by weight of the named isomer and 5% or less of the one or more other isomers. In some embodiments, the composition may contain at least 99% by weight of the named isomer and 1 % or less by weight of the one or more other isomers, or may contain 100% by weight of the named isomer and 0% by weight of the one of more other isomers. These percentages are based on the total amount of the compound of the present invention present in the composition.
[00151 ] The compounds of the first aspect may be utilized per se or in the form of a pharmaceutically acceptable ester, amide, salt, solvate, prodrug, or isomer, as appropriate. For example, the compound may be provided as a pharmaceutically acceptable salt. If used, a salt of the drug compound should be both pharmacologically and pharmaceutically acceptable, but non- pharmaceutically acceptable salts may conveniently be used to prepare the free active compound or pharmaceutically acceptable salts thereof and are not excluded from the scope of this invention. Such pharmacologically and pharmaceutically acceptable salts can be prepared by reaction of the drug with an organic or inorganic acid, using standard methods detailed in the literature.
[00152] Examples of pharmaceutically acceptable salts of the compounds useful according to the invention include acid addition salts. Salts of non- pharmaceutically acceptable acids, however, may be useful, for example, in the preparation and purification of the compounds. Suitable acid addition salts according to the present invention include organic and inorganic acids. Preferred salts include those formed from hydrochloric, hydrobromic, sulfuric, phosphoric, citric, tartaric, lactic, pyruvic, acetic, succinic, fumaric, maleic, oxaloacetic, methanesulfonic, ethanesulfonic, p-toluenesulfonic, benzenesulfonic, and isethionic acids. Other useful acid addition salts include propionic acid, glycolic acid, oxalic acid, malic acid, malonic acid, benzoic acid, cinnamic acid, mandelic acid, salicylic acid, and the like. Particular example of pharmaceutically acceptable salts include, but are not limited to, sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1 ,4-dioates, hexyne-1 ,6-dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxyenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, γ- hydroxybutyrates, glycolates, tartrates, methanesulfonates, propanesulfonates, naphthalene-1 -sulfonates, naphthalene-2-sulfonates, and mandelates.
[00153] An acid addition salt may be reconverted to the free base by treatment with a suitable base. Preparation of basic salts of acid moieties which may be present on a compound or prodrug useful according to the present invention may be prepared in a similar manner using a pharmaceutically acceptable base, such as sodium hydroxide, potassium hydroxide, ammonium hydroxide, calcium hydroxide, triethylamine, or the like.
[00154] Esters of the active agent compounds according to the present invention may be prepared through functionalization of hydroxyl and/or carboxyl groups that may be present within the molecular structure of the compound. Amides and prodrugs may also be prepared using techniques known to those skilled in the art. For example, amides may be prepared from esters, using suitable amine reactants, or they may be prepared from an anhydride or an acid chloride by reaction with ammonia or a lower alkyl amine. Moreover, esters and amides of compounds of the invention can be made by reaction with a carbonylating agent {e.g., ethyl formate, acetic anhydride, methoxyacetyl chloride, benzoyl chloride, methyl isocyanate, ethyl chloroformate, methanesulfonyl chloride) and a suitable base {e.g., 4-dimethylaminopyridine, pyridine, triethylamine, potassium carbonate) in a suitable organic solvent (e.g., tetrahydrofuran, acetone, methanol, pyridine, Ν,Ν-dimethylformamide) at a temperature of 0 °C to 60 °C. Prodrugs are typically prepared by covalent attachment of a moiety, which results in a compound that is therapeutically inactive until modified by an individual's metabolic system. Examples of pharmaceutically acceptable solvates include, but are not limited to, compounds according to the invention in combination with water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, or ethanolamine.
[00155] In the case of solid compositions, it is understood that the compounds used in the methods of the invention may exist in different forms. For example, the compounds may exist in stable and metastable crystalline forms and isotropic and amorphous forms, all of which are intended to be within the scope of the present invention.
[00156] If a compound useful as an active agent according to the invention is a base, the desired salt may be prepared by any suitable method known to the art, including treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, pyranosidyl acids such as glucuronic acid and galacturonic acid, alpha-hydroxy acids such as citric acid and tartaric acid, amino acids such as aspartic acid and glutamic acid, aromatic acids such as benzoic acid and cinnamic acid, sulfonic acids such a p-toluenesulfonic acid or ethanesulfonic acid, or the like.
[00157] If a compound described herein as an active agent is an acid, the desired salt may be prepared by any suitable method known in the art, including treatment of the free acid with an inorganic or organic base, such as an am ine (primary, secondary or tertiary), an alkali metal or alkaline earth metal hydroxide or the like. Illustrative examples of suitable salts include organic salts derived from amino acids such as glycine and arginine, ammonia, primary, secondary and tertiary amines, and cyclic amines such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium , calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum and lithium.
[00158] According to a second aspect of the invention there is provided a pharmaceutical composition comprising a compound of formula (I) to (VII), or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier, diluent and/or excipient.
[00159] Suitably, the pharmaceutically acceptable carrier, diluent and/or excipient may be or include one or more of diluents, solvents, pH buffers, binders, fillers, emulsifiers, disintegrants, polymers, lubricants, oils, fats, waxes, coatings, viscosity-modifying agents, glidants and the like.
[00160] The salt forms of the compounds of the invention may be especially useful due to their improved solubility.
[00161 ] In one embodiment, the pharmaceutical composition includes a cyclodextrin.
[00162] The cyclodextrin may be selected from alpha, beta or gamma cyclodextrins.
[00163] In one embodiment, the cyclodextrin is selected from a methyl cyclodextrin, a hydroxypropyl cyclodextrin and a sulfobutylether cyclodextrin.
[00164] It has been found that cyclodextrins provide significant advantages in formulation and delivery of the compounds of the invention.
[00165] Cyclodextrin formulations such as for example, one or more compounds of the invention with hydroxypropyl beta cyclodextrin or methyl beta cyclodextrin, may have uses in cholesterol sequestration/cholesterol lowering or via NLRP3 inhibition for Non-alcoholic steatohepatitis (NASH), alcoholic liver disease, atherosclerosis and also in Alzheimer's Disease (AD).
[00166] Diluents may include one or more of microcrystalline cellulose, lactose, mannitoi, calcium phosphate, calcium sulfate, kaolin, dry starch, powdered sugar, and the like. Binders may include one or more of povidone, starch, stearic acid, gums, hydroxypropylmethyl cellulose and the like. Disintegrants may include one or more of starch, croscarmellose sodium, crospovidone, sodium starch glycolate and the like. Solvents may include one or more of ethanoi, methanol, isopropanoi, chloroform, acetone, methylethyl ketone, methylene chloride, water and the like. Lubricants may include one or more of magnesium stearate, zinc stearate, calcium stearate, stearic acid, sodium stearyl fumarate, hydrogenated vegetable oil, glyceryl behenate and the like. A glidant may be one or more of colloidal silicon dioxide, talc or cornstarch and the like. Buffers may include phosphate buffers, borate buffers and carbonate buffers, although without limitation thereto. Fillers may include one or more gels inclusive of gelatin, starch and synthetic polymer gels, although without limitation thereto. Coatings may comprise one or more of film formers, solvents, plasticizers and the like. Suitable film formers may be one or more of hydroxypropyl methyl cellulose, methyl hydroxyethyl cellulose, ethyl cellulose, hydroxypropyl cellulose, povidone, sodium carboxymethyl cellulose, polyethylene glycol, acrylates and the like. Suitable solvents may be one or more of water, ethanoi, methanol, isopropanoi, chloroform, acetone, methylethyl ketone, methylene chloride and the like. Plasticizers may be one or more of propylene glycol, castor oil, glycerin, polyethylene glycol, poiysorbates, and the like. [00167] Reference is made to the Handbook of Excipients 6 Edition, Eds. Rowe, Sheskey & Quinn (Pharmaceutical Press), which provides non-limiting examples of excipients which may be useful according to the invention.
[00168] It will be appreciated that the choice of pharmaceutically acceptable carriers, diluents and/or excipients will, at least in part, be dependent upon the mode of administration of the formulation. By way of example only, the composition may be in the form of a tablet, capsule, caplet, powder, an injectable liquid, a suppository, a slow release formulation, an osmotic pump formulation or any other form that is effective and safe for administration.
[00169] Suitably, the pharmaceutical composition is for the treatment or prevention of a disease, disorder or condition in a mammal.
[00170] A third aspect of the invention resides in a method of treatment or prevention of a disease, disorder or condition including the step of administering an effective amount of a compound of formula (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof, or the pharmaceutical composition of the second aspect to thereby treat or prevent the disease disorder or condition.
[00171 ] A fourth aspect of the invention provides for a compound of formula (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof, or the pharmaceutical composition of the second aspect for use in the treatment or prevention of a disease, disorder or condition.
[00172] A fifth aspect of the invention provides for use of a compound of formula (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof, in the manufacture of a medicament for the treatment or prevention of a disease, disorder or condition.
[00173] As generally used herein, the terms "administering" or "administration", and the like, describe the introduction of the compound or composition to a mammal such as by a particular route or vehicle. Routes of administration may include topical, parenteral and enteral which include oral, buccal, sub-lingual, nasal, anal, gastrointestinal, subcutaneous, intramuscular and intradermal routes of administration, although without limitation thereto.
[00174] By "treat", "treatment" or treating" is meant administration of the compound or composition to a subject to at least ameliorate, reduce or suppress existing signs or symptoms of the disease, disorder or condition experienced by the subject.
[00175] By "prevent", "preventing" or "preventative" is meant prophylactically administering the formulation to a subject who does not exhibit signs or symptoms of a disease disorder or condition, but who is expected or anticipated to likely exhibit such signs or symptoms in the absence of prevention. Preventative treatment may at least lessen or partly ameliorate expected symptoms or signs.
[00176] As used herein, ' effective amount" refers to the administration of an amount of the relevant compound or composition sufficient to prevent the occurrence of symptoms of the condition being treated, or to bring about a halt in the worsening of symptoms or to treat and alleviate or at least reduce the severity of the symptoms. The effective amount will vary in a manner which would be understood by a person of skill in the art with patient age, sex, weight etc. An appropriate dosage or dosage regime can be ascertained through routine trial.
[00177] As used herein, the terms "subject" or "individual" or "patient" may refer to any subject, particularly a vertebrate subject, and even more particularly a mammalian subject, for whom therapy is desired. Suitable vertebrate animals include, but are not restricted to, primates, avians, livestock animals (e.g., sheep, cows, horses, donkeys, pigs), laboratory test animals (e.g., rabbits, mice, rats, guinea pigs, hamsters), companion animals (e.g., cats, dogs) and captive wild animals (e.g., foxes, deer, dingoes). A preferred subject is a human in need of treatment for a disease, disorder or condition as described herein. However, it will be understood that the aforementioned terms do not imply that symptoms are necessarily present.
[00178] In one particular embodiment, the disease, disorder or condition is one which is responsive to inhibition of activation of the NLRP3 inflammasome.
[00179] According to this embodiment, the compound of the first aspect, or pharmaceutically effective salt, solvate or prodrug thereof is a specific inhibitor of NLRP3.
[00180] In a further embodiment, the disease, disorder or condition is responsive to modulation of one or more of IL-Ι β, IL-17, IL-18, IL-1 , IL-37, IL- 33 and Th17 cells.
[00181 ] In one embodiment, the modulation is inhibition of one or more of IL- 1 β, IL-17, IL-18, IL-1 a, IL-37, and IL-33.
[00130] In one embodiment, the modulation of Th17 cells, is by inhibition of production and/or secretion of IL-17.
[00182] In general embodiments, the disease, disorder or condition is a disease, disorder or condition of the immune system, the cardiovascular system, the endocrine system, the gastrintestinal tract, the renal system, the respiratory system, the central nervous system, is a cancer or other malignancy and/or is caused by or associated with a pathogen.
[00183] It will be appreciated that these general embodiments defined according to broad categories of diseases, disorders and conditions are not mutually exclusive. In this regard any particular disease, disorder or condition may be categorized according to more than one of the above general embodiments. A non-limiting example is Type I diabetes which is an autoimmune disease and a disease of the endocrine system. [00184] In one embodiment, the disease, disorder or condition is of the immune system. In particular embodiments, the disease disorder or condition is an inflammatory disease disorder or condition or an autoimmune disease disorder or condition.
[00185] In one embodiment, the disease, disorder or condition is of the skin.
[00186] In one embodiment, the disease, disorder or condition is of the cardiovascular system.
[00187] In one embodiment, the disease, disorder or condition is a cancer, tumour or other malignancy. As used herein, cancers tumours and malignancies, refer to diseases disorders or conditions, or to cells or tissues associated with the diseases, disorders or conditions, characterized by aberrant or abnormal cell proliferation, differentiation and/or migration often accompanied by an aberrant or abnormal molecular phenotype that includes one or more genetic mutations or other genetic changes associated with oncogenesis, expression of tumour markers, loss of tumour suppressor expression or activity and/or aberrant or abnormal cell surface marker expression. In general embodiments, cancers, tumours and malignancies may include sarcomas, lymphomas, leukemias, solid tumours, blastomas, gliomas, carcinomas, melanomas and metastatic cancers, although without limitation thereto. A more comprehensive listing of cancers tumours and malignancies may be found at the National Cancer Institutes website http://www.cancer.gov/cancertopics/types/alphalist.
[00188] In one embodiment, the disease, disorder or condition is of the renal system .
[00189] In one embodiment, the disease, disorder or condition is of the gastro-intestinal tract. [00190] In one embodiment, the disease, disorder or condition is of the respiratory system.
[00191 ] In a further embodiment, the disease, disorder or condition is of the endocrine system.
[00192] In one embodiment, the disease, disorder or condition is of the central nervous system (CNS).
[00193] In one embodiment, the disease, disorder or condition is caused by, or is associated with, a pathogen. The pathogen may be a virus, a bacterium, a protist, a worm or a fungus or any other organism capable of infecting a mammal, although without limitation thereto.
[00194] Non-limiting examples of viruses include influenza virus, cytomegalovirus, Epstein Barr Virus, human immunodeficiency virus (HIV), alphavirus such as Chikungunya and Ross River virus, flaviviruses such as Dengue virus, Zika virus and papillomavirus, although without limitation thereto.
[00195] Non-limiting examples of pathogenic bacteria include
Staphylococcus aureus, Helicobacter pylori, Bacillus anthracis, Bordatella pertussis, Corynebacterium diptheriae, Clostridium tetani, Clostridium botulinum, Streptococcus pneumoniae, Streptococcus pyogenes, Listeria monocytogenes, Hemophilus influenzae, Pasteureiia multicida, Shigella dysenteriae, Mycobacterium tuberculosis, Mycobacterium leprae, Mycoplasma pneumoniae, Mycoplasma hominis, Neisseria meningitidis, Neisseria gonorrhoeae, Rickettsia rickettsii, Legionella pneumophila, Klebsiella pneumoniae, Pseudomonas aeruginosa, Propionibacterium acnes, Treponema pallidum, Chlamydia trachomatis, Vibrio cholerae, Salmonella typhimurium, Salmonella typhi, Borrelia burgdorferi and Yersinia pestis, although without limitation thereto. [00196] Non-limiting examples of protists include Plasmodium, Babesia, Giardia, Entamoeba, Leishmania and Trypanosomes, although without limitation thereto.
[00197] Non-limiting examples of worms include helminths inclusive of schistisimes, roundworms, tapeworms and flukes, although without limitation thereto.
[00198] Non-limiting examples of fungi include Candida and Aspergillus species, although without limitation thereto.
[00199] Further relevant disease, disorder or conditions may be selected from the group consisting of those recited in the journal article found at: http://onlinelibrary.wiley.eom/store/10.1 1 1 1/j.1365- 2249.201 1.04440. x/asset/j.1365-
2249.201 1.04440.x.pdf?v=1&t=i60c1 phf&s=d26f50a2622926cc6b4bc855bd91 1 ae9dc9750cf.
[00200] In particular embodiments, the disease, disorder or condition is selected from the group consisting of constitutive inflammation including the cryopyrin-associated periodic syndromes (CAPS): Muckle-Wells syndrome (MWS), familial cold autoinflammatory syndrome (FCAS) and neonatal-onset multisystem inflammatory disease (NOMID); including autoinflammatory diseases: familial Mediterranean fever (FMF), TNF receptor associated periodic syndrome (TRAPS), mevalonate kinase deficiency (MKD), hyperimmunoglobulinemia D and periodic fever syndrome (H IDS), deficiency of interleukin 1 receptor (DIRA) antagonist, Majeed syndrome, pyogenic arthritis, pyoderma gangrenosum and acne (PAPA), haploinsufficiency of A20 (HA20), pediatric granulomatous arthritis (PGA), PLCG2 -associated antibody deficiency and immune dysregulation (PLAID), PLCG2-associated autoinflammation, antibody deficiency and immune dysregulation (APLAID), sideroblastic anemia with B-cell immunodeficiency, periodic fevers, and developmental delay (SIFD); Sweet s syndrome, chronic nonbacterial osteomyelitis (CNO), chronic recurrent multifocal osteomyelitis (CRMO) and synovitis, acne, pustulosis, hyperostosis, osteitis syndrome (SAPHO); autoimmune diseases including multiple sclerosis (MS), type-1 diabetes, psoriasis, rheumatoid arthritis, Behcet's disease, Sjogren's syndrome and Schnitzler syndrome; respiratory diseases including chronic obstructive pulmonary disorder (COPD), steroid-resistant asthma, asbestosis, silicosis and cystic fibrosis; central nervous system diseases including Parkinson's disease, Alzheimer's disease, motor neuron disease, Huntington's disease, cerebral malaria and brain injury from pneumococcal meningitis; metabolic diseases including Type 2 diabetes, atherosclerosis, obesity, gout, pseudo-gout; ocular diseases including those of the ocular epithelium, age-related macular degeneration (AMD), corneal infection, uveitis and dry eye; kidney disease including chronic kidney disease, oxalate nephropathy and diabetic nephropathy; liver disease including non-alcoholic steatohepatitis and alcoholic liver disease; inflammatory reactions in skin including contact hypersensitivity and sunburn; inflammatory reactions in the joints including osteoarthritis, systemic juvenile idiopathic arthritis, adult-onset Still's disease, relapsing polychondritis; viral infections including alpha virus (Chikungunya, Ross River) and flavivirus (Dengue and Zika Virus), flu, HIV; hidradenitis suppurativa (HS) and other cyst-causing skin diseases; cancers including lung cancer metastasis, pancreatic cancers, gastric cancers, myelodisplastic syndrome, leukemia; polymyositis; stroke; myocardial infarction; Graft versus Host Disease; hypertension; colitis; helminth infection; bacterial infection; abdominal aortic aneurism; wound healing; depression, psychological stress; pericarditis including Dressler's syndrome, ischaemia reperfusion injury and any disease where an individual has been determined to carry a germline or somatic non-silent mutation in NLRP3.
[00201 ] In one non-limiting example of those described, the disease, disorder or condition being treated is NASH. NLRP3 inflammasome activation is central to inflammatory recruitment in NASH, and inhibition of NLRP3 may both prevent and reverse liver fibrosis. Compounds of the present invention, by interrupting the function of NLRP3 inflammasomes in liver tissue, can cause histological reductions in liver inflammation, decreased recruitment of macrophages and neutrophils, and suppression of NF-κΒ activation. Inhibition of the NLRP3 can reduce hepatic expression of pro-IL-1 β and normalized hepatic and circulating I L- p , IL-6 and MCP-1 levels thereby assisting in treatment of the disease.
[00202] In a further non-limiting example of those described, the disease, disorder or condition being treated is severe steroid resistant (SSR) asthma. Respiratory infections induce an NLRP3 inflammasome/caspase-l/IL-Ιβ signaling axis in the lungs that promotes SSR asthma. The NLRP3 inflammasome recruits, and activates, pro-caspase-1 to induce IL-1 β responses. NLRP3 inflammasome-induced IL-1 β responses are therefore important in the control of infections, however, excessive activation results in aberrant inflammation and has been associated with the pathogenesis of SSR asthma and COPD. The administration of compounds of the first aspect that target specific disease processes, are more therapeutically attractive than non- specifically inhibiting inflammatory responses with steroids or IL-1 β. Targeting the NLRP3 inflammasome/caspase-1/IL-1 β signaling axis with the compounds of the first aspect may therefore be useful in the treatment of SSR asthma and other steroid-resistant inflammatory conditions.
[00203] In one further non-limiting example of those described, the disease, disorder or condition being treated is Parkinson's disease. Parkinson's is the most common neurodegenerative movement disorder and is characterized by a selective loss of dopaminergic neurons, accompanied by the accumulation of mis-folded a-synuclein (Syn) into Lewy bodies that are pathological hallmarks of the disease. Chronic microglial neuroinflammation is evident early in the disease, and has been proposed to drive pathology. [00204] A central role for microglial NLRP3 is postulated in Parkinson's progression. The NLRP3 inflammasome is activated by fibrillar Syn via a Syk kinase dependent mechanism, and also occurs in the absence of Syn pathology at the early stages of dopaminergic degeneration, and drives neuronal loss. The compounds of the first aspect may block NLRP3 inflammasome activation by fibrillar Syn or mitochondrial dysfunction and thereby confer effective neuroprotection of the nigrostriatal dopaminergic system and assist with treatment of Parkinson's.
[00205] In a sixth aspect of the invention there is provided a method of diagnosing a disease, disorder or condition in a mammal including the step of administering a labelled compound of formula (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof, to the mammal or to a biological sample obtained from the mammal to facilitate diagnosis of the disease disorder or condition in the mammal.
[00206] Inflammasome activation, in particular that of the NLRP3 inflammasome, is known to drive initiation, progression and chronic development of a vast number of inflammatory diseases. The sulfonylureas and related compounds of the first aspect are potent and specific direct inhibitors of NLRP3. Accordingly, a chemical probe specific for NLRP3, which is present in immune cells during inflammation has potential utility in diagnosing inflammatory and other related diseases. An NRLP3 activation probe comprising a compound of the first aspect could act as an effective surrogate biomarker of inflammatory diease for ex vivo (blood) or in vivo (MRI, PET etc.) diagnostics.
[00207] The use of the compounds of the first aspect in diagnosing inflammatory and other related diseases, such as those listed above, may be achieved by near infrared fluorescent imaging and ex vivo characterisation of immune cells by degree of inhibition of IL-1 beta, pro-caspase 1 cleavage and IL-18 levels. In particular, peripheral blood monocytes (PMBCs), macrophages, dendritic cells, CD4+ T cells, Th17 cells, Th1 cells and Th2 cells are relevant. In vivo diagnostics using magnetic resonance imaging (MRI). H2 (deuterium) 13C, 19F, 15N labelled variants of [compound classes] given to a patient IV, IM, SC, PO, topical, IT, etc.
[00208] In vivo diagnostics using positron emission tomography (PET) are also appropriate. PET is a molecular imaging technique that requires specific probes radiolabeled with short-lived positron emitting radionuclides. Typical isotopes include 11C, 13N, 150, 18F, 64Cu, 62Cu, 124l, 76Br, 82Rb and 68Ga, with 18F being the most clinically utilized. In particular it is possible to produce in a simple manner a stable 6 Cu or 62Cu salt of one or more of the compounds of formula (I) by simple ion exchange with a sodium (or other monovalent cation) salt of said compounds. This enables rapid preparation of a diagnostic probe for radioimaging, PET and the like whereby the intensity, location and temporal accretion of the diagnostic probe is able to identify the degree and/or the location of immune cells with activated NLRP3 as a surrogate biomarker of the patient's inflammatory state, and site of inflammation within the body. They will also be useful for application to biological samples removed from the body i.e. in vitro diagnosis.
[00209] A seventh aspect of the invention resides in a method of modulating the activity of a biological target comprising the step of exposing the biological target to a compound of formula (I) to (VII), or a pharmaceutically effective salt, solvate or prodrug thereof.
[00210] The biological target may be selected from the group consisting of NLRP3 inflammasome, IL-1 β, IL-17, IL-18, IL-1 a, IL-37, IL-33 and Th17 cells.
[0021 1 ] The modulation may be as described previously for the third to fifth aspects. [00212] As generally used herein, a biological sample may include cells, tissues, fluids, molecules or other biological materials obtained, or obtainable, from a mammal. Non-limiting examples include urine, blood and fractions thereof such as serum, plasma, lymphocytes and erythrocytes, cerebrospinal fluid, PAP smears, nasal and ocular secretions, amniotic fluid, faeces, semen, tissue and/or organ biopsies and nucleic acid (e.g. DNA, RNA) or protein samples, although without limitation thereto.
[00213] The following experimental section describes in more detail the characterisation of certain of the compounds of the invention and their efficacy. The intention is to illustrate certain specific embodiments of the compounds of the invention and their efficacy without limiting the invention in any way.
EXPERIMENTAL
General Synthetic Methods
Figure imgf000088_0001
Method A:
A1 : To a solution of R2 amine intermediate (1 eq.) with or without base such as, but not exclusively, triethylamine (1.2 eq.) in an anhydrous aprotic solvent such as, but not exclusively, tetrahydrofuran or dichloromethane was added triphosgene (0.4 to 1.1 eq.). The reaction was stirred at ambient temperature or, where necessary, heated at reflux until completion, typically from 2 to 18 h.
A2: To di-f-butyldicarbonate (1 .2-1 .4 eq.) in anhydrous acetonitrile or THF was added DMAP (15-100 mol%), after 5 minutes, a solution of R2 amine intermediate (1.0 eq.) in acetonitrile was added. The reaction mixture was stirred for 30-60 min at room temperature.
Method B:
B1 : The R2 carboxylic acid intermediate (1 eq.) was dissolved in an aprotic solvent such as toluene with or without 2 drops of DMF and a chlorinating agent such as thionyl chloride (2 eq.) added. The reaction mixture was heated at reflux until completion, then concentrated in vacuo. To give the corresponding R2 acid chloride intermediate.
Alternative methods or forming the acid chloride are also equally useful here for example the above procedure can be carried out without toluene and DMF thereby using thionyl chloride as both solvent and chlorinating agent.
The R2 acid chloride intermediate was dissolved in acetone and added drop- wise to a solution of sodium azide (1.5 eq) in a watenacetone (50:50) solution at 0 °C. Iced water was added to precipitate the resulting R2 acylazide intermediate which was dissolved in toluene and dried (MgS04) prior to adding the solution in a drop-wise fashion to anhydrous toluene at reflux while maintaining a constant flow of inert gas. The reaction was heated until completion, typically 2 h, to give the R2 isocyanate.
B2: The R2 acid chloride (formed as indicated in method B1 ) in dry CH2CI2 was added NaN3 (2.0 eq.) at 0 °C. The reaction mixture was stirred at room temperature for 1 h and extracted into EtOAc. The organic layer was washed with H20 (15 ml_), dried (MgS04), and carefully evaporated to give acyl azide. The acyl azide was dissolved in dry toluene and heated to 100 °C for 2 h. The solvent was removed to give crude R2 isocyanate.
Method C:
C1 : R1 sulfonamide intermediate (1 eq.) was dissolved in anhydrous THF and treated with NaH (1 eq.) under reduced pressure. The mixture was heated to reflux for 2 h then cooled to room temperature and R2 isocyanate intermediate in THF added under nitrogen atmosphere. The reaction mixture was stirred at reflux until completion.
C2: R1 sulfonamide intermediate (1 eq.) was dissolved in anhydrous THF or anhydrous methanol and treated with NaH (1 eq.) under reduced pressure. Once effervescence ceased the R2 isocyanate intermediate was added and the reaction mixture was stirred at ambient temperature overnight.
C3: To R1 sulfonamide intermediate (1 eq) in anhydrous THF (5 mUmmol) was added NaH (1 eq) at 0 C and stirred for 30 min to 2 h, or until completion, at ambient temperature under nitrogen atmosphere. Again cooled to 0 °C, R2 isocyanate (1 .0 eq) in THF was added and stirred at ambient temperature until completion, typically 2 to 16 h.
C4: To crude R2 isocyanate (1.0 eq) in anhydrous THF or DCM (5-1 1 mL/mmol) was added R1 sulfonamide (1 .0 eq) followed by base such as triethylamine , DIPEA, or DBU (1 -2 eq) and the reaction mixture stirred at ambient temperature overnight.
C5: To R1 sulfonamide intermediate (1 eq) in anhydrous MeOH (5 mL/mmol) was added NaOMe (1 eq) [alternatively: a 1 .0 mM solution of freshly prepared sodium methoxide (1 eq) was added to a 1.0 mM solution of R1 sulfonamide (1 eq) in anhydrous methanol]. The solvent was then removed in vacuo. The salt was suspended in anhydrous aprotic solvent such as acetonitrile or THF, the R2 isocyanate (1.0 eq) in anhydrous aprotic solvent such as acetonitrile or THF was added and the mixture stirred at ambient temperature overnight. The solution was then heated at reflux until completion, typically 90 min.
C6: R1 sulfonamide (1.0 eq.) was dissolved in anhydrous THF under a nitrogen atmosphere. Solid sodium methoxide (1 .0 eq mmol) was added in one portion. This mixture was stirred at ambient temperature for 3 h. A solution of the R2 isocyanate (1 .17 eq) in THF was added drop wise. The reaction mixture was stirred at room temperature overnight. n 0 c 0
R-NH2 *- R-S-CI * R-S-NH2
I I I I
O O
Method D:
A solution of amine (1 .0 eq) in acetonitrile (7-12 mL/mmol) at 0 °C was treated with c.HCI (1 .25-2.25 mL/mmol) in H20 (0.5-1.2 mL/mmol) followed by aqueous solution of NaN02 (1.2 eq) dissolved in H20 (0.3-0.5 mL/mmol of NaN02). The resulting solution was stirred at 0 °C for 45 min. AcOH (0.5-1 .2 mL/mmol), CuCI2.2H20 (0.5 eq) and CuCI (0.05 eq) were sequentially added to the above mixture and purged with S02 gas for 20 min at 0 °C. The resulting reaction mixture was stirred at 0°C- 10°C until completion.
Method E:
E1 : A solution of sulfonyl chloride (1 eq) in THF (10-20 mL/mmol) was cooled to -78 °C and ammonia gas was bubbled through the solution for 15 min, stirring was continued for a further 30 min then allowed to warm to ambient temperature and stirred for 2h or until completion.
E2: A solution of sulfonyl chloride (1 eq) in acetone (20 mL/mmol) was treated with a solution of NH4HCO3 (4 eq) dissolved in water 1 .5 mL/mmol of NH4HC03) at ambient temperature and stirred for 4 h or until completion.
E3: A solution of sulfonyl chloride (1 eq) in acetone (2.5 mL/mmol) was treated with NH3 (3.5 mL/mmol, NH OH in H20, 28% NH3 basis) at 0 °C and stirred for 2 h or until completion.
Method F
Figure imgf000092_0001
General Procedure for the synthesis of triazoles
Alkyne (1 eq) and azide (1 .2 eq), 5 mol% CuS04, 10 mol% NaAsc solution in DMSO (500 μΙ_) were stirred at room temperature until completion, typically 12 h.
Synthesis of R1 sulfonamide intermediates: Cyclohexanesulfonamide
Figure imgf000092_0002
To a solution of cyclohexanesulfonyl chloride (0.1 g, 0.54 mmol) in acetone (1 mL) was added aq NH3 (2 mL, 28% NH4OH in H20) at 0 °C and the reaction mixture stirred at room temperature for ~2 h. The solvent was removed in vacuo and MeOH/dichloromethane (1 :9) (5 mL) added the NH4CI by-product was removed by filtration and remaining solution concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 0.2% MeOH-CH2Cl2 eluent to give cyclohexanesulfonamide as an off-white solid (30 mg, 34%). 1H NMR (400 MHz, DMSO-afe): δ = 6.61 (br s, 2H), 2.76-2.70 (m, 1 H), 2.09-2.04 (m, 2H), 1.80-1 .76 (m, 2H), 1.65-1 .60 (m, 1 H), 1.31 -1 .19 (m, 4H), 1 .16-1 .06 (m, 1 H).
Cyclopentanesulfonamide
Figure imgf000092_0003
To a solution of cyclopentanesulfonyl chloride (0.1 g, 0.59 mmol) in acetone (1 mL) was added aq NH3 (1 mL, 28% NH OH in H2O) at 0 °C, and the reaction mixture stirred at room temperature for ~2 h. The solvent was removed in vacuo and MeOH/dichloromethane(1 :9) (5 mL) added the NH4CI by-product was removed by filtration and remaining solution concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 35% EtOAc- hexanes eluent to give cyclopentanesulfonam ide as an off-white solid (72 mg, 81 %). 1 H NMR (400 MHz, DMSO-afe): δ = 6.69 (br s, 2H), 3.42-3.32 (m, 1 H), 1 .89-1 .84 (m, 4H), 1 .68-1.64 (m, 2H), 1 .61 -1 .52 (m, 2H).
5-((dimethylamino)methyl)furan-2-sulfonamide
Figure imgf000093_0001
Furan-2-carboxylic acid (5 g, 44.6 mmol) was dissolved in ethanol (100 mL), c.H2S04 (1.0 mL) was added and the solution heated to reflux overnight. The reaction mixture was concentrated in vacuo then partitioned between ethyl acetate (100 mL) and saturated NaHC03 (100 mL). The organic phase was washed using water then brine, dried (MgS04) and concentrated in vacuo to give ethyl furan-2-carboxylate (4.5 g, 80%). 1H NMR (400 MHz, CDCI3): δ = 7.57 (d, J = 1 .2 Hz, 1 H), 7.18 (d, J = 3.5 Hz, 1 H), 6.51 (dd, J = 3.5, 1 .2 Hz, 1 H), 4.37 (q, J = 7.1 Hz, 2H), 1 .38 (t, J = 7.1 Hz, 3H).
Ethyl furan-2-carboxylate (9.0 g, 64.3 mmol) was dissolved in dichloromethane (200 mL) and chlorosulfonic acid (7.5 g, 64.3 mmol) added. The reaction was stirred at ambient temperature for 6 hours, or until completion, then pyridine (5.6 g, 70.7 mmol) and PCI5 (14.7 g, 70.7 mmol) were added portionwise. The reaction mixture was stirred at ambient temperature for 16 hours then quenched using ice-water and stirred for 30 mins. The mixture was extracted using DCM and the combined organics washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product, ethyl 5- (chlorosulfonyl)furan-2-carboxylate (7 g, 46%) was used directly without further purification. 1 H NMR (300 MHz, CDCI3) δ = 7.33 (d, J= 3.9 Hz, 1 H), 7.27 (d, J = 3.9 Hz, 1 H), 4.44 (q, J = 7.1 Hz, 1 H), 1 .42 (t, J = 7.1 Hz, 2H).
The crude ethyl 5-(chlorosulfonyl)furan-2-carboxylate (7 g) was converted using general method E1 to give ethyl 5-sulfamoylfuran-2-carboxylate (5 g, 78%). 1H NMR (300 MHz, DMSO-afe) δ = 8.05 (s, 2H), 7.38 (d, J = 3.7 Hz, 1 H), 7.09 (d, J = 3.7 Hz, 1 H), 4.32 (q, J = 7.1 Hz, 2H), 1 .29 (t, J = 7.1 Hz, 3H).
Ethyl 5-sulfamoylfuran-2-carboxylate (2 g, 9.13 mmol) in dry THF (40 ml_) was cooled to 0 °C and lithium aluminium hydride (1.05 g, 27.3 mmol) was added portion-wise over a period of 30 mins. The reaction was heated to 70 °C for 4 hours. The reaction was cooled to 0 °C and saturated NH4CI was added (20 ml_) dropwise with great care over a period of 30 mins. The reaction mixture was diluted using ethyl acetate (100 ml_) and filtered through a pad of celite. The organic phase was washed with water (100 ml_), brine (100 ml_), dried (MgS04) and concentrated in vacuo to give 5-(hydroxymethyl)furan-2- sulfonamide (1.25 g, 78%) as a pale brown liquid. 1 H NMR (400 MHz, DMSO- d6) δ 7.71 (s, 2H), 6.88 (d, J = 3.5 Hz, 1 H), 6.44 (d, J = 3.4 Hz, 1 H), 5.47 (s, 1 H), 4.44 (d, J = 5.7 Hz, 2H), 3.36 (s, 7H), 2.51 (q, J = 1 .8 Hz, 5H), 1.36 (s, 1 H).
5-(hydroxymethyl)furan-2-sulfonamide (0.3 g, 1.7 mmol) in THF (5 ml_) was cooled to 0 °C and POCI3 (0.4 g, 2.54 mmol) added slowly. The reaction mixture was stirred at 75 °C for 2 hours then cooled to ambient temperature. The crude mixture was partitioned between ethylacetate (50 ml_) and sat. aq. NaHC03 (50 ml_) and the organic phase washed with water (50 ml_), brine (50 ml_), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 30% EtOAc-hexanes eluent to give 5- (chloromethyl)furan-2-sulfonamide as a pale-brown semi-solid (0.25 g, 76%). H NMR (300 MHz, DMSO-d6) δ = 7.85 (s, 2H), 6.93 (dd, J= 3.5, 1 .3 Hz, 1 H), 6.69 (dd, J = 3.5, 1 .3 Hz, 1 H), 4.89 (d, J = 1 .3 Hz, 2H). 5-(chloromethyl)furan-2 -sulfonamide (0.4 g, 2.05 mmol) in THF (20 ml_) was cooled to 0 °C, c.HCI (7.5 mg, 2.05 mmol) was added and the solution stirred for 20 mins at this same temperature. 5.6 M N, A/-dimethylamine in ethanol (0.28 g, 6.15 mmol, 3 eq.) was added at 0 °C and the reaction tube sealed before stirring at room temperature overnight. The solvents were removed in vacuo and azeotroped using toluene (x2) to give 5- ((dimethylamino)methyl)furan-2-sulfonamide as a gum (0.25 g, 60%). The crude product was used directly without further purification.
Furan-2-sulfonamide
Figure imgf000095_0001
Furan-2-sulfonyl chloride (0.30 g, 1 .8 mmol) was added to aqueous ammonia (1.0 ml_) at 0 °C and the mixture was stirred at ambient temperature for 1 h. Upon completion of the reaction, the excess aqueous ammonia was removed in vacuo. The residue was azeotroped with isopropanol and triturated with pentane to afford the titled compound as a light brown solid (0.21 g, 79%).1H NMR (400 MHz, DMSO-afe): δ = 7.91 (s, 1 H), 7.45 (br.s. , 2H), 6.95 (d, J = 3.6 Hz, 1 H), 6.63 (dd, J = 2.8, 1 .6 Ηζ ,Η). LC-MS 97.4% (ELSD); m/z 146.1 1 [M - H]+.
5-methylfuran-2-sulfonamide
Figure imgf000095_0002
To a solution of 2-methylfuran (2.0 g, 24.3 mmol) in anhydrous acetonitrile (4 ml_) was added S03*Py complex (5.0 g, 31 .6 mmol) and the reaction mixture heated at 40 °C under nitrogen overnight. The reaction mixture was diluted with EtOAc (5 ml_) and stirred for 2 h at 0 °C, the resulting precipitates were removed by filtration and dried to give pyridinium 5-methylfuran-2-sulfonate as an off-white solid (2.93 g, 50%). 1 H NMR (400 MHz, DMSO): δ = 8.90 (dd, J1 = 4 Hz, J2 = 8 Hz, 2H), 8.57 (tt, J1 = 1.5 Hz, J2 = 8.1 Hz, 1 H), 8.04 (dd, J1 = 4 Hz, J2 = 8 Hz, 2H), 6.27 (d, J = 3.0 Hz, 1 H), 5.98-5.94 9m, 1 H), 2.19 (s, 3H).
A slurry of pyridinium 5-methylfuran-2-sulfonate (1 .0 g, 4.41 mmol) in anhydrous DME was treated with oxalyl chloride (0.53 ml_, 6.21 mmol) then DMF (0.32 ml_, 4.41 mmol) at 0 °C under argon and the reaction stirred at room temperature until completion. The reaction was quenched with ice-water and extracted with toluene (2 χ 50 ml_), the combined organics were washed with aqueous saturated NaHC03 (20 ml_), brine (20 ml_), dried (MgS04) and concentrated in vacuo to give 5-methylfuran-2-sulfonyl chloride as pale-yellow oil (350 mg, 47%). 1H NMR (400 MHz, CDCI3): δ = 7.23-7.21 (m, 1 H), 6.27-6.25 (m, 1 H), 2.47 (s, 3H).
To a solution of 5-methylfuran-2-sulfonyl chloride (0.2 g, 1 .10 mmol) in acetone (1 ml_) was added aq. NH3 (1 ml_, 28% NH OH in H20) at 0 °C. The reaction mixture was stirred at ambient temperature for ~2 h then concentrated in vacuo. The residue was suspended in dichloromethane (5 ml_) the NH4CI by-product was removed by filtration and remaining solution concentrated in vacuo. The crude product purified by column chromatography on silica gel using 40% EtOAc-hexanes eluent to give 5-methylfuran-2-sulfonamide as an off-white solid (130 mg, 73%). 1 H NMR (400 MHz, DMSO-afe): δ = 7.60 (s, 2H), 6.83-6.82 (d, J = 4.0 Hz, 1 H), 6.26-6.25 (d, J = 4.0 Hz, 1 H), 2.34 (s, 3H).
5-ethyl-N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)furan-2- sulfonamide
To a solution of 2-ethylfuran (2.0 g, 20.8 mmol) in anhydrous acetonitrile (3 ml_) was added S03*Py complex (4.30 g, 27.0 mmol). The resulting reaction mixture was heated at 40 °C under nitrogen atmosphere for 23 h or until completion. EtOAc (5 mL) was added and the solution stirred for 2 h at 0 °C. The resulting precipitate was removed by filteration and dried to give pyridin-1 -ium 5- ethylfuran-2-sulfonate as a brown coloured hygroscopic solid (3.2 g, 60%) which was used directly in the next step without purification.
To a slurry of pyridinium 5-ethylfuran-2-sulfonate (3.2 g, 12.5 mmol) in DME (15 mL) was added oxalyl chloride (1.62 mL, 27.0 mmol) and then DMF (0.97 mL, 12.5 mmol) at O °C under argon atmosphere, the resulting reaction mixture was stirred at room temperature until completion. The reaction mixture was quenched with ice-water and then extracted with toluene (2 χ 50 mL), organic layer was washed with aqueous saturated NaHCO3(20 mL) and brine (20 mL), dried (MgS04) and concentrated in vacuo to give 5-ethylfuran-2-sulfonyl chloride as light brown oil (510 mg, 21 %). 1 H NMR (400 MHz, CDCI3): δ 7.23 (d, J = 4 Hz, 1 H), 6.26 (d, J = 8 Hz, 1 H), 2.80 (q, J = 8 Hz, 2H), 1 .33 (t, J = 8 Hz, 3H).
To a solution of 5-ethylfuran-2-sulfonyl chloride in acetone (1 mL) was added aq NH3 (1.5 mL, NH OH in H20, 28% NH3 basis) at 0 °C, resulting reaction mixture was stirred at room temperature for 2 h or until completion. The solvent was removed in vacuo and azeotroped with toluene (x2). The residue was purified by column chromatography on silica using 1 % MeOH/DCM eluant to give 5-ethylfuran-2-sulfonamide as brown coloured gum (0.36 g, 78%). 1H NMR (400 MHz, DMSO-afe): δ 7.63 (bs, 2H), 6.85 (d, J = 4 Hz, 1 H), 6.28 (d, J= 4 Hz, 1 H), 2.70 (q, J = 8 Hz, 2H), 1 .21 (t, J = 6 Hz, 3H).
4-(prop-1-en-2-yl)furan-2-sulfonamide
Figure imgf000097_0001
Synthesis of 4-(prop-1 -en-2-yl)furan-2-sulfonamide was carried out from ethyl furan-3-carboxylate using procedures detailed by Urban et.al. Synth. Commun. 2003, 33(12), 2029-2043 to give the titled compound as a white solid with all spectral data consistent with the specified literature reference.
£/6-4-(prop-1-en-2-yl)furan-2-sulfonamide
Figure imgf000098_0001
Modification of the procedures contained within Urban et.al. Synth. Commun. 2003, 33(12), 2029-2043 to use methyl- /3-magnesium iodide in place of methyl magnesium chloride gives the corresponding /6-4-(prop-1 -en-2-yl)furan-2- sulfonamide.
4-(prop-1-en-2-yl)furan-2-sulfonamide
Figure imgf000098_0002
To a solution of triphenylphosphine (0.3 g, 1 .16 mmol) in anhydrous THF (5.0 mL) was added iodine (1 .0 eq.) and the mixture stirred at room temperature for 10 min. A solution of 4-(2-hydroxypropan-2-yl)furan-2-sulfonamide in THF (3.0 ml) was added slowly and stirring was continued for 2 h or until completion. The solution was diluted with EtOAc (20 mL), washed with 10% aq. sodium bisulfite (20 mL), water (20 mL). The organic layer was separated, dried (MgS04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 20% EtOAc:hexanes eluent to give the titled compound as a white solid (0.1 g, 58%). 1H N MR (400 MHz, CDCI3) 5 = 7.51 (s, 1 H), 7.16 (s, 1 H), 5.27 (s, 2H), 2.02 (s, 3H). 13C NMR (101 MHz, CDCI3) δ 149.3, 140.7, 140.7, 132.3, 127.8, 1 12.1 , 1 1 1 .9, 76.0, 7.7, 28.7, 19.8. 4-(2-hydroxypropan-2-yl)-5-methylfuran-2-sulfonamide
Figure imgf000099_0001
To a solution of ethyl 2-methyl-3-furoate (30 g, 0.195 M) in DCM (300 mL) at - 10 °C was added chlorosulfonic acid (23.8 g, 0.204 M) drop-wise over ~15 min. The reaction was allowed to warm to ambient temperature and stirred for 72 hours. The solution was cooled to -10 °C and anhydrous pyridine (16.9 g, 0.214 M) added drop wise followed by phosphorous pentachloride (44.6 g, 0.214 M) added in ~10 g portions over 10 min. Stirred at <0 °C for 30 min then stirred at ambient temperature overnight. The reaction mixture was added drop-wise to water (550 mL) with stirring and stirring continued for 2 hours. The organic phase was separated and the aqueous phase extracted using DCM (150 mL). The combined organics were washed using water (300 mL), dried (Na2S04) and concentrated in vacuo to give 44 g dark red oil. The crude product was purified by column chromatography on silica gel using 10% EtOAc-hexane eluent to give ethyl 5-(chlorosulfonyl)-2-methylfuran-3-carboxylate as an orange oil (36 g, 73%). 1H NMR (400 MHz, CDCI3) δ = 7.55 (s, 1 H), 4.63 (q, J= 7.2 Hz, 2H), 2.75 (s, 3H), 1.38 (t, J = 7.2 Hz, 3H).
Ethyl 5-(chlorosulfonyl)-2-methylfuran-3-carboxylate (30 g, 0.12 M) in acetone (200 mL) was added drop-wise over 15 min to a solution of ammonium bicarbonate (37.6 g, 0.475 M) in water (630 mL). The reaction mixture was stirred at ambient temperature until completion (~3 h). EtOAc (250 mL) was added and the pH adjusted using drop-wise addition of cHCI to pH~2. The organics were separated and the remaining aqueous phase saturated with sodium chloride and re-extracted using EtOAc (250 mL). The combined organics were washed with brine (300 mL), dried (Na2S04) and concentrated in vacuo to give a brown oily solid which was recrystallized using EtOAc-hexane to give ethyl 2-methyl-5-sulfamoylfuran-3-carboxylate as a beige solid (1 1 .4 g, 41 %). 1H N MR (400 MHz, DMSO-afe) δ = 7.8 (s, 2H), 7.02 (s, 1 H), 4.26 (q, J = 7.2 Hz, 2H), 2.62 (s, 3H), 1 .3 (t, J = 7.2 Hz, 3H).
Ethyl 2-methyl-5-sulfamoylfuran-3-carboxylate (10 g, 0.043 M) in anhydrous THF (400 mL) at -10 °C was treated with methyl magnesium chloride solution (3.0 M in THF, 64.3 mL) drop-wise over 5 minutes with vigorous stirring. The solution was then stirred at ambient temperature for 6 hours then cooled to -5 °C and treated drop-wise with a solution of ammonium chloride (51 .8 g in 265 mL water). The aqueous solution was extracted using EtOAc (2 x 250 mL), the combined organics washed with brine (250 mL), dried (Na2S04) and concentrated in vacuo to an orange oil (10 g). The crude product was purified by column chromatography on silica gel using 40% EtOAc-hexane eluent to give the titled compound as a white solid (6.1 g, 42%). 1 H NMR (400 MHz, DMSO-afe) δ = 7.54 (br.s. , 2H), 6.78 (s, 1 H), 4.95 (s, 1 H), 2.42 (s, 3H), 1 .4 (s, 6H). d6-4-(2-hydroxypropan-2-yl)-5-methylfuran-2-sulfonamide
Figure imgf000100_0001
Methyl 2-methyl-5-sulfamoylfuran-3-carboxylate can be prepared by modification of procedures used to synthesise ethyl 2-methyl-5-sulfamoylfuran- 3-carboxylate but using methyl 2-methylfuran-3-carboxylate as starting material in place of ethyl 2-methylfuran-3-carboxylate. Methyl 2-methyl-5-sulfamoylfuran- 3-carboxylate was obtained as a white solid (3 g, 29%) 1 H NMR (400 MHz, DMSO-d6) δ = 7.89 (s, 2H), 7.03 (s, 1 H), 3.79 (s, 3H), 2.61 (s, 3H).
Methyl 2-methyl-5-sulfamoylfuran-3-carboxylate (0.7 g, 3.2 mmol) in anhydrous THF (20 mL) at -10 °C was treated with c/3-methyl magnesium iodide solution (1.0 M in Et20, 26 mL) drop-wise over 10 minutes with vigorous stirring. The solution was then stirred at ambient temperature for 12 h then cooled to 0 °C and treated drop-wise with a solution of sat. ammonium chloride. The aqueous solution was extracted using EtOAc (2 x 25 mL), the combined organics washed with brine (25 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using a gradient of 40-70% EtOAc-hexane eluent to give the titled compound as a white solid (0.37 g, 51 %). 1H NMR (400 MHz, DMSO-d6) δ = 7.57 (s, 2H), 6.79 (s, 1 H), 4.99 (s, 1 H), 2.4 (s, 3H). 13C NMR (100 MHz, CD3OD) δ = 150.3, 147.3, 128.4, 1 13.4, 67.3, 28.5 (multiplet), 12.2.
1 -benzyl-1 Η-Λ ,2,4-triazole-3-sulf onamide
Figure imgf000101_0001
A solution of 1 H-1 ,2,4-triazole-3-thiol (1 g, 9.90 mmol) in DMF (20 mL) was treated with K2C03 (4.8g, 34.7 mmol), cooled to 0 °C then benzyl bromide (4.2 g, 24.8 mmol) was added drop-wise over 5 min. The resulting reaction mixture was warmed to ambient temperature and stirred for 12 h. The reaction mixture was diluted with water (25 mL) and extracted with ethyl acetate (2 x 25 mL). The combined organics were washed with water (20 mL), brine (20 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 20% EtOAc-hexanes eluent to give 1 - benzyl-3-(benzylthio)-1 H-1 ,2,4-triazole as a white solid (1 .5 g 54%). 1H NMR (400 MHz, DMSO-de): δ = 8.67 (s, 1 H), 7.39-7.32 (m, 5H), 7.27-7.21 (m, 5H), 5.36 (s, 2H), 4.29 (s, 2H).
To a solution of 1 -benzyl-3-(benzylthio)-1 H-1 ,2,4-triazole, 2 (0.5 g, 1 .77 mmol ) in acetonitrile (5 mL) at 0 °C was added AcOH (3 mL) and H20 (2 mL) then and Cl2 gas was bubbled through the solution for 45 min. Stirring was continued at O °C for 30 min then at 20 0 C for 1 .5h. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (2 x 20 mL). The combined organics were washed with water (20 mL), brine (20 mL), dried (Na2S04) and concentrated in vacuo to give a colorless liquid. The residue was diluted with THF and cooled to -78°C. Ammonia gas was bubbled through the solution for 20 min and stirring continued for a further 30 min before warming to ambient temperature and stirring for 1 h. The reaction mixture was diluted with water (25 mL) and extracted with ethyl acetate (2 x 25 mL). The combined organics were washed with water (20 mL), brine (20 mL), dried (Na2S04) and concentrated in vacuo. The residue was triturated with diethyl ether to give 1 -benzyl-1 HA ,2,4- triazole-3-sulfonamide as an off-white solid (0.25 g, 60%). 1 H NMR (400 MHz, DMSO-de): δ = 8.88 (s, 1 H), 7.77 (s, 2H), 7.39-7.33 (m, 5H), 5.45 (s, 2H).
1 -isopropyl-1 H-1 ,2,3-triazole-4-sulfonamide
Figure imgf000102_0001
Sodium 1 H-1 ,2,3-triazole-5-thiolate (500 mg, 4.06 mmol) was dissolved in EtOH (5 mL) and cooled to 0 °C. Benzyl bromide (0.69 g, 4.06 mmol) was added drop wise over a period of 5 min. The resulting reaction mixture was warmed to RT and stirred for 1 h. Upon completion, the reaction mixture was concentrated in vacuo and residue obtained was diluted with saturated NaHCO3 solution and extracted with EtOAc (2 x 20 mL). The combined organic extract was washed with water, brine, dried (Na2SO4) and concentrated in vacuo. The residue obtained was stirred with n-pentane (30 mL), filtered and dried in vacuo to give 4-(benzylthio)-1 - -1 ,2,3-triazole as a white solid (0.7 g, 90%) which was used without further purification. 1 H N MR (400 MHz, CDCI3): δ = 7.40-7.38 (m, 1 H), 7.35-7.21 (m, 5H), 4.12 (s, 2H). LCMS (m/z): 192.0 [M+H]+
A solution of 4-(benzylthio)-1 H-1 ,2,3-triazole (5 g, 26.1 mmol) in DMF (50 mL) was cooled to 0 °C and treated with K2C03 (9.03 g, 65.4 mmol). The reaction mixture was stirred for 5 minutes at same temperature. Isopropyl iodide (8.89 g, 52.3 mmol) was added drop wise to the above mixture over 5 min. The resulting reaction mixture was warmed to RT and stirred for 2h. Upon completion, the reaction mixture was diluted with water (30 mL) and extracted with ethyl acetate (50 mL). The organic extract was washed with water, brine and dried over anhydrous Na2S04. The crude product was purified by column chromatography on silica using, 8% EtOAc-hexane eluent to give 5-(benzylthio)-1 -isopropyl-1 H- 1 ,2,3-triazole A (0.9 g), 4-(benzylthio)-2-isopropyl-2H-1 ,2,3-triazole B (1 g) and the desired product 4-(benzylthio)-1 -isopropyl-1 H-1 ,2,3-triazole C (1.4 g, 23%). 1 H NMR (400 MHz, DMSO-afe): δ = 7.29-7.18 (m, 5H), 4.78-4.71 (m, 1 H), 4.09 (s, 2H), 1.4 (d, J = 6.8 Hz, 6H). LCMS (m/z): 234.30 [M+H]+
A solution of 4-(benzylthio)-1 -isopropyl-1 H-1 ,2,3-triazole (75 mg, 0.32 mmol) in acetic acid (2.25 mL) and H20 (1.12 mL) was cooled to 0 °C . N- chlorosuccinamide (170 mg, 1 .28 mmol) was added at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 1 h. Upon completion, the reaction mixture was diluted with water and extracted with ethyl acetate (2 x 10 mL). The combined organic extracts were washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica using, 8% EtOAc-hexanes eluant to give 1 -isopropyl- 1 HA ,2,3-triazole-4-sulfonyl chloride (0.1 g, 100%) as a pale brown liquid used without further purification. LCMS (m/z): 210.10 [M+H]+.
A solution of 1 -isopropyl-1 H-1 ,2,3-triazole-4-sulfonyl chloride (100 mg) in THF (5 mL) was cooled to -40 °C. Ammonia gas was purged through the aforementioned solution for 15 min. The reaction mixture was warmed to RT and stirred for 2h. Upon completion, the reaction mixture was concentrated in vacuo and residue obtained was diluted with ethyl acetate (25 mL) and water (10 mL). The organic extract was washed with water, brine, dried (Na2S04) and concentrated in vacuo to give 1 -isopropyl-1 HA ,2,3-triazole-4-sulfonamide (0.07 g, 78%) as brown solid. 1H NMR (400 MHz, DMSO-afe): δ = 8.71 (s, 1 H), 7.66 (s, 2H), 4.91 -4.87 (m, 1 H), 1 .5 (d, J =6.8 Hz, 6H). LCMS (m/z): 191.30 [M+H]+.
1 -methyl-1 H-pyrazole-3-sulf onamide
Figure imgf000104_0001
1 -Methyl-1 /-/-pyrazol-3-amine hydrochloride was reacted to 1 -methyl-1 H- pyrazole-3-sulfonyl chloride, a pale-yellow liquid, using general method D (0.7 g, 38%). 1H NMR (300 MHz, CDCI3): δ = 7.51 -7.50 (d, J = 2.1 Hz, 1 H), 6.89- 6.88 (d, J = 2.4 Hz, 1 H), 4.06 (s, 3H). LCMS (m/z): 160.9 (M -1 )". The sulfonyl chloride was converted using general method E1 to give the titled compound as an off-white solid (0.4 g, 69%). 1 H NMR (400 MHz, DMSO-d6): δ = 7.80 (d, J = 2.1 Hz, 1 H), 7.36 (s, 2H), 6.53 (d, J = 2.1 Hz, 1 H), 3.88 (s, 3H). LCMS (m/z): 162.05 (M +1 )+.
1 -(trif luoromethyl)-1 H-pyrazole-3-sulf onamide
Figure imgf000104_0002
1 -(trifluoromethyl)-1 /-/-pyrazol-3-amine was reacted to 1 -(trifluoromethyl)-1 H- pyrazole-3-sulfonyl chloride, a brown liquid, using general method D (0.4 g, 43%). 1H NMR (300 MHz, CDCI3) δ = 8.02 (d, J = 2.8 Hz, 1 H), 7.06 (d, J = 2.8 Hz, 1 H). 19F NMR (282 MHz, CDCI3) δ = -60.46.
The sulfonyl chloride was converted using general method E1 to give the titled compound (0.22 g, 46%). 1 H NMR (300 MHz, CDCI3) δ = 7.92 (dd, J = 2.8, 0.3 Hz, 1 H),6.91 (dd, J = 2.8, 0.7 Hz, 1 H), 5.28 (s, 2H). 19F NMR (282 MHz, CDCI3) δ = -60.41.
1 -isopropyl-1 H-pyrazole-3-sulf onamide
Figure imgf000105_0001
1 -lsopropyl-1 /-/-pyrazol-3-amine was reacted to 1 -isopropyl-1 - -pyrazole-3- sulfonyl chloride, a brown liquid, using general method D (0.5 g, 43 %). 1H NMR (400 MHz, CDCIs): δ = 7.55 (s, 1 H), 6.88 (s, 1 H), 4.66-4.63 (m, 1 H), 3.6 (br.s., 2H), 1 .59 (d, J = 6.8 Hz, 6H). LCMS (m/z): 209.0 (M+1 )+. The sulfonyl chloride was converted using general method E1 to give the titled compound as yellow solid (0.45 g, 82%). 1H NMR (300 MHz, DMSO-d6): δ = 7.9 (d, J = 2.4 Hz, 1 H), 7.36 (s, 2H), 6.55 (d, J = 2.1 Hz, 1 H), 4.57-4.53 (m, 1 H), 1.42 (d, J = 6.9 Hz, 6H). LCMS (m/z): 190.0 (M+1 )+.
1 -isopropyl-1 H-pyrazole-4-sulf onamide
Figure imgf000105_0002
A solution of 4-iodo-1 H-pyrazole (1 g, 5.15 mmol) in DMF (20 mL) was treated with K2CO3 (1.42g, 10.30 mmol) and isopropyl iodide (1 .05 g, 6.19 mmol) at ambient temperature under nitrogen atmosphere. The resulting reaction mixture was heated to 90 °C and stirred for 12 h. The mixture was cooled, diluted with water (50 mL) and extracted with diethyl ether (2 x 50 mL). The combined organics were washed with water (2 x 50 mL), brine (50 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 10% EtOAc-hexanes eluent to give 4-iodo- 1 -isopropyl-1 H-pyrazole as a colorless liquid (1 .1 g, 92%). 1 H NMR (400 MHz, CDCIs): δ = 7.50-7.46 (m, 2H), 4.53-4.47 (m, 1 H), 1 .50 (d, J = 6.8 Hz, 6H). LCMS (m/z): 237.2 (M +1 )+.
A solution of 4-iodo-1 -isopropyl-1 /-/-pyrazole (1 g, 4.24 mmol) in dioxane (20 mL) was treated sequentially with benzyl mercaptan (0.8g, 6.35 mmol) and DIPEA (1 .1 g, 8.47 mmol) under nitrogen atmosphere. The solution was degassed by purging with argon gas for 15 min. Pd2(dba)3 (40 mg, 0.0423 mmol) and Xantphos (50 mg, 0.0847 mmol) were added under argon atmosphere then the resulting mixture was sealed in the reaction vessel and heated at 75 °C for 6 h. The reaction mixture was cooled, concentrated in vacuo, diluted with water (20 mL) and extracted with EtOAc (2 x 20 mL). The combined organics were washed with water (2 x 50 mL), brine (50 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 10% EtOAc-hexanes eluent to give 4- (benzylthio)-l -isopropyl-1 /-/-pyrazole as a yellow liquid (650 mg, 66%). 1H NMR (400 MHz, CDCI3): δ = 7.36 (s, 1 H), 7.26-7.22 (m, 4H), 7.1 1 -7.09 (m, 2H), 4.41 - 4.36 (m, 1 H), 3.76 (s, 2H), 1 .42 (d, J = 6.8 Hz, 6H). LCMS (m/z): 233.3 (M +1 )+
To a solution of 4-(benzylthio)-1 -isopropyl-1 /-/-pyrazole, 3 (0.35 g, 1 .508 mmol) in acetonitrile (10 mL) at 0 °C was added AcOH (0.7 mL) and H20 (0.35 mL) then DCDMH (0.6 g, 3.017 mmol) was added portion-wise over 5 min. The solution was stirred for 30 min then warmed to ambient temperature and stirred for a further 2 h. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (2 x 20 mL). The combined organics were washed with water (20 mL), brine (20 mL), dried (Na2S04) and concentrated in vacuo to give 1 -isopropyl-1 H-pyrazole-4-sulfonyl chloride as a colorless liquid. The sulfonyl chloride was diluted with THF and cooled to -78 °C then NH3 gas was bubbled through the solution for 15 minutes. The reaction mixture was stirred at -78 °C for 1 h and at ambient temperature for 2 h. The reaction mixture was diluted with water and compound extracted with ethyl acetate (2 x 25 mL). The combined organic extracts were washed with water (20 mL), brine (20 mL), dried (Na2S04) and concentrated in vacuo. The residue obtained was triturated with diethyl ether and dried under reduced pressure give 1 -isopropyl-1 - -pyrazole-4- sulfonamide as a light brown solid (0.2 g, 71 %). 1 H N MR (400 MHz, DMSO-d6): δ = 8.21 (s, 1 H), 7.71 (s, 1 H), 7.22 (s, 2H), 4.59-4.53 (m, 1 H), 1 .4 (d, J= 6.8 Hz, 6H). LCMS (m/z): 190.2 (M +1 )+.
1 -cyclopropyl-1 H-pyrazole-3-sulfonamide
Figure imgf000107_0001
1 -Cyclopropyl-1 H-pyrazole-3-amine was reacted to 1 -cyclopropyl-1 H-pyrazole- 3-sulfonyl chloride using general method D then converted using general method E1 to give the titled compound as a light brown solid (0.2 g, 33%). 1H NMR (400 MHz, CDCI3) δ = 7.51 (d, J = 2.4 Hz, 1 H), 6.69 (d, J = 2.4 Hz, 1 H), 5.04 (s, 2H), 3.67 (m, 1 H), 1 .28 - 1.05 (m, 4H).
1 -(ferf-butyl)-l H-pyrazole-3-sulf onamide
Figure imgf000107_0002
1 -(te/ -butyl)-1 /-/-pyrazol-3-amine was reacted to 1 -(terf-butyl)-1 /-/-pyrazole-3- sulfonyl chloride using general method D then converted using general method E1 to give the titled compound as a light brown solid (150 mg, 26%). 1 H NMR (400 MHz, DMSO-de): δ = 7.56 (d, J= 3.6 Hz, 1 H), 6.7 (d, J
(br.s., 1 H), 1 .60 (s, 9H). LCMS (m/z): 204.15 (M+1 )+.
1 -cyclohexyl-1 H-pyrazole-3-sulfonamide
Figure imgf000108_0001
1 -Cyclohexyl-1 H-pyrazole-3-amine was reacted to 1 -cyclohexyl-1 H-pyrazole-3- sulfonyl chloride using general method D then converted using general method E1 to give the titled compound as a white solid (0.35 mg, 50%). 1H NMR (300 MHz, DMSO-d6) δ = 7.89 (d, J = 2.3 Hz, 1 H), 7.36 (s, 2H), 6.55 (d, J = 2.3 Hz, 1 H), 4.28 - 4.08 (m, 1 H), 2.0 1 .1 (m, 6H).
1 -phenyl-1 H-pyrazole-3-sulfonamide
Figure imgf000108_0002
1 -phenyl-1 /-/-pyrazol-3-amine was reacted to 1 -phenyl-1 - -pyrazole-3-sulfonyl chloride, a yellow liquid, using general method D (0.5 g, 47 %). 1H NMR (400 MHz, CDCI3): δ = 8.04 (d, J = 2.4 Hz, 1 H), 7.73 (d, J = 9.2 Hz, 2H), 7.58 (t, J = 7.6 Hz, 2H), 7.47 (t, J = 7.2 Hz, 1 H), 7.08 (d, J = 2.8 Hz, 1 H). The sulfonyl chloride was converted using general method E1 to give the titled compound as a yellow solid (0.4 g, 87 %). 1 H NMR (400 MHz, DMSO-d6): δ = 8.62 (d, J= 2.7 Hz, 1 H), 7.86 (d, J = 8.7 Hz, 2H), 7.61 (br.s., 2H), 7.57 (t, J= 7.8 Hz, 2H), 7.41 (t, J = 7.2 Hz, 1 H), 6.85 (d, J = 2.4 Hz, 1 H). LCMS (m/z): 224.1 (M+1 )+.
1-benzyl-1 H-pyrazole-3-sulfonyl chloride
Figure imgf000109_0001
1 -benzyl-1 H-pyrazol-3-amine was reacted to 1 -benzyl-1 - -pyrazole-3-sulfonyl chloride, a light brown liquid, using general method D (0.2 g, 45%). 1 H NMR (300 MHz, CDCIs): δ 7.42-7.38 (m, 3H), 7.33-7.28 (m, 3H), 6.8 (d, J =2.4Hz, 1 H), 5.42 (s, 2H). The sulfonyl chloride was converted using general method E1 to give the titled compound as a light brown liquid (0.15 g, 81 %). 1H NMR (400 MHz, CDCI3): δ = 7.42-7.36 (m, 4H), 7.24 (d, J= 1 .6Hz, 2H), 6.7 (d, J= 2.4Hz, 1 H), 5.35 (s, 2H), 5.10 (s, 2H). LCMS (m/z): 238.10 (M +1 )+
1 -(1 -phenylethyl)-1 H-pyrazole-3-sulf onamide
Figure imgf000109_0002
1 -(1 -phenylethyl)-1 H-pyrazol-3-amine was reacted to 1 -(1 -phenylethyl)-1 H- pyrazole-3-sulfonyl chloride using general method D then converted using general method E1 to give the titled compound as a white solid (0.25 mg, 68%). 1 H NMR (300 MHz, CDCI3) δ = 7.43 - 7.18 (m, 6H), 6.72 (d, J = 2.4 Hz, 1 H), 5.57 (q, J = 7.1 Hz, 1 H), 5.02 (s, 2H), 1 .92 (d, J = 7.1 Hz, 3H).
1 -(2-(piperidin-1 -yl)ethyl)-1 H-pyrazole-3-sulfonamide
Figure imgf000109_0003
1 -(2-(piperidin-1 -yl)ethyl)-1 /-/-pyrazol-3-am ine was reacted to 1 -(2-(piperidin-1 - yl)ethyl)-1 H-pyrazole-3-sulfonyl chloride, a pale-brown liquid, using general method D then converted using general method E1 to give the titled compound as an off-white solid (0.3 g, 46%). 1H NMR (300 MHz, DMSO-d6): δ = 7.84 (d, J = 2.1 Hz, 1 H), 7.36 (s, 2H), 6.54 (s, J = 2.4 Hz, 1 H), 4.26 (t, J = 6.9 Hz, 2H), 2.66 (t, J= 6.6 Hz, 2H), 2.36 (s, 4H), 1.46-1 .34 (m, 6H). LCMS (m/z): 259.10 (M
1 ,5-dimethyl-1 H-pyrazole-3-sulfonamide
Figure imgf000110_0001
1 ,5-dimethyl-1 - -pyrazol-3-amine was reacted to 1 ,5-dimethyl-1 /-/-pyrazole-3- sulfonyl chloride, a yellow liquid, using general method D (0.45 g, 26%). 1H NMR (300 MHz, CDCI3): δ = 5.92 (s, 1 H), 3.71 (s, 3H), 2.23 (s, 3H). LCMS (m/z):2M (M +Na)+. The sulfonyl chloride was converted using general method E1 to give the titled compound as an off-white solid (0.25 g, 55%). 1H NMR (400 MHz, DMSO-d6): δ = 7.30 (s, 2H), 6.36 (s, 1 H), 3.76 (s, 3H), 2.27 (s, 3H). LCMS (m/z): 175.9 (M +1 )+.
1 -methyl-5-(trif luoromethyl)-1 H-pyrazole-3-sulf onamide
Figure imgf000110_0002
1 -methyl-5-(trifluoromethyl)-1 /-/-pyrazol-3-amine was reacted to 1 -methyl-5- (trifluoromethyl)-l - -pyrazole-3-sulfonyl chloride, a pale-brown liquid, using general method D (1 .1 g, 37%). 1 H NMR (300 MHz, CDCI3): δ = 7.21 (s, 1 H), 4.16 (s, 3H). The sulfonyl chloride was converted using general method E1 to give the titled compound as a yellow solid (0.45 mg, 82%). 1H NMR (300 MHz, CDCI3): δ = 7.06 (s, 1 H), 5.02 (br.s. , 2H), 4.03 (s, 3H). 1 -isopropyl-5-(trif luoromethyl)-1 H-pyrazole-3- sulfonamide
Figure imgf000111_0001
A mixture of NaH (2.14 g, 89.3 mmol) in DMF (20 ml_) was cooled to -10 °C. A solution of 1 , 1 , 1 -trifluoropropan-2-one (5 g, 44.6 mmol) in DMF (80 mL) was added very carefully to the above mixture and stirred at -10 °C for 5 min. CS2 (10.2 g, 133.9 mmol) was added drop-wise to the above mixture over 30 min then the reaction mixture was warmed to ambient temperature and stirred for 1 h. The reaction mixture was cooled to 0 °C and treated with CH3I (7.5 mL) over 10 min. The resulting reaction mixture was warmed to ambient temperature and stirred for 12 h. The reaction mixture was diluted with cold water (50 mL) and extracted with diethyl ether (2 x 100 mL). The combined organic extracts were washed with water (50 mL), brine (50 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 5% EtOAc-hexanes eluent to give 1 , 1 , 1 -trifluoro-4,4-bis(methylthio)but-3- en-2-one as a light brown solid (3.5 g, 36%). 1H NMR (300 MHz, CDCI3): δ = 6.24 (s, 1 H), 2.57 (m, 6H). LCMS (m/z): 217.20 (M +1 )+.
A solution of 1 , 1 , 1 -trifluoro-4,4-bis(methylthio)but-3-en-2-one (2.5 g, 1 1.6 mmol) in EtOH (25 mL) was treated with isopropyl hydrazine hydrochloride (2 g, 13.9 mmol) at 0 °C, Et3N (2.4g, 40.98 mmol) was added and the mixture heated at 80°C for 12 h. The reaction mixture was concentrated in vact/o,di luted with sat. aq. NaHC03 solution and extracted with EtOAc (2 x 250 mL). The combined organics were washed with water (200 mL), brine (200 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 100% EtOAc eluent to give 1 -isopropyl-3- (methylthio)-5-(trifluoromethyl)-1 H-pyrazole as a light brown liquid (1.5 g, 58%). 1 H NMR (400 MHz, CDCI3): δ = 6.47 (s, 1 H), 4.58-4.53 (m, 1 H), 2.49 (s, 3H), 1 .50 (d, J = 6.8 Hz, 6H). LCMS (m/z): 225.20 (M +1 )+.
A solution of 1 -isopropyl-3-(methylthio)-5-(trifluoromethyl)-1 - -pyrazole (0.5g, 2.23 mmol) in chloroform (10 mL) at 0°C was treated with mCPBA (0.38 g, 2.23 mmol) and stirred at 10 °C for 1 h. The reaction mixture was diluted with saturated NaHC03 solution (10 mL) and extracted with CHCI3 (2 x 30 mL). The combined organics were washed with water (30 mL), brine (30 mL), dried (Na2S04) and concentrated in vacuo The residue obtained was dissolved in CHCI3 (10 mL) and treated with trifluoroacetic anhydride (1 .4 g, 6.7 mmol) the reaction mixture was heated at 50 °C for 3 h, cooled to ambient temperature and concentrated in vacuo. The residue obtained was diluted with MeOH (5 mL)-THF (5 mL)-H20 (5 mL), cooled to 0°C, treated with Na2C03 (0.7 g, 6.7 mmol) and stirred for 3h. The solution was diluted with water (30 mL) and extracted with CHCI3 (2 x 50 mL). The combined organics were washed with water (50 mL), brine (50 mL), dried (Na2S04) and concentrated in vacuo. The crude residue (0.2 g) containing 1 -isopropyl-3-((1 -isopropyl-5-(trifluoromethyl)- 1 H-imidazol-4-yl)disulfanyl)-5-(trifluoromethyl)-1 H-pyrazole was used in the next in step without further purification. 1H NMR (400 MHz, CDCI3): δ = 6.85 (s, 2H), 6.70 (s, 1 H), 6.60 (s, 1 H), 4.6 (m, 2H), 1 .53 (m, 6H). LCMS (m/z): 416.75 (M -1 )"
A solution of crude 1 -isopropyl-3-((1 -isopropyl-5-(trifluoromethyl)-1 H-imidazol-4- yl)disulfanyl)-5-(trifluoromethyl)-1 H-pyrazole (0.2 g crude, 0.478 mmol) in acetonitrile (10 mL) was cooled to 0°C and treated with AcOH (1 mL) and H20 (1.5mL). DCDMH (0.19 g, 0.956 mmol) was added portion-wise over 5 minutes and stirred for 2h. The mixture was diluted with water (20 mL) and extracted with DCM (2 x 20 mL). The combined organics were washed with water (50 mL), brine (50 mL), dried (Na2S04) and concentrated in vacuo to give 1 - isopropyl-5-(trifluoromethyl)-1 H-pyrazole-3-sulfonyl chloride as a colorless liquid. The (1 -isopropyl-5-(trifluoromethyl)-1 --pyrazole-3-sulfonyl chloride) was diluted with THF, cooled to -78 °C and NH3 gas was bubbled through the solution for 10 min then stirred for 1 h before warming to ambient temperature and stirring for a further 1 h. The reaction mixture was diluted with water and extracted with ethyl acetate (2 x 25 mL). The combined organics were washed with water (50 mL), brine (50 mL), dried (Na2S04) and concentrated in vacuo. The residue obtained was triturated with diethyl ether and n-pentane to give 1 - isopropyl-5-(trifluoromethyl)-1 /-/-pyrazole-3-sulfonamide as a white solid (75 mg, 61 %). 1H N MR (400 MHz, CDCI3): δ = 7.01 (s, 1 H), 5.06 (s, 2H), 4.73-4.70 (m, 1 H), 1 .5 (d, J = 6.8 Hz, 6H). LCMS (m/z): 256.0 (M -1 )".
5-isopropyl-1 -methyl-1 ft-pyrazole-3-sulf onamide
Figure imgf000113_0001
A solution of 1 -methyl-5-(prop-1 -en-2-yl)-1 -/-pyrazol-3-amine (0.25 g, 1.824 mmol) in acetonitrile (10 mL) at 0 °C was treated with c.HCI (1.2 mL) in H20 (0.5 mL) followed by aqueous solution of NaNO2 (0.15 g, 2.19 mmol) dissolved in H20 (2 mL). The resulting solution was stirred at O °C for 45 min. AcOH (0.25 mL), CuCI2.2H20 (0.15 g, 0.91 mmol) and CuCI (10 mg , 0.091 mmol) were sequentially added to the above mixture and purged with S02 gas for 20 min at 0 °C. The resulting reaction mixture was stirred at 0°C- 10°C for 60 min. Upon completion, the reaction mixture was diluted with water (20 mL) and extracted with EtOAc (2 x 20 mL). The combined organics were washed with water (20 mL), brine (20 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 20% EtOAc- hexanes eluent to give 1 -methyl-5-(prop-1 -en-2-yl)-1 - -pyrazole-3-sulfonyl chloride as a colourless liquid (0.15 g, 38%). 1H NMR (300 MHz, CDCI3): δ = 6.77 (s, 1 H), 5.51 (s, 1 H), 5.28 (s, 1 H), 4.02 (s, 3H), 2.1 1 (s, 3H).
A solution of 1 -methyl-5-(prop-1 -en-2-yl)-1 /-/-pyrazole-3-sulfonyl chloride (0.075 g, 0.34 mmol) in THF (7 ml_) was cooled to -78 °C and ammonia gas was bubbled through the solution for 15 min, stirring was continued for a further 30 min then allowed to warm to ambient temperature and stirred for 2h or until completion. The reaction mixture was diluted with ethyl acetate (25 ml_) and filtered through a pad of celite. The filtrate was dried (Na2S04) and concentrated in vacuo to give 1 -methyl-5-(prop-1 -en-2-yl)-1 - -pyrazole-3- sulfonamide as an off-white solid used without purification 0.04 g (crude).
A solution of crude 1 -methyl-5-(prop-1 -en-2-yl)-1 /-/-pyrazole-3-sulfonamide (0.12 g, 0.6 mmol) in MeOH (10 ml_) - EtOAc (4 ml_) was treated with 10% palladium on carbon (30 mg) under nitrogen atmosphere. The reaction flask was evacuated, filled with hydrogen (balloon) and stirred for 4 h. The reaction mixture was diluted with ethyl acetate (25 ml_), filtered through a pad of celite, dried (Na2S04) and concentrated in vacuo. The solid obtained was futher washed with diethyl ether to give 5-isopropyl-1 -methyl-1 - -pyrazole-3- sulfonamide as an off-white solid (0.1 1 g, 91 %). 1H N MR (400 MHz, CDCI3): 5 = 6.50 (s, 1 H), 5.00 (br.s., 2H), 3.87 (s, 3H), 2.97-2.93 (m, 1 H), 1 .28 (d, J =7.2 Hz, 6H).
5-(2-hydroxypropan-2-yl)-1-methyl-1 H-pyrazole-3- sulfonamide
Figure imgf000114_0001
ethyl 3-amino-1 -methyl-1 H-pyrazole-5-carboxylate was reacted to ethyl 3- (chlorosulfonyl)-l -methyl-1 H-pyrazole-5-carboxylate, a light-yellow liquid, using general method D (0.35 g, 47%). 1 H NMR (300 MHz, Chloroform-a δ 7.39 (s, 1 H), 4.40 (q, J= 7.1 Hz, 2H), 4.32 (s, 3H), 1.40 (t, J= 7.1 Hz, 3H).. The sulfonyl chloride was converted using general method E2 to give ethyl 1 -methyl-3- sulfamoyl-1 H-pyrazole-5-carboxylate as an off-white solid (0.3 g, 94%). 1H NMR (300 MHz, DMSO-de) δ = 7.59 (s, 2H), 7.09 (s, 1 H), 4.33 (q, J = 7.1 Hz, 2H), 4.14 (s, 3H), 1.31 (t, J = 7.1 Hz, 3H).
To a solution of ethyl 1 -methyl-3-sulfamoyl-1 H-pyrazole-5-carboxylate (0.25 g, 1 .07 mmol) in anhydrous THF (10 mL) at 0 °C was added methyl magnesium chloride (3 M in THF, 5 equivalents) drop-wise. The resulting reaction mixture was gradually warmed to ambient temperature and stirred for 6h or until completion. The solution was cooled to 0°C, quenched with sat. aq. NH4CI (2.0 mL) then diluted with cold water (20 mL) and extracted with EtOAc (2 x 25 mL). The combined organics were washed with brine (50 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 50% gradient of EtOAc in hexanes eluent to give the titled compound as a white solid. (0.2 g, 87%). 1 H NMR (400 MHz, DMSO-de) δ = 7.34 (s, 2H), 6.40 (s, 1 H), 5.48 (s, 1 H), 4.0 (s, 3H), 1 .50 (s, 6H).
1 -benzyl-5-(2-hydroxypropan-2-yl)-1 H-pyrazole-3-sulf onamide
Figure imgf000115_0001
Ethyl 3-amino-1 -benzyl-1 - -pyrazole-5-carboxylate was reacted to ethyl 1 - benzyl-3-(chlorosulfonyl)-1 - -pyrazole-5-carboxylate, a light-brown liquid, using method D (0.35 g, 47%). 1H NMR (400 MHz, CDCI3): δ = 7.41 (s, 1 H), 7.34-7.26 (m, 5H), 5.87(s, 2H), 4.37 (q, J = 7.2 Hz, 2H), 1 .38 (t, J = 7.2 Hz, 3H). The sulfonyl chloride was converted using general method E2 to give ethyl 1 -benzyl- 3-sulfamoyl-1 - -pyrazole-5-carboxylate as a white solid (0.7 g, 88%). 1 H NMR (400 MHz, DMSO-d6): δ = 7.66 (s, 2H), 7.39-7.27 (m, 3H), 7.2-7.18 (m, 3H), 5.77(s, 2H), 4.33 (q, J = 7.2 Hz, 2H), 1.29 (t, J = 7.2 Hz, 3H). LCMS (m/z): 310.05 (M +1 )+.
To a solution of ethyl 1 -benzyl-3-sulfamoyl-1 /-/-pyrazole-5-carboxylate (0.5 g, 1 .62 mmol) in anhydrous THF (10 mL) at 0 °C was added methyl magnesium chloride (3 M in THF, 2.77 mL, 8.1 mmol) drop-wise. The resulting reaction mixture was gradually warmed to ambient temperature and stirred for 4h or until completion. The solution was cooled to 0°C, quenched with sat. aq. NH4CI (2.0 mL) then diluted with cold water (20 mL) and extracted with EtOAc (2 x 25 mL). The combined organics were washed with brine (50 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 70-100% gradient of EtOAc in hexanes eluent to give the titled compound as a white solid. (0.27 g, 57%). 1 H NMR (300 MHz, DMSO-d6): δ = 7.37 (s, 2H), 7.39-7.27 (m, 3H), 7.2-7.18 (m, 2H), 6.45 (s, 1 H), 5.66 (s, 2H), 5.60 (s, 1 H), 1 .44 (s, 6H). LCMS (m/z): 296.1 (M +1 )+.
5-(2-hydroxypropan-2-yl)-1-phenyl-1 H-pyrazole-3-sulfonamide
Figure imgf000116_0001
2-(3-amino-1 -phenyl-1 - -pyrazol-5-yl)propan-2-ol was reacted to 5-(2- hydroxypropan-2-yl)-1 -phenyl-1 - -pyrazole-3-sulfonyl chloride, a yellow liquid, using method D (0.4 g, 36%). 1H NMR (300 MHz, CDCI3): δ = 7.55-7.45 (m, 5H), 6.91 (s, 1 H), 1 .51 (s, 6H). The sulfonyl chloride was converted using general method E2 to give the titled compound as a yellow solid (0.32 g, 87%). 1 H NMR (300 MHz, DMSO-d6): δ = 7.5 (s, 5H), 7.47 (s, 2H), 6.65 (s, 1 H), 5.41 (s, 1 H), 1.30 (s, 6H).
5-(dimethylamino)naphthalene-1-sulfonamide
Figure imgf000117_0001
5-(dimethylamino)naphthalene-1 -sulfonamide 3-azidobenzenesulfonamide was synthesized according to procedures contained in Satish K. Nair, Daniel Elbaum and David W. Christianson. J. Biol. Chem. 1996, 277.1003-100 and Lixuan Mu, Wensheng Shi, Guangwei She, Jack C. Chang, and Shuit-Tong Lee. Angew. Chem. Int. Ed. 2009, 48, 3469 -3472.
A solution of 5-(dimethylamino)naphthalene-1 -sulfonyl chloride (0.12 g, 0.44 mmol) in acetone (5 ml_) was added drop-wise to a solution of ammonium bicarbonate (0.17 g, 1 .76 mmol) in water (1 .0 ml_) and the reaction stirred at ambient temperature for 2 h, or until completion. The pH was adjusted using c. HCI to pH 2.0. The organic phase was separated and the aqueous phase was saturated with NaCI and extracted with ethyl acetate. The combined organic phases were washed with brinem dried (MgS04) and concentrated in vacuo to give the titled compound as a white solid (0.075 g, 67% yield). 1H NMR (600 MHz, CD3OD) δ = 8.54 (d, J= 8.5 Hz, OH), 8.36 (d, J= 8.7 Hz, 1 H), 8.23 (d, J = 7.3 Hz, 1 H), 7.58 (ddd, J = 17.0, 8.6, 7.4 Hz, 2H), 7.28 (d, J= 7.6 Hz, 1 H), 2.89 (s, 6H). 13C NMR (151 MHz, CD3OD) δ = 151 .6, 138.9, 129.7, 129.4, 129.2, 127.4, 126.6, 122.9, 1 19.5, 1 14.8, 44.4.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-2,3- dihydrobenzo[b]thiophene-6-sulfonamide 1 ,1 -dioxide
Figure imgf000117_0002
m-Chloroperbenzoic acid (77%, 6.35 g, 27.9 mmol) was added portion wise to a solution of benzo[£>]thiophene (1 .50 g, 1 1 .1 mmol) in anhydrous dichloromethane (100 ml_) at room temperature with vigorous stirring, the resulting reaction mixture was stirred for 16 h at the same temperature. A saturated aqueous NaHC03 solution (250 mL) was added and aqueous layer was extracted with dichloromethane (2 x 100 m L), organic layer was separated, combined organic layers dried (MgS04) and concentrated in vacuo. Crystallization from ethanol afforded benzo[b]thiophene 1 , 1 -dioxide (1 .56 g, 84%) as an off-white solid. 1 H NMR (600 MHz, CDCI3): δ = 7.73 (d, J = 6 Hz, 1 H), 7.58-7.53 (m, 2H), 7.38 (d, J = 12 Hz, 1 H), 7.23 (d, J= 6 Hz, 1 H), 6.73 (d, J = 6 Hz, 1 H). LCMS (m/z): 167 [M +H] +
A solution of benzo[b]thiophene 1 , 1 -dioxide (0.75 g, 4.51 mmol) in ethanol (55 mL) was degassed with nitrogen for 10 minutes then 10% Pd/C (10 mg) was added and the mixture stirred under hydrogen atmosphere (1 atm) for 24 h. The reaction mixture was filtered through a Celite pad, filtrate was concentrated to give 2,3-dihydrobenzo[£>]thiophene 1 , 1 -dioxide (0.74 g, 97%) as an off-white solid. 1 H NMR (600 MHz, CDCI3): 5 = 7.75 (d, J = 6 Hz, 1 H), 7.59 (t, J = 9 Hz, 1 H), 7.49 (t, J= 6 Hz, 1 H), 7.40 (d, J= 6 Hz, 1 H), 3.51 (t, J= 6 Hz, 2H), 3.41 (t, J = 6 Hz, 2H). LCMS (m/z): 169 [M +H] +
2,3-dihydrobenzo[b]thiophene 1 , 1 -dioxide (0.75 g, 4.45 mmol) was heated in chlorosulfonic acid (1 .5 mL, 22.2 mmol ) at 80 °C for 4 h. Reaction mixture was poured onto crushed ice and stirred for 5 minutes. The aqueous solution was extracted with dichloromethane (2x50 mL) and the combined organics dried (MgS04) and concentrated in vacuo to give 2,3-dihydrobenzo[£>]thiophene-6- sulfonyl chloride 1 , 1 -dioxide (0.45 g, 38%) as a light brown oil. The crude product was used directly in the next step without purification. 1H NMR (600 MHz, CDCIs): δ = 8.42 (s, 1 H), 8.25 (d, J = 12 Hz, 1 H), 7.69 (d, J = 6 Hz, 1 H), 3.64 (t, J = 9 Hz, 2H), 3.55 (t, J = 6 Hz, 2H).
To a solution of 2,3-dihydrobenzo[b]thiophene-6-sulfonyl chloride 1 , 1 -dioxide (0.45 g, 1.68 mmol) in acetone (1 mL) was added aq NH3 (2 mL, 28% NH OH in H20) at 0 °C, the resulting reaction mixture was stirred at room temperature for 2 h or until completion. The solvent was removed in vacuo and azeotroped with toluene (x2). The crude residue was purified by column chormatograpy on silica using 4% MeOH/CH2Cl2 eluent to give 2,3-dihydrobenzo[£>]thiophene-6- sulfonamide 1 ,1 -dioxide (0.16 mg, 39%) as an off-white solid. 1H-NMR (DMSO- d6): δ = 8.09 (s, 1 H), 8.06 (d, J = 12 Hz, 1 H), 7.75 (d, J = 6 Hz, 1 H), 7.60 (bs, 2h) 3.70 (t, J = 6 Hz, 2H), 3.44 (t, J = 9 Hz, 2H).
3-azidobenzenesulfonamide
Figure imgf000119_0001
Synthesised according to procedure contained in Pawan Kumar, Navneet Chandak, Poul Nielsen, Pawan K. Sharma. Bioorg. Med. Chem. 2012, 20, 3843-3849. A solution of 3-aminobenzenesulfonamide (0.3 g, 1 .7 mmol) in CH3CN (8 ml_) was cooled to 0 °C. To this stirred mixture was added f-BuONO (250 μΙ_, 2.1 mmol) followed by TMSN3 (276 uL, 2.1 mmol). The resulting solution was stirred at room temperature for 1 h. The reaction mixture was concentrated in vacuo and the crude product purified by column chromatography on silica gel using 100% hexanes eluant to give the titled compound as a pale yellow solid (0.31 g, 91 %). 1H N MR (600 MHz, CD3OD) 5 7.68 - 7.62 (m, 1 H), 7.56 (d, J = 1 .8 Hz, 1 H), 7.47 (d, J = 7.8 Hz, 1 H), 7.19 - 7.15 (m, 1 H). 13C NMR (151 MHz, DMSO-d6) δ 146.2, 140.8, 131.2, 122.9, 122.4, 1 16.5.
A-(3-Sulfamoylphenyl)pent-4-ynamide
Figure imgf000119_0002
To a solution of pent-4-ynoic acid (0.1 g, 1 .02 mmol) and 3- aminobenzenesulfonamide (0.21 g, 1 .22 mmol) in dry DMF (5.0 ml) was added HBTU (0.46 g, 1 .22 mmol) followed by DIPEA (212 uL, 1 .22 mmol). The reaction mixture was stirred at ambient temperature for 2 h, or until completion. The mixture was diluted with EtOAc (30 mL), washed with H20 (20 mL), brine (20 mL) then the organics dried (MgS04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 100% hexanes eluant to give the titled compound as a to give the titled compound as a pale-yellow solid (0.2 g, 79%). 1H NMR (400 MHz, CD3OD) δ = 8.22 (dd, J = 2.2, 1 .7 Hz, 1 H), 7.75 - 7.68 (m, 1 H), 7.65 - 7.58 (m, 1 H), 7.51 - 7.42 (m, 2H), 2.64 - 2.59 (m, 2H), 2.58 - 2.54 (m, 2H), 2.32 - 2.25 (m, 1 H). 13C NMR (101 MHz, CD3OD) 5 = 171 .3, 143.8, 138.9, 129.2, 122.9, 121 .0, 1 17.1 , 82.1 , 69.1 , 35.4, 14.0.
Benzene-1 ,3-disulfonamide
Figure imgf000120_0001
Benzene-1 ,3-disulfonyl dichloride (0.50 g, 0.726 mmol) was dissolved in tetrahydrofuran (4 mL) and the solution was cooled to 0 °C. aqueous ammonia (0.4 mL) was added at 0 °C and the mixture was stirred at ambient temperature for 1 h. Upon completion of the reaction, the mixture was poured into chilled water and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. The resulting solid was triturated with pentane to afford the titled compound as a light brown solid (0.16 g, 87%).1 H NMR (400 MHz, DMSO-afe): δ = 8.27 (t, J = 2.0 Hz, 1 H), 8.06 (dd, J = 2.0, 8.0 Hz, 2H), 7.81 (t, J = 8.0 Hz, 1 H), 7.64 (s, 4H).
A1,A1-dimethylbenzene-1 ,3-disulfonamide
Figure imgf000121_0001
3-amino-A/,A/-dimethylbenzenesulfonamide was converted to 3-(N,N- dimethylsulfamoyl)benzenesulfonyl chloride (0.45 g, 80%) using method D.1H NMR (300 MHz, CDCI3) δ = 8.42 (t, J =2.0 Hz, 1H), 8.27 (d, J= 7.9 Hz, 1H), 8.14 (d, J= 7.9 Hz, 1H), 7.85 (t, J= 7.9 Hz, 1H), 2.79 (s, 6H). The sulfonyl chloride was converted using general method E1 to give the titled compound as a yellow solid (0.45 g, 93%).1H NMR (400 MHz, DMSO-afe) δ = 8.13 (m, 2H), 7.98 (d, J= 7.9 Hz, 1H), 7.87 (t, J= 7.9 Hz, 1H), 7.65 (s, 2H), 2.65 (s, 6H).
Methyl 3-sulfamoylbenzoate
Figure imgf000121_0002
Methyl 3-(chlorosulfonyl)benzoate (1.00 g, 4.26 mmol) was dissolved in anhydrous tetrahydrofuran (15 mL) and the solution was cooled to 0 °C. Aqueous ammonia (5.0 mL) was added drop-wise and the mixture stirred at ambient temperature for 2 h. Upon completion the reaction mixture was poured into chilled water and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. The resulting solid was triturated with pentane to afford the titled compound as a light brown solid (0.75 g, 82%).1H NMR (400 MHz, DMSO-d6): δ = 8.40 (s, 1H), 8.19 (d, J=8Hz, 1H), 8.1 (d, J=8Hz, 1H), 7.77 (t, J=8Hz, 1H), 7.6 (s, 2H), 3.92 (s, 3H); m/z 214.0 [M -H+]\
3-(4-phenyl-1 H-1 ,2,3-triazol-1-yl)benzenesulfonamide
Figure imgf000122_0001
Ethynylbenzene (1 eq) and 3-azidobenzenesulfonamide (1 .2 eq), 5 mol% CuS04, 10 mol% NaAsc solution in DMSO (500 μΙ_) were stirred at room temperature for 12 h. The crude product was purified directly from the reaction mixture using reverse phase column chromatography (Reveleris flash column chromatography, 4 g, 18 mL/min.) and freeze dried to give the product as a white solid (32 mg, 70%). 1H NMR (400 MHz, DMSO-d6) δ = 9.57 - 9.36 (m, 1 H), 8.46 (d, J= 5.7 Hz, 1 H), 8.20 (s, 1 H), 7.98 (d, J= 8.1 Hz, 3H), 7.88 (d, J = 7.6 Hz, 1 H), 7.62 (s, 2H), 7.53 (d, J = 7.4 Hz, 2H), 7.42 (d, J = 7.5 Hz, 1 H). yV-(prop-2-yn-1-yl)-3-(4-sulfamoylphenyl)propanamide
Figure imgf000122_0002
To a solution of 3-(4-sulfamoylphenyl)propanoic acid (0.3 g, 1.5 mmol) and propargyl amine (0.1 1 g, 1 .5 mmol) in dry DMF (5.0 ml) was added HBTU (0.74 g, 1.5 mmol) followed by DIPEA (342 uL, 1 .22 mmol). The reaction mixture was stirred at RT for 2 h. The reaction was monitored by LCMS and after the completion of reaction, it was diluted with EtOAc (30 mL) washed with H20 (20 ml_), brine (20 mL). The organic layer was separated; dried (MgS04) and evaporated to give the crude product. The crude product was purified by silica gel column chromatography (1 : 1 , EtOAc:Hexane) to isolate the title compound as a white solid (0.22 g, 63%). 1 H NMR (400 MHz, CD3OD) 5 = 7.85 (d, J = 7.9 Hz, 2H), 7.38 (d, J = 7.9 Hz, 2H), 3.97 (t, J = 2.4 Hz, 2H), 3.04 (t, J = 7.6 Hz, 2H), 2.55 (t, J = 7.6 Hz, 2H), 2.33 (d, J = 2.8 Hz, 1 H). benzo[d][1 ,3]dioxole-5-sulfonamide
Figure imgf000123_0001
Sulfuryl chloride (2.18 ml, 26.7 mmol) was added to anhydrous DMF (2.10 ml, 26.7 mmol) at 0 °C under nitrogen atmosphere, then the ice bath was removed and the solution stirred for 15 minutes. The solution was cooled once more to 0 °C and benzo[d][1 ,3]dioxole was added. The reaction mixture was allowed to reach room temperature then heated at 100 °C for 2 h. The reaction mixture was poured onto crushed ice, stirred for 5 minutes, then extracted with dichloromethane (100 ml then 2 x 50 ml_). The combined organics were dried (MgS04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 15% DCM-hexanes eluent to give benzo[ ][1 ,3]dioxole-5-sulfonyl chloride as an off-white solid (1 .78 g, 33%). 1H N MR (400 MHz, CDCI3): 5 = 7.64 (d, J = 8.0 Hz, 1 H), 7.43 (s, 1 H), 6.95 (d, J = 8.0 Hz, 1 H), 6.16 (s, 2H).
To a solution of benzo[ ][1 ,3]dioxole-5-sulfonyl chloride (0.30 g, 1 .35 mmol) in acetone (1 ml_) was added aq. NH3 (1.5 ml_, 28% NH OH in H20) at 0 °C, the reaction mixture was stirred at room temperature until completion, typically 2 h, then concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 2% MeOH-DCM eluent to give the titled compound as an off-white solid (210 mg, 77%). 1 H NMR (400 MHz, DMSO-d6): δ = 7.32 (d, J= 8.0 Hz, 1 H), 7.25 (s, 1 H), 7.21 (bs, 2H), 7.02( d, J= 8.0 Hz, 1 H), 6.1 1 (s, 2H).
Pyridine-4-sulfonamide
Figure imgf000123_0002
Pyridine-4-sulfonyl chloride was converted using general method E3 to give the titled compound as a pale-yellow solid (50 mg, 56%).1 H NMR (300 MHz, DMSO-de): δ = 8.56 (d, J = 4.5 Hz, 1 H), 7.49 (d, J = 4.5 Hz, 1 H), 7.24 (br.s., 2H).
Pyridine-3-sulfonamide
Figure imgf000124_0001
Pyridine-3-sulfonyl chloride was converted using general method E3 to give the titled compound as a pale-yellow solid (0.7 g, 79%).1H NMR (300 MHz, DMSO- de) δ = 8.96 (dd, J= 2.5, 0.9 Hz, 1 H), 8.77 (dd, J= 4.8, 1 .6 Hz, 1 H), 8.17 (ddd, J = 8.0, 2.4, 1 .6 Hz, 1 H), 7.67 - 7.56 (m, 3H).
Pyridine-2-sulfonamide
Figure imgf000124_0002
A 1.0 M solution of HCI (45 mL) and DCM (45 mL) was cooled to -10 °C and pyridine-2-thiol (1 .0 g, 9.0 mmol) added. After 10 min, NaOCI (6% solution, 47 mL, 3.3 eq.) was added drop-wise over 5 min and stirring continued at -10 °C for 10 min. The organic phase was separated, dried using Na2S04 and filtered. The resulting solution was added drop-wise to a pre-cooled solution of sat. methanolic ammonia and DCM (1 : 1 , 40 mL) at 0 °C then allowed to warm to ambient temperature and stirred until completion, typically 2 h. The solvent was removed in vacuo to give a white solid which was dissolved in hot EtOAc and filtered to remove solid impurities. The solvent was removed in vacuo and recrystallized with EtOAc-hexanes to give the titled compound as a yellow solid (0.5 g, 35%). 1 H NMR (300 MHz, DMSO-d6) δ = 8.70 (ddd, J = 4.7, 1 .7, 0.9 Hz, 1 H), 8.05 (td, J = 7.7, 1 .7 Hz, 1 H), 7.91 (dt, J = 7.9, 1 .1 Hz, 1 H), 7.62 (ddd, J = 7.6, 4.7, 1 .2 Hz, 1 H), 7.45 (s, 2H).
4-(trifluoromethyl)pyridine-2-sulfonamide
Figure imgf000125_0001
4-(trifluoromethyl)pyridine-2-sulfonamide was synthesized according to the procedures used to synthesise pyridine-2-sulfonamide but using 4- (trifluoromethyl)pyridine-2-thiol in place of pyridine-2-thiol. The product 4- (trifluoromethyl)pyridine-2-sulfonamide was given as a solid (0.7 g, 56%). 1H N MR (300 MHz, DMSO-d6) δ = 9.02 (d, J = 5.0 Hz, 1 H), 8.16 (s, 1 H), 8.07 (d, J = 5.0 Hz, 1 H), 7.68 (s, 2H).
3-(3-(trifluoromethyl)-3H-diazirin-3-yl)benzenesulfonamide
Figure imgf000125_0002
3-(3-(trifluoromethyl)-3 - -diazirin-3-yl)aniline was converted using general method D to 3-(3-(trifluoromethyl)-3 - -diazirin-3-yl)benzenesulfonyl chloride, a yellow liquid (1 .1 g, 52%). 1H NMR (300 MHz, CDCI3) δ = 8.15 - 8.08 (m, 1 H), 7.82 - 7.77 (m, 1 H), 7.76 - 7.68 (m, 1 H), 7.68 - 7.61 (m, 1 H). 19F NMR (282 MHz, CDCI3) 5 -65.06.
3-(3-(trifluoromethyl)-3 - -diazirin-3-yl)benzenesulfonyl chloride was converted using general method E2 to the titled compound as a white solid (0.6 g, 60%). 1 H NMR (300 MHz, CDCI3) δ = 7.99 (dt, J= 7.9, 1 .5 Hz, 1 H), 7.71 (t, J= 2.0 Hz, 1 H), 7.60 (t, J = 7.9 Hz, 1 H), 7.49 (d, J = 7.9 Hz, 1 H), 4.87 (s, 2H). 19F NMR (282 MHz, CDCI3) 5 -65.13.
2-(methyl(7-nitrobenzo[c][1 ,2,5]oxadiazol-4-yl)amino)-N-(4- sulfamoylphenethyl)acetamide
Figure imgf000126_0001
2-(Methylamino)acetic acid (0.24 g, 2.75 mmol) and sodium hydrogencarbonate (0.694 g, 8.26 mmol) were dissolved in a mixture of water (10 ml_) and MeOH (20 ml_). Then, 4-chloro-7-nitrobenzo[c][1 ,2,5]oxadiazole (0.50 g, 2.50 mmol) was added and the mixture stirred at 60 °C for 2 h. Upon completion of the reaction, volatiles were removed under reduced pressure and the crude residue obtained was purified by column chromatography on silica gel using 0-5% gradient of methanol in dichloromethane to obtain 2-(methyl(7- nitrobenzo[c][1 ,2,5]oxadiazol-4-yl)amino)acetic acid as a brick-red solid (1 .10 g, 87%).
2-(Methyl(7-nitrobenzo[c][1 ,2,5]oxadiazol-4-yl)amino)acetic acid (1.00 g, 3.96 mmol) was dissolved in anhydrous tetahydrofuran (25 ml_) under nitrogen atmosphere and the solution was cooled to 0 °C. Diisopropylethylamine (0.76 g, 5.55 mmol) and 1 ,1 '-carbonyldiimidazole (0.90 g, 4.75 mmol) were added and the mixture stirred at 50 °C until all of the 2-(methyl(7- nitrobenzo[c][1 ,2,5]oxadiazol-4-yl)amino)acetic acid had reacted. The reaction mixture was then cooled to 0 °C, 4-(2-aminoethyl)benzenesulfonamide (0.95 g, 4.75 mmol) was added and stirred at ambient temperature until completion, typically 6 h. The solvents were removed in vacuo and the residue was purified by reverse phase preparative HPLC to afford the titled compound as a brick-red solid (1 .20 g, 70%). LCMS (m/z): 435.4 (M +1 )+.
4-(2-(7-Nitrobenzo[c][1 ,2,5]oxadiazol-4-ylamino)ethyl)benzenesulfonamide
Figure imgf000127_0001
4-(2-aminoethyl)benzenesulfonamide (0.55 g, 2.75 mmol) and diisopropylethylamine (0.64 g, 2.75 mmol) were dissolved in ethanol (20 ml_) and the solution cooled to 0 °C. 4-Chloro-7-nitrobenzo[c][1 ,2,5]oxadiazole (0.50 g, 2.50 mmol) was added at 0 °C and the mixture was stirred at ambient temperature for 16 h. Upon completion of the reaction, the reaction mass was poured into brine and extracted with ethyl acetate. Solvents evaporated from the combined organic extract under reduced pressure and the crude obtained was purified by reverse phase prep HPLC to afford the titled product as a dark yellow solid (0.250 g, 7%). 1H NMR (400 MHz, CD3OD) δ= 8.5 (d, J = 8.8 Hz, 1 H), 7.82 (d, J = 8.4 Hz, 2H), 7.47 (d, J = 8.4 Hz, 2H), 6.35 (d, J = 8.8 Hz, 1 H), 3.83 (m, 2H), 3.15 (t, J = 7.6 Hz, 2H).
2-(7-(Dimethylamino)-2-oxo-2H-chromen-4-yl)-N-(4- sulfamoylphenethyl)acetamide (171):
Figure imgf000127_0002
Molecular Weight: 429.49
2-(7-(dimethylamino)-2-oxo-2 - -chromen-4-yl)acetic acid (0.50 g, 2.02 mmol), EDC HCI (0.47 g, 3.03 mmol), HOBt (0.464 g, 3.03 mmol) and N- methylmorpholine (0.409 g, 4.04 mmol) were mixed in anhydrous tetrahydrofuran (5 ml_) and stirred at 0 °C for 30 min. 4-(2- Aminoethyl)benzenesulfonamide (0.445 g, 2.224 mmol) was added and stirring continued at ambient temperature for 18 h. Upon completion, the reaction was poured onto chilled water and extracted with ethyl acetate. The solvent was removed in vacuo and the residue was purified by column chromatography on silica gel using a gradient of 0-5% methanol in dichloromethane to give 2-(7- (dimethylamino)-2-oxo-2 - -chromen-4-yl)-A/-(4-sulfamoylphenethyl)acetamide as a greenish-yellow solid (0.25 g, 29%). LCMS (m/z): 430.2 (M +1 )+.
6,7-dihydro-5H-pyrrolo[1 ,2-a]imidazole-2-sulfonamide
Figure imgf000128_0001
A solution of 3-chlorobutanenitrile (20 g, 193.1 mmol) in diethyl ether (100 mL) was treated with MeOH (7.41 g, 231 .7 mmol) and cooled to 0 °C. HCI gas was bubbled into the reaction mixture for 4 h at 0 °C. The reaction mixture was stirred at -20 °C for 24 h and reaction mixture was concentrated in vacuo. The solid residue obtained was washed with diethyl ether (3 x 100 mL), n-pentane (2 x 100 mL) and dried in vacuo at 45 °C to give methyl 4-chlorobutanimidate hydrochloride as a white solid.
Methyl 4-chlorobutanimidate hydrochloride (25 g, 146.1 mmol) was dissolved in DCM (250 mL) treated with Et3N (44.3 g, 4.38 mmol) and resulting solution was cooled to 0 °C. 2,2-Dimethoxyethan-1 -amine (12.2 g, 1 16.9 mmol) was added dropwise to the above mixture over a period of 5 min. The resulting reaction mixture was warmed to 60 °C and stirred for 3 h. The reaction mixture was concentrated in vacuo and residue obtained was treated with in formic acid (150 mL) and heated at 80 °C for 24 h. Upon completion, the reaction mixture was concentrated in vacuo and residue obtained azeotroped with toluene (2 x 100 mL). The crude mixture was basified with saturated NaHC03 solution and extracted with DCM (3 x 200 mL). The combined organic extract was washed with water, brine, dried (Na2S04) and concentrated in vacuoio give 6,7-dihydro- 5 - -pyrrolo[1 ,2-a]imidazole (8 g, 39% over 3 steps) as a low melting dark solid. 1 H NMR (300 MHz, CDCI3): δ = 7.0 (s, 1 H), 6.83 (s, 1 H), 3.95 (t, J=7.2 Hz, 2H), 2.84 (t, J =7.2 Hz, 2H), 2.61 -2.51 (m, 2H).
A solution of 6, 7-dihydro-5 - -pyrrolo[1 ,2-a]imidazole (4 g, 37.0 mmol) in acetonitrile (120 mL) was cooled to 0 °C. A/-lodosuccinimide (9.16g, 40.7 mmol) was added portion wise at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 12h. Upon completion, the reaction mixture was diluted with saturated Na2S203 solution and extracted with ethyl acetate (2 x 50 mL). The combined organic extract was washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 4-40% EtOAc-hexanes eluant to give 2- iodo-6,7-dihydro-5H-pyrrolo[1 ,2-a]imidazole (1 .0 g, 19%) as a white solid. 1H NMR (300 MHz, CDCI3): δ = 7.03 (s, 1 H), 3.89 (t, J =7.2 Hz, 2H), 3.02 (t, J=7.2 Hz, 2H), 2.65-2.55 (m, 2H). LCMS (m/z): 235 [M+H]+
In a 50 mL re-sealable reaction tube, a solution of 2-iodo-6,7-dihydro-5 - - pyrrolo[1 ,2-a]imidazole (0.3 g, 1 .28 mmol) and phenylmethane thiol (0.24 g, 1 .92 mmol) in 1 ,4-dioxane (10 mL) was treated with DIPEA (0.41 g, 3.20 mmol) at RT under nitrogen atmosphere. Nitrogen gas was purged through the solution for 5 minutes. Xantphos (74mg, 0.128mmol) and Pd2(dba)3 (60 mg, 0.064 mmol) were sequentially added to the aforementioned solution and the vessel purged with nitrogen gas for 5 minutes. The resulting mixture was stirred at 1 10 °C for 12h. Upon completion, the mixture was cooled to RT, diluted with EtOAc (25 mL) and filtered through celite. The filtrate was dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 50-70% EtOAc-hexanes eluant to give 2-(benzylthio)-6,7-dihydro-5 --pyrrolo[1 ,2-a]imidazole, (0.15 g, 51 %) as a brown liquid. 1H NMR (300 MHz, CDCI3): δ = 7.26-7.22 (m, 3H), 7.12 (s, 1 H), 7.05-7.02 (m, 2H), 3.73 (s, 2H), 3.14 (t, J = 6.9 Hz, 2H), 2.81 (t, J= 7.2 Hz, 2H), 2.32-2.27 (m, 2H). LCMS (m/z): 231 .3 [M+H]+.
A solution of 2-(benzylthio)-6,7-dihydro-5 - -pyrrolo[1 ,2-a]imidazole (250 mg, 1 .08 mmol) in acetonitrile (2.5 mL), acetic acid (0.5 mL) and H20 (1. 2 mL) was cooled to 0 °C . DCDMH (170 mg, 0.869 mmol) was added at 0 °C and resulting reaction mixture was stirred at 0 °C for 2 h. The reaction mixture was diluted with water and extracted with ethyl acetate (2 x 20 mL). The combined organic extract was washed with water, brine, dried (Na2S04) and concentrated in vacuo to give 6,7-dihydro-5 - -pyrrolo[1 ,2-a]imidazole-2-sulfonyl chloride as a pale brown liquid used directly in the next step.
A solution of 6,7-dihydro-5 - -pyrrolo[1 ,2-a]imidazole-2-sulfonyl chloride (300 mg) in THF (15 mL) was cooled to -40 °C. Ammonia gas was purged through the aforementioned solution for 15 min and solution was stirred at -40 °C for 1 h. The reaction mixture was warmed to RT, stirred for 1 h then, upon completion, concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 10% MeOH-CHCI3 eluant to give 6,7-dihydro-5 --pyrrolo[1 ,2-a]imidazole-2-sulfonamide (1 17 mg, 88%) as a white solid. 1H NMR (300 MHz, DMSO-afe): δ = 7.54 (bs, 2H), 7.26 (s, 1 H), 4.06 (s, 2H), 2.80 (s, 2H), 2.32 (s, 2H). LCMS (m/z): 187.95 [M+H]+.
4-Nitrobenzenesulfonamide
Figure imgf000131_0001
4- Nitrobenzenesulfonyl chloride (1.0 eq.) dissolved in acetone (0.8 mL/mmol) was added drop-wise to Ammonium bicarbonate (4.0 eq) dissolved in water (0.8 mL/mmol). The reaction mixture was stirred at room temperature for 2 h before acidification with 1 M HCI (pH ~2). The mixture was extracted with ethyl acetate (3 x 10 mL), dried (MgS04) and concentrated in vacuo to give the titled compound as a pale orange solid (157 mg, 57%). 1 H NMR (600 MHz, DMSO- d6) δ = 8.42 (d, J = 8.8 Hz, 2H), 8.06 (d, J = 8.8 Hz, 2H), 7.74 (s, 2H). HRMS calculated for C6H5N204Si [M-H]" 200.9976, found 200.9984.
5- Methyl-A-(4-sulfamoylphenethyl)isoxazole-3-carboxamide
Figure imgf000131_0002
5-Methylisoxazole-3-carbonyl chloride (1 .0 eq) (prepared using general method B1 ) was dissolved in anhydrous THF (4 mL/mmol) and treated with triethylamine (1.0 eq). After stirring for 5 minutes 4-(2- aminoethyl)benzenesulfonamide (1 .0 eq) was added to the acid chloride solution. The reaction was stirred at room temperature, under an argon atmosphere overnight. The solvent was removed in vacuo, and the residue purified by reverse phase column chromatography using acetonitrile/10 mM ammonium bicarbonate (aq) as mobile phase to give the titled compound as a white solid (205 mg, 48%). 1H NMR (600 MHz, DMSO-afe) δ = 8.79 (t, J = 5.8 Hz, 1 H), 7.74 (d, J = 8.3 Hz, 2H), 7.41 (d, J = 8.3 Hz, 2H), 7.30 (s, 2H), 6.50 (q, J = 0.6 Hz, 1 H), 3.52 - 3.46 (m, 2H), 2.91 (t, J = 7.2 Hz, 2H), 2.45 (d, J = 0.6 Hz, 3H). HRMS calculated for Ci3H14N304Si [M-H]" 308.071 1 , found 308.0708. 4-(2-(7-Methoxy-4,4-dimethyl-1 ,3-dioxo-3,4-dihydroisoquinolin-2(1 H)- yl)ethyl)benzenesulfonamide
Figure imgf000132_0001
A/-(cyclohexylcarbamoyl)-4-(2-(7-methoxy-4,4-dimethyl-1 ,3-dioxo-3,4- dihydroisoquinolin-2(1 -/)-yl)ethyl)benzenesulfonamide (1 .0 eq) dissolved in anhydrous pyridine (8 mL/mmol) was treated with phthalic anhydride (1 eq.) and DMAP (0.1 eq) and heated to reflux under an inert atmosphere for 4 hours. The solvent was removed in vacuo, and the residue purified by reverse phase column chromatography using acetonitrile/1 0 mM ammonium bicarbonate (aq) as mobile phase to give the titled compound as a white solid (291 mg, 75%). 1 H NMR (600 MHz, DMSO-afe) δ = 7.72 (d, J = 8.4 Hz, 2H), 7.61 (d, J f= 8.7 Hz, 1 H), 7.53 (d, J = 2.8 Hz, 1 H), 7.40 (d, J = 8.2 Hz, 2H), 7.33 - 7.25 (m, 3H), 4.13 (d, J = 7.5 Hz, 2H), 3.83 (s, 3H), 2.93 (t, J = 7.3 Hz, 2H), 1 .45 (s, 6H). HRMS calculated for C20H2i N205Si [M-H]" 401 .1 177, found 401 .1 174.
Synthesis of R1 and R2 amine intermediates:
1-methyl-1 H-pyrazol-3-amine HCI
Figure imgf000132_0002
In a 20 ml_ microwave vial, a solution of 2-chloroacrylonitrile (2 g, 22.85 mmol) in EtOH (10 ml_) was treated with methyl hydrazine (1 .93g, 41 .13 mmol,). The resulting reaction mixture was heated at 100 °C for 10 minutes in a Biotage microwave synthesizer. The reaction mixture was left at < 5 °C for 12 h during this time a solid precipitated. The precipitate was removed by filtration and dried in vacuo to give the titled compound as white solid (0.12 g, 55%). 1H NMR (400 MHz, DMSO-d6): δ 7.76 (s, 1 H), 6.13 (s, 1 H), 3.80 (s, 3H), 2.58 (s, 2H). LCMS (m/z): 98.3 (M +1 )+.
1 -(trif luoromethyl)-1 H-pyrazol-3-amine
Figure imgf000133_0001
3-Nitro-1 H-pyrazole (5 g, 44 mmol) was dissolved in A/,A/-dimethylformamide (100 ml_), cooled to -5 °C and NaH (3.8 g, 93.6 mmol) added portionwise. The reaction mixture was stirred for 15 mins before adding dibromodifluoromethane (8.6 g, 44 mmol) and allowing to warm to ambient temperature overnight. The reaction mixture was quenched using ice-water and extracted using ethyl acetate. The organic phase was washed using water, brine, dried (Na2S04) and concentrated in vacuo to give 3-nitro-1 -(trifluoromethyl)-1 H-pyrazole (2.1 g, 22%) which was used without further purification. 19F NMR (282 MHz, CDCI3) = -34.20.
1 -(bromodifluoromethyl)-3-nitro-1 H-pyrazole (2.1 g, 9.76 mmol) was dissolved in DCM (50 ml_) and cooled to -78 °C before adding AgBF (5.7 g, 28.3, 3 equivalents). The reaction mixture was allowed to warm to ambient temperature overnight then cooled to 0 °C and quenched by addition of sat.aq. NaHC03 (50 ml_). The aqueous phase was extracted using DCM and the combined organics washed with water, brine, dried (Na2S04) and concentrated in vacuo to give 3- nitro-1 -(trifluoromethyl)-1 H-pyrazole (0.9 g, 51 %). 19F NMR (282 MHz, CDCI3) δ = -60.96. 3-nitro-1 -(trifluoromethyl)-l H-pyrazole (1 .0 g) was dissolved in THF:EtOAc (1 : 1 , 50 mL), Pd/C (200 mg) was added and the mixture stirred under a hydrogen atmosphere (balloon) overnight. The mixture was filtered through celite and washed through using ethyl acetate. The solvent was removed in vacuo and the residue purified by column chromatography on silica gel using 40% EtOAc in hexanes eluant to give the titled product (0.75 g, 87%). 1 H NMR (300 MHz, CDCIs) δ = 7.54 (d, J = 2.7 Hz, 1 H), 5.84 (d, J = 2.7 Hz, 1 H). 19F NMR (282 MHz, CDCI3) 5 = -61 .13.
1 -isopropyl-1 H-pyrazol-3-amine
Figure imgf000134_0001
Isopropyl hydrazine hydrochloride (5 g, 45.45 mmol) in water (40 mL) was treated sequentially with K2C03 (12.5 g, 91 mmol) and 2-chloroacrylonitile (4 g, 45.45 mmol). The resulting reaction mixture was stirred at 50 °C for 1 h, cooled to RT and extracted with ethyl acetate (50 mL). The organic extract was washed with water (40 mL), brine(40 mL), dried (Na2S04) and concentrated in vacuo to give the titled compound as a yellow solid (3.5 g, 62%). 1H NMR (400 MHz, CDCI3): δ = 7.15 (s, 1 H), 5.56 (s, 1 H), 4.27-4.23 (m, 1 H), 3.6 (br.s., 2H), 1 .43 (d, J = 6.4 Hz, 6H). LCMS (m/z): 126.0 (M+1 )+.
1 -cyclopropyl-1 H-pyrazol-3-amine
Figure imgf000134_0002
A solution of 1 /-/-pyrazol-3-amine (2 g, 24.1 mmol,) in AcOH (20 ml_) was treated with 2,5-hexane dione (5.7 g, 50.6 mmol) at ambient temperature under nitrogen atmosphere. The resulting reaction mixture was heated to 100 °C for6 h. The reaction mixture was concentrated under reduced pressure and azeotroped with toluene. The crude product was purified by column chromatography on silica gel using a gradient of 50-100% EtOAc-hexanes eluent to give 3-(2,5-dimethyl-1 H-pyrrol-1 -yl)-1 H-pyrazole as a red solid (2.25 g, 59%). 1 H NMR (400 MHz, DMSO-afe) δ 12.92 (s, 1 H), 7.85 (t, J = 1 .8 Hz, 1 H), 6.28 (t, J = 2.1 Hz, 1 H), 5.75 (s, 2H), 2.00 (s, 6H).
Copper (II) acetate (0.56 g, 3.1 mmol), 2, 2;-bipyridine (0.48 g, 3.1 mmol) and dichloroethane (10 ml_) were heated to 75 °C for 20 min. 5 ml_ of this pre- prepared solution was added to a m ixture of 3-(2,5-dimethyl-1 H-pyrrol-1 -yl)-1 H- pyrazole (0.5 g, 3.1 mmol), potassium cyclopropyltrifluoroborate (2 eq) and sodium carbonate (2eq.), in dichloroethane (5 ml_) then the reaction stirred at 75 °C for 6 h. The reaction mixture was diluted using DCM, washed using water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 10% EtOAc-hexanes eluent to give 1 -cyclopropyl-3-(2,5-dimethyl-1 H-pyrrol-1 -yl)-1 H-pyrazole as a yellow liquid (0.2 g, 32%). 1 H NMR (300 MHz, Chloroform-a δ 7.48 (dd, J= 2.3, 0.5 Hz, 1 H), 6.12 (d, J = 2.3 Hz, 1 H), 5.84 (s, 2H), 3.61 (tt, J = 7.3, 3.6 Hz, 1 H), 2.09 (s, 6H), 1 .22 - 0.95 (m, 4H).
To a solution of ammonium hydroxide hydrochloride (1 .64 g, 1 1 .8 mmol) in ethanol ((1 O mL) was added a solution of potassium hydroxide (0.66 g) in water (10 mL) at O °C. After 10 min stirring a solution of 1 -cyclopropyl-3-(2,5-dimethyl- 1 H-pyrrol-1 -yl)-1 H-pyrazole (0.95 g) in ethanol (10 mL) was added and the reaction heated at 100 °C for 20 h. The solvent was removed in vacuo and the residue partitioned between ethyl acetate and water. The organic phase was washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using a gradient of 70-100% EtOAc-hexanes eluent to give the titled product as a brown solid (0.4 g, 69%). 1H NMR (400 MHz, CDCI3) δ = 7.51 (d, J = 2.4 Hz, 1 H), 6.69 (d, J = 2.4 Hz, 1 H), 5.04 (s, 2H), 3.67 (m, 1 H), 1 .19 (m, 2H), 1 .12 (m, 2H).
1 -(ferf-butyl)-l H-pyrazol-3-amine
Figure imgf000136_0001
Isopropyl hydrazine hydrochloride (1 .42 g, 1 1 .4 mmol) in water (25 mL) at 0 °C was treated sequentially with K2C03 (1.57 g, 1 1 .4 mmol), NaHC03 (1.91 g, 22.9 mmol) and 2-chloroacrylonitile (1 g, 1 1 .4 mmol) then warmed to ambient temperature and stirred for 12h. The reaction mixture was diluted with water (20 mL) and extracted with ethyl acetate (2 x 25 mL). The combined organics were washed with water (30 mL), brine (30 mL), dried (Na2S04) and concentrated in vacuo to give the titled compound as a brown liquid (0.9 g, 60%). 1H NMR (400 MHz, DMSO-d6): δ = 7.34 (s, 1 H), 5.35 (s, 1 H), 4.51 (br.s., 2H), 1.40 (s, 9H). LCMS (m/z): 140.10 (M+1 )+.
1 -cyclohexyl-1 H-pyrazol-3-amine
Figure imgf000136_0002
3-nitro-1 H-pyrazole (1 g, 8.85 mmol) was dissolved in N,N-dimethylformamide (20 mL) and treated with potassium carbonate (1.47 g, 10.62 mmol) and bromocyclohexane (1 .8 g, 10.62 mmol). The mixture was heated to 100 °C for 16 hours (or until completion) then cooled to ambient temperature diluted using water (100 mL) and extracted using ethyl acetate (2 x 75 mL). The combined organics were washed using water (100 mL), brine (100 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using a gradient of 10% EtOAc-hexanes eluent to give 1 -cyclohexyl-3-nitro-1 - -pyrazole as a colourless liquid (1 .3 g, 76%). 1H NMR (300 MHz, CDCI3) δ = 7.47 (d, J = 2.5 Hz, 1 H), 6.88 (d, J = 2.5 Hz, 1 H), 4.26 - 4.1 1 (m, 1 H), 2.18 (m, 2H), 1 .93 (m, 2H), 1 .82 - 1 .20 (m, 6H).
In a 100 mL Parr shaker reaction vessel, a solution of 1 ,5-dimethyl-3-nitro-1 H- pyrazole (0.65 g, 3.3 mmol) in MeOH (4 mL) and EtOAc (20 mL) was treated with 10% palladium on carbon (200 mg) under nitrogen atmosphere. The flask was evacuated then filled with hydrogen gas (60 psi) and stirred at ambient temperature for 12h. The reaction mixture was diluted with ethyl acetate (50 mL) and filtered through a bed of Celite. The filtrate was dried (Na2S04) and concentrated in vacuo to give 1 -cyclohexyl-1 H-pyrazol-3-amine as a light brown solid (0.3 g 55%). 1H NMR (300 MHz, CDCI3) δ = 7.15 (d, J = 2.3 Hz, 1 H), 5.56 (d, J = 2.3 Hz, 1 H), 3.85 (m, 1 H), 3.62 (s, 1 H), 2.1 (m, 2H), 1 .8 (m, 2H), 1.77 - 1 .10 (m, 6H).
1 -phenyl-1 H-pyrazol-3-amine
Figure imgf000137_0001
Potassium te/ -butoxide (1 1 .9 g, 106.3 mmol) was dissolved in tBuOH (1 OOmL) and the solution was heated to 100 °C. Phenyl hydrazine (5 g, 46.2 mmol) and 3-ethoxy acrylonitrile (4.5 g, 46.2 mmol) were sequentially added and heating continued for 16 h. The mixture was concentrated in vacuo. The residue obtained was partitioned between water (500 mL) and ethyl acetate (500 mL). The organic extract was washed with water (250 mL), brine (250 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 25% EtOAc-hexanes eluent to give 1 - phenyl-1 H-pyrazol-3-amine as a pale brown solid (3.5 g, 48%). 1H NMR (400 MHz, CDCI3): δ = 7.69 (s, 1 H), 7.57 (d, J= 8.0 Hz, 2H), 7.41 (d, J= 8.0 Hz, 2H), 7.2 (t, J = 7.6 Hz, 1 H), 5.85 (s, 1 H), 3.83 (br.s., 2H). LCMS (m/z): 160.3 (M+1 )+.
1 -benzyl-1 H-pyrazol-3-amine
Figure imgf000138_0001
A solution of 3-nitro-1 H-pyrazole (1 g, 8.85 mmol) in THF (20 mL) was cooled to 0 °C and NaH (0.53 g, 13.27 mmol) was added. The suspension was stirred for 20 min then benzyl bromide (1.5 g, 8.85 mmol) was added drop-wise. The reaction was stirred until completion ~6 h, diluted with saturated NaHC03 solution (20 mL) and extracted with EtOAc (2 x 50 mL). The organics were washed with water (30 mL), brine (30 mL), dried (Na2S04) and concentrated in vacuo to give 1 -benzyl-3-nitro-1 H-pyrazole as a white solid (1.5 g, 84%). 1H NMR (300 MHz, CDCI3): δ = 7.40-7.36 (m, 4H), 7.31 -7.27 (m, 2H), 6.90 (d, J =2.7Hz, 1 H), 5.37 (s, 2H). LCMS (m/z): 204.20 (M +1 )+.
A solution of 1 -benzyl-3-nitro-1 H-pyrazole (1.5 g, 7.39 mmol) in THF (20 mL) and MeOH (5 mL) was cooled to 0 °C. Zinc powder (2.4 g, 36.9 mmol) and NH4CI solution (1.97g, 36.94 mmol; in 5 mL of water) was added. The resulting reaction m ixture was heated at 70 °C for 12 h. The reaction m ixture was cooled to ambient temperature, diluted with EtOAc (50 mL) and filtered through a bed of Celite. The filtrate was dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 50% EtOAc- hexanes eluent to give 1 -benzyl-1 /-/-pyrazol-3-amine as light brown liquid (0.85 g, 67%). 1H NMR (300 MHz, CDCI3): δ = 7.34-7.26 (m, 3H), 7.14-7.1 1 (m, 2H), 7.05 (d, J=2.4Hz, 1 H), 5.59 (d, J=2.4Hz, 1 H), 5.14(s, 2H). LCMS (m/z): 174.10 (M +1 )+
1-(1-phenylethyl)-1 H-pyrazol-3-amine
Figure imgf000139_0001
A solution of 3-nitro-1 H-pyrazole (1 g, 8.85 mmol) in THF (20 mL) was cooled to 0 °C and NaH (0.7 g, 17.7 mmol) was added. The suspension was stirred for 30 min then (l -bromoethyl)benzene (1.96 g, 10.6 mmol) was added drop-wise. The reaction was heated to 80 °C overnight or until completion, cooled to ambient temperature, diluted using water (40 mL) and extracted with EtOAc (2 x 50 mL). The organics were washed with water (30 mL), brine (30 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 20% EtOAc-hexanes eluent to give 3-nitro- 1 -(1 -phenylethyl)-1 H-pyrazole as a yellow liquid (1.2 g, 63%). 1 H NMR (300 MHz, CDCIs) δ 7.46 - 7.18 (m, 6H), 6.88 (d, J= 2.5 Hz, 1 H), 5.59 (q, J= 7.1 Hz, 1 H), 1 .96 (d, J = 7.1 Hz, 3H).
A solution of 3-nitro-1 -(1 -phenylethyl)-1 H-pyrazole (1 g, 4.6 mmol) in THF (20 mL) and MeOH (5 mL) was cooled to 0 °C. Zinc powder (1 .49 g, 23.04 mmol) and NH4CI solution (1.23 g, 23.04 mmol; in 5 mL of water) was added. The resulting reaction mixture was stirred for 30 mins then heated at 80 °C for 6 h. The reaction mixture was cooled to ambient temperature, diluted with EtOAc (50 mL) and filtered through a bed of Celite. The organic phase was washed using water (20 mL), brine (20 mL), dried (Na2SO4) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 50% EtOAc-hexanes eluent to give 1 -(1 -phenylethyl)-1 H-pyrazol-3-amine as a yellow liquid (0.85 g, 67%). 1 H NMR (300 MHz, CDCI3) δ = 7.39 - 7.04 (m, 6H), 5.59 (d, J = 2.4 Hz, 1 H), 5.35 (q, J = 7.1 Hz, 1 H), 3.83 (s, 2H), 1.78 (d, J = 7.1 Hz, 3H).
1 -(2-(piperidin-1 -yl)ethyl)-1 H-pyrazol-3-amine
Figure imgf000140_0001
A solution of 3-nitro-1 H-pyrazole (2 g, 17.7 mmol) in DMF (20 mL) was treated with 1 -(2-chloroethyl)piperidine hydrochloride (4.8 g, 26.5 mmol) at ambient temperature. The solution was cooled to 0 °C, and treated with K2CO3 (6.1 g, 44.27 mmol) in portions over a period of 5 min. The resulting reaction mixture was stirred at ambient temperature for 4 h. The reaction mixture was diluted with water and extracted with ethyl acetate (2 x 40 mL). The combined organics were washed with water (40 mL), brine (40 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 25% EtOAc-hexanes eluent to give 1 -(2-(3- nitro-1 /-/-pyrazol-1 -yl)ethyl)piperidine as a pale yellow solid (2.5 g, 64%). 1H NMR (400 MHz, CDCI3): δ = 7.60 (d, J = 2.4 Hz, 1 H), 6.81 (d, J = 2.4 Hz, 1 H), 4.29 (t, J= 6.4 Hz, 2H), 2.78 (t, J= 6.4 Hz, 2H), 2.41 (s, 4H), 1 .57-1 .53 (m, 4H), 1 .45 (t, J = 6 Hz, 2H). LCMS (m/z): 225.10 (M +1 )+.
A solution of 1 -(2-(3-nitro-1 /-/-pyrazol-1 -yl)ethyl)piperidine 3 (2.5 g, 1 1 .16 mmol) in THF (20 mL) and MeOH (5 mL) was cooled to 0 °C. The solution was sequentially treated with zinc powder (3.6 g, 55.8 mmol) and aqueous NH4CI (3 g, 55.8 mmol) solution then warmed to ambient temperature and stirred for 5 h. The reaction mixture was diluted with ethyl acetate (50 mL), filtered through a bed of Celite and concentrated in vacuo. The residue was diluted with ethyl acetate (60 mL) and washed with water (40 mL), brine (40 mL), dried (Na2S04) and concentrated in vacuoio give 1 -(2-(piperidin-1 -yl)ethyl)-1 /-/-pyrazol-3-amine as light yellow liquid (1 .75 g, 81 %). 1H NMR (400 MHz, DMSO-d6): δ = 7.28 (d, J = 2 Hz, 1 H), 5.30 (s, J = 2 Hz, 1 H), 4.50 (s, 2H), 3.90 (t, J= 6.8 Hz, 2H), 2.5- 2.53 (m, 4H), 2.39-2.33 (m, 6H), 1 .2 (s, 2H). LCMS (m/z): 195.10 (M +1 )+.
1 ,5-dimethyl-1 H-pyrazol-3-amine
Figure imgf000141_0001
A solution of 5-methyl-3-nitro-1 H-pyrazole (2 g, 15.7 mmol) in THF (20 mL) was cooled to 0 °C. NaH (0.7 g, 17.32 mmol) was added portion-wise over 10 min under nitrogen atmosphere. The resulting suspension was stirred for 10 min then treated with Mel (2.2 g, 15.7 mmol), warmed to ambient temperature and stirred for 4h. The reaction mixture was diluted with saturated NH4CI solution (20 mL) and extracted with EtOAc (2 x 30 mL). The organics were washed with water (30 mL), brine (30 mL), dried (Na2S04) and concentrated in vacuo to give 1 ,5-dimethyl-3-nitro-1 H-pyrazole as a white solid (2 g, 91 %). 1 H NMR (300 MHz, CDCIs): δ = 6.71 (s, 1 H), 3.87 (s, 3H), 2.34 (s, 3H).
In a 100 mL Parr shaker reaction vessel, a solution of 1 ,5-dimethyl-3-nitro-1 H- pyrazole (2 g, 14.18 mmol) in MeOH (4 mL) and EtOAc (20 mL) was treated with 10% palladium on carbon (400 mg) under nitrogen atmosphere. The flask was evacuated then filled with hydrogen gas (60 psi) and stirred at ambient temperature for 12h. The reaction mixture was diluted with ethyl acetate (50 mL) and filtered through a bed of Celite. The filtrate was dried (Na2S04) and concentrated in vacuo to give 1 ,5-dimethyl-1 H-pyrazol-3-amine as a light brown solid (1.36 g 87%). 1 H NMR (400 MHz, DMSO-d6): δ = 5.19 (s, 1 H), 4.33 (br.s. , 2H), 3.43 (s, 3H), 2.07 (s, 3H). LCMS (m/z): 1 12.3 (M +1 )+
1 -methyl-5-(trif luoromethyl)-1 H-pyrazol-3-amine
Figure imgf000142_0001
A solution of 1 , 1 , 1 -triethoxyethane (20 g, 123 mmol) in DCM (250 ml_) and pyridine (20.5g, 259 mmol) was cooled to 0 °C. A solution of trifluoroacetic anhydride (52 g, 246 mmol) in DCM (50 ml_) was added drop-wise over a period of 30 min. The reaction mixture was warmed to ambient temperature, stirred for 12h, then diluted with sat. aq. NaHC03 solution and extracted with DCM (2 x 250 ml_). The combined organics were washed with water (20 ml_), brine (20 ml_), dried (Na2S04) and concentrated in vacuo to give 4,4-diethoxy-1 , 1 , 1 - trifluorobut-3-en-2-one as a pale brown liquid (20 g, 76%). 1 H NMR (400 MHz, CDCI3): δ = 4.93 (s, 1 H), 4.39 (q, J= 7.2 Hz, 2H), 4.18 (q, J= 7.2 Hz, 4H), 1 .46- 1 .40 (m, 6H).
A solution of 4,4-diethoxy-1 ,1 , 1 -trifluorobut-3-en-2-one (10 g, 47.16 mmol) in acetonitrile (100 ml_) was treated with aqueous NH3 solution (15 ml_) at 0 °C then stirred at RT for 12 h. The reaction mixture was concentrated in vacuo then the residue was treated with water (250 ml_) and extracted with DCM (2 x 250 ml_). The combined organics were washed with water (250 ml_), brine (250 ml_), dried (Na2S04) and concentrated in vacuo to give (£)-4-amino-4-ethoxy- 1 ,1 , 1 -trifluorobut-3-en-2-one as an off white solid (7.5 g, 87%). 1H NMR (300 MHz, CDCI3): δ = 5.6 (br.s., 1 H), 4.17 (q, J = 7.2 Hz, 2H), 1 .42 (t, J = 7.2 Hz, 3H).
A solution of (E)-4-amino-4-ethoxy-1 , 1 , 1 -trifluorobut-3-en-2-one (5 g, 27.3 mmol) in EtOH (30 ml_) was treated with methyl hydrazine sulphate (4.72 g, 32.8 mmol) and Et3N (4.1 g, 41 .0 mmol) at ambient temperature. The resulting reaction mixture was heated at 85 °C for 12 h then cooled to ambient temperature and concentrated in vacuo. The residue obtained was diluted with sat. aq. NaHC03 solution (250 ml_) and extracted with EtOAc (2 x 250 ml_). The combined organics were washed with water (250 mL), brine (250 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 20% EtOAc-hexanes eluent to give 1 - methyl-5-(trifluoromethyl)-1 /-/-pyrazol-3-amine as a pale brown liquid (0.17 g, 38%). 1H NMR (400 MHz, CDCI3): δ = 5.93 (s, 1 H), 3.79 (s, 3H), 3.68 (br.s., 2H).
1 -methyl-5-(prop-1 -en-2-yl)-1 H-pyrazol-3-amine
Figure imgf000143_0001
A solution of 1 -methyl-1 -/-pyrazol-3-amine (2 g, 20.6 mmol,) in AcOH (50 mL) was treated with 2,5-hexane dione (4.9 g, 43.29 mmol) at ambient temperature under nitrogen atmosphere. The resulting reaction mixture was heated to 100 °C for 1 h then stirred at ambient temperature for 5 h. The reaction mixture was concentrated under reduced pressure and azeotroped with toluene. The crude product was purified by column chromatography on silica gel using 10% EtOAc- hexanes eluent to give 3-(2,5-dimethyl-1 H-pyrrol-1 -yl)-1 -methyl-1 H-pyrazole as a liquid (2.5 g, 69%). 1 H NMR (300 MHz, CDCI3): δ = 7.39 (d, J = 2.1 Hz, 1 H), 6.15 (d, J = 2.4 Hz, 1 H), 5.84 (s, 2H), 3.92 (s, 3H), 2.10 (s, 6H).
A solution of 3-(2,5-dimethyl-1 -/-pyrrol-1 -yl)-1 -methyl-1 H-pyrazole (1 g, 5.71 mmol) in dry THF (10 mL) was cooled to -78 °C under nitrogen atmosphere, n- BuLi (1 .6 M in hexanes, 4.4 mL, 6.86 mmol) was added drop-wise to the above solution over a period of 10 minutes then stirred at-78 °C for 1 h before treating with a solution of l2 (1 54g, 5.71 mmol) in THF (5 mL) at-78 °C stirring was continued at this temperature until completion (2 h). The reaction mixture was quenched with sat. aq. NH4CI solution and extracted with ethyl acetate (2 x 25 mL). The combined organics were washed with water (20 mL), brine (20 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 50% EtOAc-hexanes eluent to give 3-(2,5-dimethyl-1 H-pyrrol-1 -yl)-5-iodo-1 -methyl-1 /-/-pyrazole as an off-white solid (0.75g, 43.6%). 1 H N MR (400 MHz, CDCI3): δ = 6.33 (s, 1 H), 5.84 (s, 2H), 3.95 (s, 3H), 2.09 (s, 6H).
A solution of 3-(2,5-dimethyl-1 H-pyrrol-1 -yl)-5-iodo-1 -methyl-1 /-/-pyrazole (1 g, 3.32 mmol) in DME:water (8:2, 10 mL) was treated with 4,4,5,5-tetramethyl-2- (prop-1 -en-2-yl)-1 ,3,2-dioxaborolane (0.67g, 3.98 mmol) and Na2C03 (0.52g, 4.98 mmol) at ambient temperature under nitrogen atmosphere. The resulting solution was degassed by purging with argon for 15 min then treated with Pd(PPh3)4 (190 mg, 0.166 mmol) under argon atmosphere. The resulting mixture was heated at 90 °C for 24 h then cooled to ambient temperature and concentrated in vacuo. The residue obtained was diluted with cold water (20 mL) and extracted with EtOAc (2 x 20 mL). The combined organics were washed with water (20 mL), brine (20 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 5% EtOAc-hexanes eluent to give 3-(2,5-dimethyl-1 H-pyrrol-1 -yl)-1 - methyl-5-(prop-1 -en-2-yl)-1 /-/-pyrazole as a pale-yellow liquid (0.765 g, 92%). 1H NMR (300 MHz, CDCI3): δ = 6.08 (s, 1 H), 5.84 (s, 2H), 5.39 (s, 1 H), 5.23 (s, 1 H), 3.92 (s, 3H), 2.12 (s, 9H).
A solution of 3-(2,5-dimethyl-1 H-pyrrol-1 -yl)-1 -methyl-5-(prop-1 -en-2-yl)-1 H- pyrazole (0.7 g, 3.25 mmol) in EtOH-H20 (8:2, 12 mL) was treated with NH2OH.HCI (2.26 g, 32.55 mmol) and KOH (1 .8 g, 32.55 mmol). The resulting reaction mixture was heated at 100 °C for 48 h. The reaction mixture was cooled and concentrated in vacuo. The residue was treated with saturated NaHC03 to give a solution of pH~8 then extracted with EtOAc (2 x 50 mL). The combined organics were washed with water (20 mL), brine (20 mL), dried (Na2SO4) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 100% EtOAc eluent to give 1 -methyl-5- (prop-1 -en-2-yl)-1 H-pyrazol-3-amine as a light brown liquid (0.4 g, 91 %). 1H NMR (400 MHz, CDCI3): δ = 5.54 (s, 1 H), 5.27 (s, 1 H), 5.08 (s, 1 H), 3.71 (s, 3H), 2.41 (s, 2H), 1 .92 (s, 3H).
Ethyl 1-benzyl-3-nitro-1 H-pyrazole-5-carboxylate
Figure imgf000145_0001
A solution of 3-nitro-1 - -pyrazole-5-carboxylic acid (5 g, 31 .8 mmol) in ethanol (50 mL) was treated with thionyl chloride (4.5 g, 38.2 mmol) drop wise over a period of 10 min at 0 °C under a nitrogen atmosphere. The resulting mixture was stirred at 80 °C for 6 h then cooled to ambient temperature and concentrated in vacuo. The residue obtained was basified to pH 8 with saturated NaHC03 solution before extracting with ethyl acetate (2 x 100 mL). The combined organics were washed with water (100 mL), brine (100 mL), dried (Na2S04) and concentrated in vacuo. The crude product was triturated with diethyl ether, filtered and dried under reduced pressure to give ethyl 3- nitro-1 H-pyrazole-5-carboxylate as a white solid (5 g, 85%). 1H NMR (400 MHz, DMSO-de): δ = 7.44 (s, 1 H), 4.36 (q, J = 6.8 Hz, 2H), 1 .33 (t, J = 7.2 Hz, 3H). LCMS (m/z): 184 (M-1 ) \
Ethyl 3-nitro-1 - -pyrazole-5-carboxylate (1 g, 5.4 mmol) was dissolved in DMF (10 mL) at ambient temperature and treated with K2C03 (1.34 g, 9.7 mmol). The resulting mixture was cooled to 0 °C and methyl iodide (1.15 g, 8.1 mmol) was added drop-wise, the reaction mixture was sealed, allowed to warm to ambient temperature and stirred for 12 h. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (2 x 30 mL). The combined organics were washed with water (30 mL), brine (30 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 5% EtOAc-hexanes eluent to give ethyl 1 -methyl-3-nitro-1 - -pyrazole-5- carboxylate as a white solid (0.65 g, 61 %). 1H NMR (400 MHz, DMSO-afe) δ = 7.54 (s, J = 1.1 Hz, 1 H), 4.35 (q, J = 7.1 Hz, 2H), 4.19 (d, J = 1 .2 Hz, 3H), 1 .33 (t, J = 7.1 Hz, 3H).
Ethyl 1 -methyl-3-nitro-1 --pyrazole-5-carboxylate (0.65 g, 3.3 mmol) was dissolved in THF (20 mL) and MeOH (5 mL) at 0 °C. Zinc powder (1 .0 g, 16.3 mmol) and aqueous NH4CI (0.87 g, 16.3 mmol) were added sequentially. The resulting reaction mixture was stirred at ambient temperature for 4h, then heated to 70 C for 1 hour. The solvents were removed in vacuo. The residue obtained was dissolved in EtOAc (30 mL) and filtered through a bed of Celite. The filtrate was washed with water (30 mL), brine (30 mL), dried (Na2SO4) and concentrated in vacuo to give ethyl 3-amino-1 -methyl-1 - -pyrazole-5- carboxylate as a white solid (0.5 g, 91 %). 1 H NMR (400 MHz, DMSO-afe) δ 5.95 (s, 1 H), 4.24 (q, J = 7.1 Hz, 2H), 3.83 (s, 3H), 3.41 (s, 2H), 1 .27 (t, J = 7.1 Hz, 4H).
Ethyl 1-benzyl-3-nitro-1 H-pyrazole-5-carboxylate
Figure imgf000146_0001
A solution of 3-nitro-1 - -pyrazole-5-carboxylic acid (5 g, 31 .8 mmol) in ethanol (50 mL) was treated with thionyl chloride (4.5 g, 38.2 mmol) drop wise over a period of 10 min at 0 °C under a nitrogen atmosphere. The resulting mixture was stirred at 80 °C for 6 h then cooled to ambient temperature and concentrated in vacuo. The residue obtained was basified to pH 8 with saturated NaHCO3 solution before extracting with ethyl acetate (2 x 100 mL). The combined organics were washed with water (100 mL), brine (100 mL), dried (Na2S04) and concentrated in vacuo. The crude product was triturated with diethyl ether, filtered and dried under reduced pressure to give ethyl 3- nitro-1 H-pyrazole-5-carboxylate as a white solid (5 g, 85%). 1H NMR (400 MHz, DMSO-d6): δ = 7.44 (s, 1 H), 4.36 (q, J = 6.8 Hz, 2H), 1 .33 (t, J = 7.2 Hz, 3H). LCMS (m/z): 184 (M-1 ) \
Ethyl 3-nitro-1 - -pyrazole-5-carboxylate (2 g, 10.8 mmol) was dissolved in DMF (10 mL) at ambient temperature and treated with K2C03 (3 g, 21 .6 mmol). The resulting mixture was cooled to 0 °C and benzyl bromide (2.7 g, 16.2 mmol) was added drop-wise, the reaction mixture was allowed to warm to ambient temperature and stirred for 2 h. The reaction mixture was diluted with water (20 mL) and extracted with EtOAc (2 x 30 mL). The combined organics were washed with water (30 mL), brine (30 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 5% EtOAc-hexanes eluent to give ethyl 1 -benzyl-3-nitro-1 - -pyrazole-5- carboxylate as a white solid (1.2 g, 40%). 1 H NMR (400 MHz, CDCI3): δ = 7.44 (s, 1 H), 7.34-7.31 (m, 5H), 5.83(s, 2H), 4.39 (q, J= 7.2 Hz, 2H), 1 .38 (t, J= 7.2 Hz, 3H). LCMS (m/z): 276.15 (M +1 )+.
Ethyl 1 -benzyl-3-nitro-1 --pyrazole-5-carboxylate (1.2 g, 4.36 mmol) was dissolved in THF (20 mL) and MeOH (5 mL) at 0 °C. Zinc powder (1.4 g, 21 .8 mmol) and aqueous NH4CI (1.16g, 21 .8 mmol) were added sequentially. The resulting reaction mixture was stirred at ambient temperature for 4h, then concentrated in vacuo. The residue obtained was dissolved in EtOAc (30 mL) and filtered through a bed of Celite. The filtrate was washed with water (30 mL), brine (30 mL), dried (Na2S04) and concentrated in vacuo io give ethyl 3-amino- 1 -benzyl-1 H-pyrazole-5-carboxylate as a white solid (1 g, 94%). 1H NMR (300 MHz, DMSO-d6): δ = 7.30-7.19 (m, 3H), 7.1 1 -7.08(m, 2H), 6.00 (s, 1 H), 5.43 (s, 2H), 4.91 (s, 2H), 4.23 (q, J= 7.2 Hz, 2H), 1 .24 (t, J= 7.2 Hz, 3H). LCMS (m/z): 245.9 (M +1 )+. 3-(2,5-dimethyl-1 H-pyrrol-1-yl)-1 -phenyl-1 ft-pyrazole
Figure imgf000148_0001
A solution of 1 -phenyl-1 /-/-pyrazol-3-amine (3.5 g, 21 .9 mmol) in acetic acid (20 ml_) was treated with 2,5-hexadione (5.2 g, 45.9 mmol) and heated to 100 °C for 4 h. The mixture was cooled and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 5% EtOAc-hexanes eluent to give 3-(2,5-dimethyl-1 - -pyrrol-1 -yl)-1 -phenyl-1 H-pyrazole as a colorless liquid (2.8 g, 54%). 1 H NMR (400 MHz, CDCI3): δ = 7.98 (s, 1 H), 7.74 (d, J = 8.4 Hz, 2H), 7.49 (d, J = 8.4 Hz, 2H), 7.32 (t, J = 7.6 Hz, 1 H), 6.39 (s, 1 H), 5.9 (s, 2H), 2.19 (s, 6H).
A solution of 3-(2,5-dimethyl-1 -/-pyrrol-1 -yl)-1 -phenyl-1 /-/-pyrazole (2.7 g, 1 1.4 mmol) in THF (70 ml_) at -78 °C was treated drop-wise with n-BuLi (1 .6 M in THF, 10 ml_, 23.91 mmol) over 10 min. The reaction mixture was stirred at -78 °C for 1 .5 h then treated with freshly dried acetone (1 g, 17.0 mmol) and stirring continued at -78 °C for 1 .5 h. The reaction mixture was quenched with sat. ammonium chloride (2 ml_), concentrated in vacuo then partitioned between water (100 ml_) and ethyl acetate (100 ml_). The organic extract was washed with water (100 ml_), brine (100 ml_), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 20% EtOAc-hexanes eluent to give 2-(3-(2,5-dimethyl-1 H-pyrrol-1 -yl)-1 -phenyl- 1 H-pyrazol-5-yl)propan-2-ol as an off white solid (1 .4 g, 42%). 1H NMR (400 MHz, CDCI3): δ = 7.57-7.56 (m, 2H), 7.47-7.46 (m, 3H), 6.23 (s, 1 H), 5.85 (s, 2H), 2.19 (s, 6H), 1 .52 (s, 6H). LCMS (m/z): 296.1 (M+1 ) +. In an 100 mL re-sealable reaction tube, 2-(3-(2,5-dimethyl-1 -/-pyrrol-1 -yl)-1 - phenyl-1 - -pyrazol-5-yl)propan-2-ol (1.4 g, 4.74 mmol) was dissolved in EtOH- H20 (1 : 1 , 50 mL) at ambient temperature. Hydroxyl amine hydrochloride (3.3 g, 47.45 mmol), and KOH (2.6 g, 47.45 mmol) were added sequentially and resulting reaction mixture was heated at 120 °C for 16 h. The reaction mixture was concentrated in vacuo, diluted with water (50 mL) and extracted with ethyl acetate (2 x 50 mL). The organic extracts were washed with water (50 mL), brine (50 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 50% EtOAc-hexanes eluent to give 2-(3-amino-1 -phenyl-1 - -pyrazol-5-yl)propan-2-ol as a colorless liquid (0.8 g, 78%). LCMS (m/z): 218.1 (M+1 )+.
8-bromo-1 ,2,3,5,6,7-hexahydro-s-indacen-4-amine
Figure imgf000149_0001
A/-Bromosuccinimide (1.02 g, 5.78 mmol) was added portion-wise to a solution of 1 ,2,3,5,6,7-hexahydro-s-indacen-4-amine (1 g, 5.78 mmol) in DCM (20 mL) at 0 °C. The solution was gradually warmed to ambient temperature and stirred for 12 h. The reaction mixture was diluted with sat. aqueous Na2S203 (50 mL) and extracted with DCM (2 x 25 mL). The combined organic extracts were washed with water (25 mL), brine (25 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 5% EtOAc-hexane eluent to give 8-bromo-1 ,2,3,5,6,7-hexahydro-s- indacen-4-amine as a brown solid (1 .2 g, 83%). 1 H N MR (300 MHz, CDCI3): 5 = 3.45 (br.s., 2H), 2.92-2.88 (m, 4H), 2.81 -2.77 (m, 4H), 2.16-2.09 (m, 4H); LCMS 94% (210 nM); m/z 252.15 [M +H]+.
8-chloro-1 ,2,3,5,6,7-hexahydro-s-indacen-4-amine
Figure imgf000150_0001
A/-Chlorosuccinimide (0.46 g, 3.46 mmol) was added portion-wise to a solution of 1 ,2,3,5,6,7-hexahydro-s-indacen-4-amine, 1 (0.6 g, 3.46 mmol) in CHCI3 (10 mL) at 0 °C. The solution was gradually warmed to ambient temperature and stirred for 10 h. The reaction mixture was diluted with sat. aqueous Na2S203 (50 mL) and extracted with DCM (2 x 25 mL). The combined organic extracts were washed with water (25 mL), brine (25 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 10% EtOAc-hexane eluent to give 8-chloro-1 ,2,3,5,6,7-hexahydro-s- indacen-4-amine as a brown solid (0.45 g, 63%). 1H NMR (300 MHz, CDCI3): δ = 2.94 (t, J = 7.2 Hz, 4H), 2.77 (t, J = 8.1 Hz, 4H), 2.18 (m , 4H); m/z 207.8 [M
+H]+.
8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacen-4-amine
Figure imgf000150_0002
8-Bromo-1 ,2,3,5,6,7-hexahydro-s-indacen-4-amine (400 mg, 1 .59 mmol) was dissolved in 1 ,4-dioxane-water (8:2, 10 mL) and the reaction flask purged with argon gas for 15 min. K2C03 (650 mg, 4.78 mmol), methyl boronic acid (100 mg, 1.75 mmol) and Pd(PPh3)4 (100 mg, 0.079 mmol) were sequentially added under argon atmosphere. The resulting mixture was sealed and heated at 100 °C for 2 h. The reaction mixture was cooled, diluted with water and extracted using EtOAc (2 x 20 mL). The combined organic extracts were washed with water (25 mL), brine (25 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 5% EtOAc-hexane eluent to give 8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacen-4- amine as a colorless liquid (0.220 g, 76%). 1H NMR (400 MHz, CDCI3): δ = 3.41 (br.s., 2H), 2.88-2.8 (m, J = 7.5 Hz, 4H), 2.75-2.67 (m, 4H), 2.18-2.09 (m, 7H); m/z 188.2 [M +H]+.
3,5,6,7-tetrahydro-2H-indeno[5,6-d]furan-8-amine
Figure imgf000151_0001
A solution of 2,3-dihydrobenzofuran-5-carbaldehyde (10 g, 67.6 mmol), malonic acid (10.5 g, 101.35 mmol) and piperidine (0.47 ml_, 4.73 mmol, 0.07 eq) was heated in pyridine (60 ml_) at 100 °C for 5 h. The reaction mixture was acidified to ~pH 3 using 1 N HCI and the product extracted using 10% IPA/chloroform (2 x 250 ml_). The combined organic extracts were washed with water (250 ml_), brine (250 ml_), dried (Na2S04) and concentrated in vacuo. The crude product was triturated using diethyl ether to give (E)-3-(2,3-dihydrobenzofuran-5- yl)acrylic acid as a yellow solid (10 g, 78%). 1 H NMR (300 MHz, Chloroform-a δ = 7.73 (d, J = 15.9 Hz, 1 H), 7.43 (s, 1 H), 7.33 (dd, J = 8.1 , 1 .8 Hz, 1 H), 6.80 (d, J = 8.1 Hz, 1 H), 6.29 (d, J = 15.9 Hz, 1 H), 4.64 (t, J = 8.7 Hz, 2H), 3.24 (t, J = 8.7 Hz, 2H).
A solution of (E)-3-(2,3-dihydrobenzofuran-5-yl)acrylic acid (8.0 g, 42.1 mmol) in acetic acid (80 ml_) and water (1.0 ml_) was treated with 10% palladium on carbon (1 .0 g) in two portions. The reaction mixture was stirred under an atmosphere or hydrogen gas (balloon) until completion, typically 4 h. The mixture was diluted using ethyl acetate (100 mL) and filtered through a bed of celite washing through with further ethyl acetate. The solvents were removed in vacuo and the crude residue azeotroped using toluene (2 x 50 mL) to give an off white solid which was triturated using diethyl ether (50 mL) to give 3-(2,3- dihydrobenzofuran-5-yl)propanoic acid as a white solid (6.5 g, 80%). 1 H NMR (400 MHz, CDCI3) δ = 7.04 (s, 1 H), 6.93 (d, J= 8.4, 1 H), 6.7 (d, J= 8.4 Hz, 1 H), 4.55 (t, J = 8.4 Hz, 2H), 3.18 (t, J = 8.4 Hz, 2H), 2.89 (t, J= 7.6 Hz, 2H), 2.64 (t, J = 7.6 Hz, 2H).
A solution of 3-(2,3-dihydrobenzofuran-5-yl)propanoic acid (6.0 g, 31 mmol) in thionyl chloride (8 mL) was heated at 80 °C for 1 h. On completion of the reaction the thionyl chloride was removed in vacuo and the crude 3-(2,3- dihydrobenzofuran-5-yl)propanoyl chloride dissolved in anhydrous 1 ,2- dichloroethane (30 mL). In a separate flask aluminium trichloride (2 g, 15 mmol) was added to anhydrous 1 ,2-dichloroethane (40 mL) at 0 °C followed by the acid chloride solution (10 mL) drop-wise over 5 min and the resulting solution was stirred for 30 min at 0 °C. A further portion of aluminium trichloride (3 g, 22.5 mmol) was added followed by drop-wise addition of the remaining acid chloride solution (20 mL) at 0 °C. The reaction mixture was stirred at room temperature for 1 h or until completion, diluted with water and extracted using EtOAc (2 x 50 mL). The combined organic extracts were washed with 1 N HCI (50 mL), 1 N NaOH (50 mL), water (25 mL), brine (25 mL), dried (Na2SO4) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 10% EtOAc-hexanes eluent to give 2,3,5,6- tetrahydro-7H-indeno[5,6-b]furan-7-one as a white solid (3.8 g, 70%). 1 H NMR (300 MHz, CD3OD) δ = 7.36 (s, 1 H), 6.91 (s, 1 H), 4.61 (t, J = 8.6 Hz, 3H), 3.26 (t, J = 8.6 Hz, 2H), 3.05 (t, J = 5.5 Hz, 3H), 2.68 (t, J = 5.5 Hz, 2H).
2,3,5,6-tetrahydro-7H-indeno[5,6-b]furan-7-one (1 .5 g, 8.61 mmol) was dissolved in c.H2SO4 (6.0 mL) at 0 °C followed by drop-wise addition of f.HN03:c.H2S04, 1 : 1 (1 .2 mL) stirring was continued at 0 °C for 1 h. The reaction mixture was added to ice-cold water (60 mL) and stirred for 10 min, the resulting light brown ppt was removed by filtration, washed with ice cold water (20 mL) and dried in vacuo to give 8-nitro-2,3,5,6-tetrahydro-7H-indeno[5,6- b]furan-7-one (1.2 g, 64%). 1 H NMR (300 MHz, CD3OD) δ = 7.54 (s, 1 H), 4.80 (t, J = 8.6 Hz, 2H), 3.42 (t, J = 8.6 Hz, 2H), 3.09 (t, J = 5.6 Hz, 2H), 2.74 (t, J = 5.6 Hz, 2H).
A solution of 8-nitro-2,3,5,6-tetrahydro-7H-indeno[5,6-b]furan-7-one (1 .0 g, 4.57 mmol) in methanol (20 mL) 0 °C was treated with methane sulfonic acid (0.2 mL) followed by 20% palladium hydroxide (0.5 g). The reaction mixture was stirred under an atmosphere or hydrogen gas at 60 psi until completion. The reaction mixture was filtered through a bed of celite washing through with methanol (50 mL) and concentrated in vacuo. The residue was diluted with ethyl acetate (50 mL) and washed using sat. aq. NaHC03 (50 mL), water (20 mL), brine (20 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 10% EtOAc- hexanes eluent to give 3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-amine as a white solid (0.5 g, 63%). 1H NMR (300 MHz, CDCI3) δ = 6.54 (s, 1 H), 5.30 (s, 2H), 4.61 (t, J = 8.7 Hz, 2H), 3.21 (t, J = 8.7 Hz, 2H), 2.95 (t, J = 5.5 Hz, 2H), 2.66 (t, J = 5.5 Hz, 2H).
4-bromo-3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-amine
Figure imgf000153_0001
3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-amine (0.5 g, 2.86 mmol) and triethylamine (0.51 mL, 3.71 mmol) in dichloromethane (6.0 mL) at 0 °C was treated drop-wise with a solution of pivolyl chloride (0.41 g, 3.43 mmol) in DCM (4.0 mL). The reaction was stirred at ambient temperature for 6 h. The reaction mixture was added to sat. aq. NaHC03 (30 mL), and extracted using DCM (2 x 25 mL). The combined organics were washed with water (25 mL), brine (25 mL), dried (Na2S04) and concentrated in vacuo to give N-(3,5,6,7-tetrahydro- 2H-indeno[5,6-b]furan-8-yl)pivalamide as a white solid (0.55 g, 74%). 1H NMR (300 MHz, Chloroform-aO δ = 6.91 (s, 1 H), 4.56 (t, J = 8.6 Hz, 2H), 3.17 (t, J = 8.6 Hz, 2H), 2.83 (t, J= 7.4 Hz, 2H), 2.75 (t, J= 7.4 Hz, 2H), 2.04 (p, J= 7.4 Hz, 2H), 1 .32 (s, 9H).
N-(3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-yl)pivalamide (0.55 g, 2.12 mmol) in acetic acid (10 mL) was treated drop-wise with a solution of bromine (0.4 g, 2.55 mmol) in acetic acid (2.0 mL) and the reaction stirred at ambient temperature for 3 h. Ice cold water was added to the reaction mixture and stirred for 10 min. The resulting precipitate was removed by filtration, washed with water (20 mL) and dried in vacuo to give N-(4-bromo-3,5,6,7-tetrahydro- 2H-indeno[5,6-b]furan-8-yl)pivalamide as a pale-brown solid (0.65 g, 91 %). 1H NMR (300 MHz, Chloroform-aO δ = 6.94 (s, 1 H), 4.61 (t, J= 8.7 Hz, 2H), 3.18 (t, J = 8.7 Hz, 2H), 2.92 - 2.80 (m, 4H), 2.06 (p, J = 7.4 Hz, 2H), 1 .31 (s, 9H).
N-(4-bromo-3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-yl)pivalamide (0.6 g, 1 .78 mmol) in EtOH (10 mL) and cHCI (15 mL) was heated at 90 °C for 36 h. The solution was concentrated in vacuo then basified using aq NH4OH solution. The aqueous phase was extracted using ethyl acetate (2 x 20 mL) and the combined organics dried (Na2S04) and concentrated in vacuo to give 4-bromo- 3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-amine as a brown solid (0.3 g, 67%). 1 H NMR (400 MHz, Chloroform-aO δ = 4.61 (t, J = 8.6 Hz, 2H), 3.49 (s, 2H), 3.17 (t, J = 8.6 Hz, 2H), 2.84 (t, J = 7.4 Hz, 2H), 2.78 (t, J = 7.4 Hz, 2H), 2.12 (p, J = 7.4 Hz, 2H).
3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-4-amine
Figure imgf000155_0001
3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-amine (0.5 g, 1 .98 mmol) in ethanol (10 mL) and acetic acid (1 .5 mL) was treated with a solution of sodium nitrate (1.3 g, 19.8 mmol) in water (3.0 mL) and the reaction stirred at ambient temperature for 4 h. The ethanol was removed in vacuo then the residue diluted with water (30 mL), extracted using 10% IP A/chloroform (2 x 25 mL), dried (Na2S04) and concentrated in vacuo. . The crude product was purified by column chromatography on silica gel using 5% EtOAc-hexanes eluent to give 4- bromo-3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan as a yellow solid (0.28 g, 60%).
4-bromo-3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan (0.28 g, 1.18 mmol) in DMSO (10 mL) was treated with copper iodide (0.22 g, 1 .18 mmol), L-proline (0.21 g, 1 .88 mmol) and sodium azide (0.19 g, 2.94 mmol). The reaction mixture was heated in a sealed tube at 135 °C for 36 h. The reaction mixture was cooled, diluted with water and extracted using EtOAc (2 x 25 mL). The combined organic extracts were washed with water (25 mL), brine (25 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 10% EtOAc-hexanes eluent to give 3,5,6,7- tetrahydro-2H-indeno[5,6-b]furan-4-amine as a grey solid (0.17 g, 85%).1H NMR (300 MHz, Chloroform-a δ = 6.21 (s, 1 H), 4.59 (t, J= 8.5 Hz, 2H), 3.51 (s, 1 H), 2.98 (t, J = 8.5 Hz, 2H), 2.83 (t, J = 7.5 Hz, 2H), 2.64 (t, J = 7.5 Hz, 2H), 2.10 (p, J = 7.5 Hz, 2H). benzo[1 ,2-b:4,5-b']difuran-4-amine
Figure imgf000156_0001
2,3,6,7-tetrahydrobenzo[1 ,2-b:4,5-b']difuran-4-carboxylic acid (0.8 g, 3.88 mmol), 2, 3-dichloro-5,6-dicyanobenzoquinone (2.64 g, 1 1 .65 mmol) in anhydrous dioxane (20 mL) was heated in a sealed tube at 120 °C for 18 h. The reaction mixture was cooled to room temperature and sat. aq. Na2S203 (30 mL) added before extraction with ethyl acetate (2 x 25 mL). The combined organics dried (Na2S04) and concentrated in vacuo to give the crude benzo[1 ,2-b:4,5- b']difuran-4-carboxylic acid (1 .5 g). The crude acid (1 .5 g), triethylamine (2.05 mL) and diphenylphosphoryl azide (4.08 g, 14.85 mmol) in tertiary butanol (20 mL) was heated in a sealed tube at 90 °C for 12 h. The solution was cooled to room temperature, diluted with water (50 mL) and extracted using EtOAc (2 x 50 mL). The combined organic extracts were washed with water (25 mL), brine (25 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 10% EtOAc-hexanes eluent to give tert-butyl benzo[1 ,2-b:4,5-b']difuran-4-ylcarbamate (0.75 g) with minor impurities from the phosphine reagent, the product was dissolved in DCM (10 mL) and TFA (3.0 mL) added drop-wise over 5 min at 0 °C. The reaction was stirred at ambient temperature for 2 h then added carefully to sat. aq. NaHC03 (50 mL). The aqueous phase was extracted using DCM (2 x 30 mL) and the combined organic extracts were washed with water (25 mL), brine (25 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 10% EtOAc-hexanes eluent to give benzo[1 ,2-b:4,5-b']difuran-4-amine as an off-white solid (0.2 g, 30% over three steps). 1 H NMR (400 MHz, CDCI3): δ = 7.6 (d, J= 2.2 Hz, 1 H), 7.53 (d, J= 2.2 Hz, 1 H), 7.12 (s, 1 H), 6.78 (m, 2H), 4.17 (br.s., 1 H).
3-(3-(trifluoromethyl)-3H-diazirin-3-yl)aniline
Figure imgf000157_0001
Figure imgf000157_0002
To a solution of 2,2,2-trifluoro-1 -phenylethan-1 -one (5 g, 28.7 mmol) in C.H2SO4 (10 mL) at -5 °C was added a solution of c.H2S04 and f.HN03 (1 A , 16 mL) and the reaction mixture was stirred for 3 hours. The resulting solution was poured onto ice/water (100 mL) and extracted using ethyl acetate (2 x 100 mL). The combined organics were washed with water (100 mL), brine (100 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 20% EtOAc-hexanes eluent to give 2,2,2- trifluoro-1 -(3-nitrophenyl)ethan-1 -one as a yellow liquid (4.2 g , 67%). 1H NMR (400 MHz, CDCI3) δ = 8.92 (s, 1 H), 8.59 (dd, J = 8.1 , 1 .4 Hz, 1 H), 8.41 (d, J = 7.8 Hz, 1 H), 7.82 (t, J = 8.1 Hz, 1 H). 19F NMR (233.33 MHz, CDCI3): -71 .82 (s,
3F).
A solution of 2,2,2-trifluoro-1 -(3-nitrophenyl)ethan-1 -one (4.2 g, 19.2 mmol), hydroxylamine hydrochloride (4.0 g, 57.5 mmol) and pyridine (25 mL) in ethanol (25 mL) were heated at reflux for 3 h. or until completion. The solvent was removed in vacuo and the crude product was purified by column chromatography on silica gel using 40% EtOAc-hexanes eluent to give 2,2,2- trifluoro-1 -(3-nitrophenyl)ethan-1 -one oxime as a colourless liquid (4.0 g, 89%). 19F NMR (233.33 MHz, CDCI3): -66.42 and 62.28 (E and Z oxime).
To a solution of 2,2,2-trifluoro-1 -(3-nitrophenyl)ethan-1 -one oxime (4.0 g, 17.1 mmol) in dichloromethane (20 mL) was cooled to 0 °C treated with triethyiamine (1.5 eq), A/,A/-dimethylamine pyridine (0.5 eq), tosylchloride (1 .1 eq) and stirred at ambient temperature until completion, typically 16 h. The reaction mixture was diluted using dichioromethane (50 mL), washed with sat.aq. NH4CI (100 mL), water (100 mL), brine (100 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 5% EtOAc-hexanes eluent to give 2,2,2-trifluoro-1 -(3-nitrophenyl)ethan-1 - one O-tosyl oxime as a white solid (4.0 g, 60%). 1H N MR (300 MHz, CDCI3) 5 = 8.41 (ddd, J = 5.6, 3.5, 2.2 Hz, 1 H), 8.21 (t, J= 1 .5 Hz, 1 H), 7.89 (d, J = 8.3 Hz, 2H), 7.81 - 7.65 (m, 2H), 7.42 (d, J = 7.8 Hz, 2H), 2.50 (s, 3H). 19F NMR (282 MHz, cdcl3) 5 -61.55, -66.90.
A solution of 2,2,2-trifluoro-1 -(3-nitrophenyl)ethan-1 -one O-tosyl oxime (4.0 g, 10.3 mmol) in diethyl ether was cooled to -78 °C and a solution of ammonia gas was bubbled through for 30 min. The reaction mixture was sealed, allowed to warm to ambient temperature then stirred for 16 h. The mixture was filtered through a pad of celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 7% EtOAc-hexanes eluent to give 3-(3-nitrophenyl)-3-(trifluoromethyl)diaziridine as a colourless liquid (2.4 g, 100%). 1H NMR (300 MHz, CDCI3) δ = 8.52 (t, J = 2.0 Hz, 1 H), 8.33 (ddd, J = 8.3, 2.3, 1 .1 Hz, 1 H), 7.99 (d, J = 8.0 Hz, 1 H), 7.65 (tt, J = 7.8, 0.4 Hz, 1 H), 2.95 (d, J = 8.8 Hz, 1 H), 2.31 (d, J = 8.9 Hz, 1 H). 19F NMR (282 MHz, CDCI3) 5 = -75.10.
A solution of 3-(3-nitrophenyl)-3-(trifluoromethyl)diaziridine (2.4 g, 10.3 mmol) in methanol (30 mL) was treated with triethylamine (2 eq.) and iodine (1 eq.) and the reaction mixture stirred until completion, typically 2 h. The solution was diluted using diethyl ether, washed with 10% aq citric acid, water, aq. sodium thiosulfate, brine then dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 10% EtOAc- hexanes eluent to give 3-(3-nitrophenyl)-3-(trifluoromethyl)-3H-diazirine as a colourless liquid (2.1 g, 88%). 1 H NMR (300 MHz, CDCI3) δ 8.30 (ddd, J = 7.9, 2.2, 1 .4 Hz, 1 H), 8.09 - 8.01 (m, 1 H), 7.70 - 7.54 (m, 2H). 19F NMR (282 MHz, CDCI3) 5 = -65.14.
3-(3-nitrophenyl)-3-(tnfluoromethyl)-3H-diazirine (3.0 g, 13 mmol) in THF (70 mL) was treated with a solution of sodium dithionate (10 eq.) in water (30 mL) and the mixture was stirred at ambient temperature until completion, typically overnight. The solution was diluted using water, extracted using ethyl acetate (x2), washed with water, brine then dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 40% EtOAc-hexanes eluent to give the titled compound, 3-(3-(trifluoromethyl)- 3H-diazirin-3-yl)aniline, as a yellow solid (1 .5 g, 58%). 1H NMR (300 MHz, CDCIs) δ = 7.16 (t, J = 7.9 Hz, 1 H), 6.70 (ddd, J = 8.1 , 2.3, 0.9 Hz, 1 H), 6.52 (ddt, J= 7.9, 1.9, 0.9 Hz, 1 H), 6.45 br.(.s, 1 H), 3.77 (s, 2H). 19F NMR (282 MHz, CDCI3) 5 -65.07. -di-ieri-butylpyrimidin-2-amine
Figure imgf000159_0001
2,4,6-Trichloropyrimidine (2.7 g, 14.7 mmol) was dissolved in anhydrous THF (30 mL) at 0 °C under nitrogen atmosphere. Cul (280 mg, 1.47 mmol) was added to the aforementioned solution and subsequently treated with 2M tert- butylmagnesium chloride in THF (3.78 g, 16.15 mL, 32.3 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 3 h. Upon completion, the reaction mixture was diluted with saturated NH4CI solution and extracted with EtOAc (2 x 50 mL). The combined organic extract was washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 100% hexanes eluant to give 4,6-di-te/t-butyl-2-chloropyrimidine (1.3 g, 39%) as a pale brown liquid. 1 H NMR (300 MHz, CDCI3): δ = 7.20 (s, 1 H), 1 .33 (s, 18H). LCMS (m/z): 227.3 [M+H]+
In a 100 mL re-sealable reaction tube, a solution of 4,6-di-terf-butyl-2- chloropyrimidine (1.3 g) in EtOH (15 mL) was cooled to -50 °C. Ammonia gas was purged through the aforementioned solution for 15 min. The reaction mixture was warmed to 70 °C and stirred for 12 h. Upon completion, the reaction mixture was concentrated in vacuo and the residue obtained was diluted with water and extracted with ethyl acetate (50 mL). The organic extract was washed with water, brine, dried (Na2SO4) and concentrated in vacuo to give 4,6-di-teri-butylpyrimidin-2-amine (0.7 g, 59%) as a white solid. 1 H NMR (300 MHz, CDCIs): δ = 6.64 (s, 1 H), 4.83 (s, 2H), 1 .26 (s, 18H). LCMS (m/z): 208.4 [M+H]+
4-chloro-2,6-diisopropylaniline
Figure imgf000160_0001
2,6-Diisopropylaniline (5.0 g, 28.2 mmol) in DMF (100 mL) was treated with N- chlorosuccinimide (3.97 g, 29.7 mmol) and the reaction mixture stirred at room temperature overnight. The solution was poured onto water (500 mL) and extracted using diethyl ether (2 x 150 mL). The combined organics were washed with water (2 x 200 mL), brine (200 mL), dried (MgSO4) and concentrated in vacuo. The product was purified by short path distillation to give the titled compound as a red oil (3.0 g, 50%). 1H NMR (600 MHz, DMSO-d6): δ = 6.84 (s, 2H), 4.75 (s, 2H), 3.01 (hept, J = 6.8 Hz, 2H), 1.13 (d, J = 6.8 Hz, 12H). 13C NMR (151 MHz, DMSO-afe): δ = 141 .1 , 133.8, 122.5, 120.5, 27.2, 22.8.
4-chloro-2,6-dicyclopropylaniline
Figure imgf000161_0001
In a 50 ml_ re-sealable reaction tube, a solution of 2,6-dibromo-4-chloroaniline (0.25 g, 0.88 mmol) and cyclopropyl boronic acid (0.22 g, 2.62 mmol) along with K3PO4 (0.74 g, 3.50 mmol) was dissolved in toluene:water (10 ml_: 1 ml_). The resulting solution was degassed by purging with nitrogen gas for 5 minutes. Pd(OAc)2 (20mg, 0.087 mmol) and tricyclohexylphospine (25 mg, 0.087 mmol) were added and the solution was purged with nitrogen gas for another 5 minutes. The resulting mixture was stirred at 100 °C for 12 h. Upon completion of reaction the mixture was diluted with water (25 ml_), extracted with EtOAc (2 x 25 ml_) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 5% EtOAc-hexanes eluant to give 4-chloro-2,6-dicyclopropylaniline (150 mg, 83%) as brown liquid. 1H NMR (300 MHz, DMSO-afe): δ = 6.69 (s, 2H), 4.98 (s, 2H), 1 .74-1 .64 (m, 2H), 0.90- 0.84 (m, 4H), 0.52 -0.47 (m, 4H). LCMS (m/z): 208.30 [M+H]+.
Synthesis of 4-chloro-2-methyl-6-(trifluoromethyl)aniline
Figure imgf000161_0002
A solution of 2-methyl-6-(trifluoromethyl)aniline (0.4g, 2.20 mmol) in acetonitrile (4 ml_) and AcOH (0.3 ml_) was cooled to 0 °C . A/-Chlorosuccinimide (0.36g, 2.70 mmol) was added at 0 °C and the solution then allowed to warm to RT and stirred for 12 h. Upon completion of reaction the reaction mixture was diluted with ice cold water and the resulting precipitate removed by filtration and washed sequentially with saturated NaHC03, Na2S203 solution, n-pentane and dried in vacuo to give 4-chloro-2-methyl-6-(trifluoromethyl)aniline (0.25 g, 52%) as a white solid. 1 H NMR (400 MHz, CDCI3): δ = 7.30 (d, J = 2.4 Hz, 1 H), 7.18 (d, J = 2.0 Hz, 1 H), 2.17 (s, 3H). 19F NMR (400 MHz, CDCI3): δ = -63.03
4-chloro-2,6-diethylaniline
Figure imgf000162_0001
A solution of 2,6-dibromo-4-chloroaniline (0.5 g, 1 .75 mmol) and ethyl boronic acid (0.4 g, 5.25 mmol) in toluene (15 ml_) and water (4 ml_) was treated with K3PO4 (1.5 g, 7.0 mmol) at RT under argon atmosphere. Argon gas was used to purge the solution for 5 minutes before treating with Pd(OAc)2 (40 mg, 0.175 mmol) and tricyclohexyl phospine (50 mg, 0.175 mmol). The reaction mixture was again purged with argon for 5 minutes. The resulting mixture was stirred at 100 °C for 12 h. Upon completion the reaction mixture diluted with water extracted with EtOAc (2 x 25 ml_) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 8% EtOAc-hexanes eluant to give 4-chloro-2,6-dicyclopropylaniline (100 mg, 31 %) as yellow liquid. 1 H NMR (300 MHz, CDCI3): δ = 6.94 (s, 2H), 3.61 (s, 2H), 2.53 (q, J = 7.5 Hz, 4H), 1 .27 (t, J = 7.5 Hz, 6H). LCMS (m/z): 184.00 [M+H]+.
4-chloro-2,6-dimethoxyaniline
Figure imgf000162_0002
2,6-dibromo-4-chloroaniline (4 g, 14.0 mmol) was dissolved in 25% NaOMe soln in MeOH (48 mL) and treated with Cul (2.9g, 15.4 mmol) at RT under nitrogen atmosphere. The resulting mixture was stirred at 70 °C for 12 h under nitrogen atmosphere. Upon completion, the reaction mixture was cooled to RT and concentrated in vacuo. The residue obtained was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 50 mL) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 1 % EtOAc-hexanes to give 4-chloro-2,6-dimethoxyaniline (1.0 g, 38%) as a pale brown liquid. 1H NMR (300 MHz, CDCI3): δ = 6.52 (s, 2H), 3.83 (s, 6H). LCMS (m/z): 187.9 [M+H]+
2-amino-5-chloro-3-cyclopropyl-N,N-dimethylbenzamide
Figure imgf000163_0001
In a 50 mL resealable reaction tube, 2-amino-3-bromobenzoic acid (2.0 g, 9.25 mmol) was dissolved in DMF (20 mL) and cooled to 0 °C. EDC-HCI (2.1 g, 1 1.0 mmol), HOBt (1.49 g, 1 1 .0 mmol), DIPEA (2.8 mL, 27.7 mmol) and dimethylamine hydrochloride (1.13 g, 13.8 mmol) were sequentially added at 0 °C. The reaction mixture was warmed to 70 °C and stirred for 12 h. Upon completion the reaction mixture was diluted with water extracted with EtOAc (2 x 50 mL) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo to give 2-amino-3-bromo-N,N- dimethylbenzamide (2.0 g. 89%) as a white solid. 1H NMR (300 MHz, CDCI3): δ = 7.44 (dd, J = 7.8, 1 .5 Hz, 1 H), 7.06 (dd, J = 7.8, 1 .5 Hz, 1 H), 6.61 (t, J = 7.8 Hz, 1 H), 4.82 (bs, 2H), 3.05 (s, 6H). LCMS (m/z): 243.10, 245.10 [M+H]+.
In a 50 mL resealable reaction tube, a solution of 2-amino-3-bromo-A/,A/- dimethylbenzamide (2 g, 8.23 mmol), cyclopropyl boronic acid (850 mg, 9.87 mmol) and K3P04 (5.23 g, 24.06 mmol) were dissolved in toluene (30 ml_) and water (3 ml_). The solution was degassed by purging with nitrogen gas for 5 minutes then Pd(OAc)2 (184 mg, 0.823 mmol) and tricyclohexylphospine (230 mg, 0.823 mmol) were added and solution was once again purged with nitrogen gas for 5 minutes. The resulting mixture was stirred at 100 °C for 12 h. Upon completion the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 50 ml_) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 20% EtOAc-hexanes to give 2-amino-3-cyclopropyl-A/,A/-dimethylbenzamide (1.0 g, 60%) as light brown color solid. 1 H N MR (300 MHz, CDCI3): δ = 7.06 (d, J = 7.5 Hz, 1 H), 6.99 (dd, J = 7.8, 1 .5 Hz, 1 H), 6.66 (t, J = 7.8 Hz, 1 H), 4.70 (bs, 2H), 3.05 (s, 6H), 1 .68-1 .61 (m, 1 H), 0.94-0.88 (m, 2H), 0.62-0.57 (m, 2H). LCMS (m/z): 205.3 [M+H]+.
A solution of 2-amino-3-cyclopropyl-A/,A/-dimethylbenzamide (0.5g, 2.44 mmol) in acetonitrile (10 ml_) and AcOH (0.3 ml_) was cooled to 0 °C. N- Chlorosuccinimide (0.5 g, 3.67 mmol) was added at 0 °C and the resulting reaction mixture was warmed to RT and stirred for 12 h. Upon completion the reaction mixture was diluted with saturated Na2S203 solution and extracted with ethyl acetate (2 x 50 ml_). The combined organic extracts were washed with NaHC03 solution, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 25% EtOAc-hexanes eluant to give 2-amino-5-chloro-3-cyclopropyl-N,N- dimethylbenzamide (0.2 g, 34%) as a brown solid. 1H NMR (300 MHz, CDCI3): δ = 7.02 (d, J = 1.5 Hz, 1 H), 6.96 (d, J = 2.7 Hz, 1 H), 4.66 (bs, 2H), 3.05 (s, 6H), 1 .68-1 .61 (m, 1 H), 0.94-0.88 (m, 2H), 0.62-0.57 (m, 2H). LCMS (m/z): 239.0 [M+H]+.
4-chloro-2-methoxy-6-(trifluoromethyl)aniline
Figure imgf000165_0001
In a 50 ml_ resealable reaction tube, a solution of 2-bromo-4-chloro-6- (trifluoromethyl)aniline (0.5 g, 1.82 mmol), bis(pinacolato diborane) (0.92 g, 3.64 mmol) and KOAc (0.44 g, 4.55 mmol) in 1 ,4-dioxane (10 ml_) was degassed by purging with nitrogen gas for 5 minutes. Pd(dppf)Cl2 (015g, 0.182 mmol) was added and the solution purged again with nitrogen gas for 5 minutes. The resulting mixture was stirred at 1 10 °C for 12 h. Upon completion, the reaction mixture diluted with water, extracted with EtOAc (2 x 50 ml_) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 2% EtOAc-hexanes eoluant to give 4-chloro-2-(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)-6-(trifluoromethyl)aniline (0.5 g, 85%). LCMS (m/z): 324.10 [M+H]+.
4-Chloro-2-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-6- (trifluoromethyl)aniline (500 mg, 1 .55 mmol) was dissolved in THF (5 ml_) and H20 (2 ml_) at RT. NaB03.H20 (0.62 g, 6.23 mmol) was added portion-wise and the reaction stirred at RT for 4 h. Upon completion the reaction mixture was diluted with water extracted with EtOAc (2 x 50 ml_) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 15% EtOAc-hexanes eluant to give 2-amino-5-chloro-3- (trifluoromethyl)phenol (0.5 g, 100%) as a yellow liquid. 1H NMR (300 MHz, DMSO-afe): δ = 10.50 (s, 1 H), 6.85 (s, 2H), 5.12 (bs, 2H). 19F NMR (300 MHz, DMSO-afe): δ = -61.46. LCMS (m/z): 21 1 .6 [M+H]+.
2-amino-5-chloro-3-(trifluoromethyl)phenol (250 mg, 1 .18 mmol) was dissolved in anhydrous DMF (5 mL) and treated with K2CO3 (240 mg, 1 .77 mmol). The resulting mixture was stirred at RT for 30 minutes. Methyl iodide (185mg, 1.303 mmol) was added dropwise and the reaction stirred at RT for 2 h. Upon completion the reaction mixture was diluted with water extracted with EtOAc (2 x 50 mL) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 10% EtOAc-hexanes eluant to give 4-chloro-2-methoxy-6-(trifluoromethyl)aniline (0.2 g, 75%) as a pale brown solid. 1 H NMR (400 MHz, DMSO-a6): δ = 7.10 (d, J = 2.0 Hz, 1 H), 6.98 (d, J= 2.0 Hz, 1 H), 5.34 (bs, 2H) 3.85 (s, 3H). 19F NMR (400 MHz, DMSO-afe): δ = -61.45.
7-chloro-5-cyclopropyl-2,3-dihydro-1 H-inden-4-amine
Figure imgf000166_0001
A solution of 2,3-dihydro-1 H-inden-4-amine, 1 (500 mg, 3.75 mmol) in EtOH (5 mL) was cooled to 0 °C and treated dropwise with acetic anhydride (0.95 g, 9.37 mmol) under nitrogen atmosphere. The resulting reaction mixture was warmed to RT and stirred for 3h. Upon completion of reaction, (TLC, 30% ethyl acetate-hexanes, Rf, 0.2), the reaction mixture was concentrated in vacuo. The residue obtained was diluted with diethyl ether, filtered and dried in vacuo to give A/-(2,3-dihydro-1 /-/-inden-4-yl)acetamide (0.3 g, 45%) as a white solid. 1H NMR (400 MHz, DMSO-afe): δ = 9.29 (s, 1 H), 7.41 (d, J = 8.0 Hz, 1 H), 7.08 (t, J = 7.6 Hz, 1 H), 6.99 (d, J = 6.8 Hz, 1 H), 2.87 (t, J = 7.2 Hz, 2H), 2.80 (t, J = 7.2 Hz, 2H), 2.04 (s, 3H), 1 .99-1 .95 (m, 2H). LCMS (m/z): 176.40 [M+H]+
A/-(2,3-dihydro-1 /-/-inden-4-yl)acetamide (200 mg, 1 .1 1 mmol) was dissolved in AcOH (5 mL) and cooled to 0 °C. A/-Chlorosuccinimide (220 mg, 1 .69 mmol) was added then the reaction mixture was warmed to RT and strirred overnight. Upon completion the reaction mixture was diluted with ice cold water and the solid formed removed by filtration, washed saturated NaHC03, Na2S203 solution and dried in vacuo to give N-(7-chloro-2,3-dihydro-1 H-inden-4- yl)acetamide (0.12 g, 50%) as a white solid. 1H NMR (300 MHz, CDCI3): δ = 7.73 (d, J = 8.4 Hz, 1 H), 7.14 (d, J = 8.4 Hz, 1 H), 6.86 (s, 1 H), 3.02-2.85 (m, 4H), 2.19 (s, 3H), 1 .99 (m, 2H). LCMS (m/z): 209.80 [M+H]+
A/-(7-chloro-2,3-dihydro-1 /-/-inden-4-yl)acetamide (120 mg, 0.57 mmol) was dissolved in 3 M HCI (5 mL) and warmed to 90 °C for 4 h. Upon completion the reaction mixture was cooled to RT and basified (pH ~8) with saturated NaHC03 solution before extracting with EtOAc (2 x 20 mL). The combined organic extracts were washed with water, brine, dried (Na2S04) and concentrated in vacuo to give 7-chloro-2,3-dihydro-1 /-/-inden-4-amine (70 mg, 74%) as a white solid. 1 H NMR (300 MHz, DMSO-afe): δ = 6.85 (d, J = 8.4 Hz, 1 H), 6.40 (d, J = 8.4 Hz, 1 H), 4.97(s, 2H), 2.82 (t, J = 8.1 Hz, 2H), 2.71 (t, J = 7.5 Hz, 2H), 2.01 - 1 .96 (m, 2H). LCMS (m/z): 168.20 [M+H]+.
A solution of 7-chloro-2,3-dihydro-1 /-/-inden-4-amine (0.8g, 4.79 mmol) in acetonitrile (10 mL) was cooled to 0 °C and treated with A/-iodosuccinimide (1.61 g, 7.18 mmol) at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 12 h. Upon completion the reaction mixture was diluted with saturated Na2S203 solution and extracted with ethyl acetate (2 x 50 mL). The combined organic extract was washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatograpy on silica gel (60-120 mesh) using 4-5% EtOAc-hexanes eluant to give 7-chloro-5-iodo-2,3-dihydro-1 /-/-inden-4-amine (0.45 g, 32%) as a pale brown solid. 1 H NMR (300 MHz, DMSO-afe): δ = 7.36 (s, 1 H), 5.04 (s, 2H), 2.82- 2.72 (m, 4H), 2.03-1 .98 (m, 2H). LCMS (m/z): 293.7 [M+H]+.
In a 50 mL resealable reaction tube, a solution of 7-chloro-5-iodo-2,3-dihydro- 1 H-inden-4-amine , (0.35 g, 1.19 mmol) and cyclopropyl boronic acid (0.41 g, 4.77 mmol) in 1 ,4-dioxane (14 ml_) and water (4 ml_) was treated with CS2CO3 (1.16 g, 3.57 mmol) at RT under nitrogen atmosphere. Nitrogen gas was purged through the solution for 5 minutes and treated with Pd(OAc)2 (26mg, 0.1 19 mmol) and Catacxium-A (42 mg, 0.1 19 mmol) under nitrogen atmosphere. The resulting mixture was again degassed with nitrogen gas for another 5 minutes. The resulting mixture was stirred at 100 °C for 24 h. Upon completion the reaction mixture was diluted with water and extracted with EtOAc (2 x 25 ml_). The combined organic extract was washed with water, brine, dried (Na2S0 ) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 5% EtOAc-hexanes eluant to give 7-chloro-5-cyclopropyl-2,3-dihydro-1 /-/-inden-4-amine (70 mg, 28%) as a light brown solid. 1H NMR (300 MHz, CDCI3): 5 = 6.86 (s, 1 H), 2.94 (t, J = 7.5 Hz, 2H), 2.80 (t, J = 7.5 Hz, 2H), 2.15-2.10 (m, 2H), 1 .44-1 .43 (m, 1 H), 0.91 - 0.88 (m, 2H), 0.58-0.55 (m, 2H). LCMS (m/z): 208.3 [M+H]+.
Synthesis of R2 acid intermediates:
2,3,6,7-tetrahydrobenzo[1 ,2-b:4,5-b']difuran-4-carboxylic acid
Figure imgf000168_0001
Synthesis of 2,3,6, 7-tetrahydrobenzo[1 ,2-b:4,5-b']difuran-4-carboxylic acid was carried out from hydroquinone using procedures detailed by Monte et.al. J.Med.Chem. 1996, 39, 2953-2961 to give the 2,3,6,7-tetrahydrobenzo[1 ,2- b:4,5-b']difuran-4-carbaldehyde as a bright yellow solid; 1H NMR (400 MHz, CDCI3): δ = 10.27 (s, 1 H), 6.87 (s, 1 H), 4.67 (t, J = 8.8 Hz, 2H), 4.59 (t, J = 8.8 Hz, 2H), 4.59 (t, J = 8.8 Hz, 2H), 3.46 (t, J = 8.8 Hz, 2H), 3.18 (t, J = 8.8 Hz, 2H).
The aldehyde (0.68 g, 3.58 mmol) was oxidized using silver (I) oxide (1 .5 eq.) in 5% aqueous sodium hydroxide at rt for 20 days. The crude reaction mixture was filtered through celite, extracted using diethyl ether (2 x 50 mL) to remove unreacted aldehyde then the aqueous phase was acidified to pH 1 using 3.0 M aqueous HCI drop-wise at 0 °C. The product was extracted using dichloromethane (2 x 50 mL) and the combined organics washed using brine (50 mL), dried (MgS04) and concentrated in vacuo to give 2,3,6,7- tetrahydrobenzo[1 ,2-b:4,5-b']difuran-4-carboxylic acid as a white solid (0.44 g; 60%).
Alternatively, the aldehyde (0.5 g, 2.77 mmol) in acetone (5.0 mL) was treated with sulfamic acid (0.4 g, 4.17 mmol) in two portions at 0 °C. After 2 min a solution of sodium chlorite (0.32 g, 3.6 mmol) in water (1 .0 mL) was added drop-wise and stirring continued at 0 °C for 4 h. The reaction mixture was diluted with water (20 mL) and extracted using 10% IPA/chloroform (2 x 20 mL). The combined organics were washed with water (25 mL), brine (25 mL), dried (Na2S04) and concentrated in vacuo. The crude solid was triturated with diethyl ether to give 2,3,6,7-tetrahydrobenzo[1 ,2-b:4,5-b']difuran-4-carboxylic acid (0.4 g; 70%). 1H NMR (400 MHz, DMSO-d6): δ = 6.86 (s, 1 H), 4.52 (t, J = 8.8 Hz, 2H), 4.47 (t, J = 8.8 Hz, 2H), 3.30 (t, J = 8.8 Hz, 2H), 3.10 (t, J = 8.8 Hz, 2H). 13C (100 MHz, DMSO-de): δ = 166.4, 154.2, 153.9, 128.9, 127.2, 1 1 1 .4, 1 10.43, 71.9, 71 .6, 31 .5, 29.5.
Benzo[d][1 ,3]dioxole-4-carboxylic acid
C02H C02CH3 C02CH3 C02H Synthesised using procedures modified from Pie et.al. J.Med.Chem. 2004, 47, 871 -887 as follows:
2,3-Dihydroxybenzoic acid (5.0 g, 32.4 mmol) in anhydrous methanol (50 mL) was treated with concentrated sulfuric acid (10 drops) and heated at reflux overnight. The reaction mixture was concentrated in vacuo, diluted using EtOAc (100 mL) washed using sat. aqueous NaHC03 (2 x 50 mL), brine (50 mL) then dried (MgS04) and concentrated in vacuoio give methyl 2,3-dihydroxybenzoate (2.92 g; 54%). 1H NMR (400 MHz, CDCI3): δ = 10.9 (s, 1 H), 7.32 (dd, J = 8.0, 1 .2 Hz, 1 H), 7.09 (m, 1 H), 6.78 (t, .7 = 8.0 Hz, 1 H), 5.65 (s, 3H). 13C NMR (100 Hz, CDCIs) 170.7, 148.8, 145.0, 120.5, 1 19.8, 1 19.2, 1 12.4, 52.4.
Methyl 2,3-dihydroxybenzoate (1.0 g, 5.95 mmol) in DMF (16 mL) was treated with KF (1 .79 g, 30.9 mmol) and stirred at ambient temperature for 30 minutes. Diiodomethane (1 .79 g, 6.7 mmol) was added and the reaction heated at 100 °C for 5 hours. The reaction mixture was cooled to rt, poured onto water (100 mL) and extracted using diethyl ether (2 x 50 mL). The combined organics were washed with water (50 mL), brine(50 mL), dried (MgS04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 10% EtOAc-petroleum ether eluent to give methyl benzo[ ][1 ,3]dioxole-4- carboxylate as a white crystalline solid (0.56 g; 52%); 1 H NMR (400 MHz, CDCI3): δ = 7.41 (dd, J= 8.0, 1.2 Hz, 1 H), 6.97 (dd, J= 8.0, 1 .2 Hz, 1 H), 6.86 (t, J = 8.0 Hz, 1 H), 6.1 (s, 2H), 3.93 (s, 3H).
A solution of methyl benzo[ ][1 ,3]dioxole-4-carboxylate (0.4 g, 2.22 mmol) in methanol (8.0 mL) was treated with 2.0 M aqueous KOH (2.2 mL) and the solution stirred at rt for 3 hours. The mixture was concentrated to ~3 mL volume, diluted with water (5 mL) and acidified to pH ~3 using 2.0 M HCI. The resulting precipitate was removed by filtration, washed with water then diethyl ether and dried in vacuo to give benzo[ ][1 ,3]dioxole-4-carboxylic acid as a beige solid (0.38 g, 97%). 1H NMR (400 MHz, DMSO-d6): δ = 7.28 (dd, J= 8.0, 1.2 Hz, 1H),6.97 (dd, J=8.0, 1.2 Hz, 1H),6.89 (t, J=8.0Hz, 1 H), 6.12 (s, 2H); 13C NMR(100 Hz, DMSO-de) 165.5, 148.9, 148.5, 122.9, 121.6, 113.8, 112.5, 102.1.
Synthesised Compounds by Substituent Class
ALIPHATICS
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)cyclohexanesulfonamide
Figure imgf000171_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and cyclohexanesulfonamide were used in general method C3 to give the titled compound as a white solid (12 mg, 41%).1H NMR (400 MHz, CD3OD): δ = 6.97 (s, 1H), 3.50-3.43 (m, 1H), 2.87 (t, 4H, J= 8.0 Hz), 2.78 (t, 4H, J= 8.0 Hz), 2.22-2.18 (m, 2H), 2.10-2.02 (m, 4H), 1.94-1.71(m, 2H),1.63- 1.59 (m, 1H), 1.64-1.53 (m, 2H); 1.41-1.21 (m, 3H); 13C NMR (100 MHz, CD3OD): δ = 143.7, 137.8, 126.4, 118.4, 110.2, 59.9, 35.5, 30.0 , 28.5, 25.8, 25.1 , 24.8; LCMS Purity: >95%; LCMS (m/z): 363 [M +H]+ ; HRMS calculated for C19H26N2O3S [M+H]+:363.1737, found 363.1729.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)cyclopentanesulfonamide
Figure imgf000171_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and cyclopentanesulfonamide were used in general method C3 to give the titled compound as a white solid (26 mg, 42%) 1 H NMR (400 MHz, CD3OD): δ = 6.97 (s, 1 H), 4.08-4.02 (m, 1 H), 2.83 (t, J= 8.0 Hz, 4H), 2.80 (t, J = 8.0 Hz, 4H), 2.13-2.01 (m, 8H), 1 .84-1 .77 (m, 2H), 1 .71 -1 .65 (m, 2H); 13C NMR (100 MHz, CD3OD): δ = 145.1 , 139.2, 127.8, 1 19.8, 1 1 1 .7, 62.2, 33.9, 31.4, 29.9, 28.6, 26.9; LCMS Purity: >95%; LCMS (m/z): 349 [M +H]+; HRMS calculated for C18H24N2O3S: 349.1580, found 349.1588.
A/-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)tetrahydro-2H-pyran-4- sulfonamide.
Figure imgf000172_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and tetrahydro-2H-pyran-4-sulfonamide were used in general method C2 to give the titled compound as a white solid (12 mg, 57%). 1 H NMR (400 MHz, CD3OD): δ = 7.00 (s, 1 H), 4.09 (dd, J1 = 4 Hz, J2 = 12 Hz, 2H), 3.82- 3.76 (m, 1 H), 3.49-3.43, (m, 2H), 2.89 (t, J = 8 Hz, 4H), 2.81 (t, J = 8 Hz, 4H), 2.12-2.05 (m, 6H), 1 .98-1.87 (m, 2H); 13C NMR (100 MHz, CD3OD): δ = 154.0, 143.1 , 137.7, 126.5, 1 10.4, 66.0, 57.0, 32.5, 28.5, 25.9, 25.1 ; LCMS Purity: >95%; LCMS (m/z): 365 [M +H]+ ; HRMS calculated forCi8H24N204S , 365.1530, found 365.1541 .
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)tetrahydrofuran-3- sulfonamide
Figure imgf000173_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and tetrahydrofuran-3-sulfonamide were used in general method C2 to give the titled compound as a white solid (12 mg, 60%).1H NMR (400 MHz, DMSO-de): δ = 8.04 (s, 1 H), 6.93 (s, 1 H), 4.33-4.27 (m, 1 H), 4.04-4.00 (m, 1H),3.91-3.89 (m, 1H), 3.85-3.79 (m, 1H), 3.72-3.66 (m, 1H), 2.80 (t, J = 16.0 Hz, 4H), 2.70 (t, J = 16.0 Hz, 4H), 2.24-2.17 (m, 2H), 1.99 -1.95 (m, 4H).13C NMR (100 MHz, DMSO-d6): δ = 142.4, 139.6, 136.6, 124.7, 108.2, 68.7, 61.7, 32.5, 30.3, 28.8, 28.1, 24.9; LCMS Purity: >95%; LCMS (m/z): 351 [M +H]+; HRMS calculated for C17H22N2O4S 351.1373, found 351.1389.
FURANS
A-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)furan-2-sulfonamide
Figure imgf000173_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1) and furan-2-sulfonamide were used in general method C4 to give the titled compound as a white solid (75 mg, 16%).1H NMR (400 MHz, DMSO- d6): δ = 11.08 (br.s, 1H), 8.08 (s, 1H), 8.02 (s, 1H), 7.22 (q, J = 2.0 Hz, 1H), 6.94 (s, 1H), 6.71 (q, J =2.0 Hz, 1H), 2.78 (t, J =1.2 Hz, 4H), 2.59 (t, J =1.2 Hz, 4H), 1.94 (quin, J=7.2Hz, 4H).13C NMR (100 MHz, DMSO-d6): δ = 148.9, 147.9, 147.3, 143.1, 137.3, 128.7, 118.0, 117.5, 111.7, 54.9, 32.5, 30.1, 25.1. LCMS, Purity: 96.26%; m/z 345.1 (M-H+). HRMS (FAB") calcd for C17H-18N2O4S [Λ/-Η]" : 345.0987, found: 345.0866.
/V-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl )-5-methylfuran-2- sulfonamide
Figure imgf000174_0001
4- lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 5-methylfuran-2 -sulfonamide were used in general method C2 to give the titled compound as a white solid (28 mg, 53%). 1 H NMR (400 MHz, DMSO-d6): δ = 7.96 (s, 1 H), 7.00-6.99 (d, J = 4.0 Hz, 1 H), 6.91 (s, 1 H), 6.29- 6.28 (d, J = 4.0 Hz, 1 H), 1 H), 2.78 (t, J = 8.0 Hz, 4H), 2.61 (t, J = 8.0 Hz, 4H), 2.34 (s, 3H), 2 (t, J = 8.0 Hz, 4H), 1.98-1 .90 (m, 4H); 13C NMR (100 MHz, DMSO-d6): δ = 143.3, 137.6, 129.9, 125.2, 1 18.0, 1 14.6, 108.7, 108.2, 107.8, 32.9, 30.6, 25.4, 13.8; LCMS Purity: >95%; LCMS (m/z): 361 [M +H]+; HRMS calculated for C18H20N2O4S [M+H]+ 361 .1216, found 361 .1217.
5- ethyl-N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)furan-2- sulfonamide
Figure imgf000174_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 5-ethylfuran-2-sulfonamide were used in general method C2 to give the titled compound as a white solid (51 mg, 47%). 1 H NMR (400 MHz, DMSO-de): δ = 7.97 (bs, 1 H), 7.02 (s, 1 H), 6.91 (d, J= 4.0 Hz, 1 H), 6.31 (d, J = 4.0 Hz, 1 H), 2.78 (t, J= 8.0 Hz, 4H), 2.68 (q, J= 8.0 Hz, 2H), 2.59 (t, J= 6.0 Hz, 4H), 1 .97-1 .90 (m, 4H), 1 .19 (t, J= 8.0 Hz, 3H). 13C NMR (150 MHz, DMSO-d6): δ = 143.5, 143.3, 142.9, 137.6, 129.8, 1 18.0, 108.7, 106.8, 106.3, 32.9, 30.5, 25.4, 21 .3, 12.1 ; LCMS Purity: >95%; LCMS (m/z): 375 [M +H] +; HRMS calculated for C19H22N2O4S [M+H]+ 375.13730, found 375.13910.
5-((dimethylamino)methyl)-yV-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)furan-2-sulfonamide
Figure imgf000175_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 5-((dimethylamino)methyl)furan-2-sulfonamide were used in general method C2 to give the titled compound as a white solid (25 mg, 6 %).1H N MR (400 MHz, CD3OD) δ = 7.17 (d, J= 3.5 Hz, 1 H), 6.96 (s, 1 H), 6.86 (d, J = 3.5 Hz, 1 H), 4.43 (s, 2H), 2.86 (s, 3H),2.86 (t, J = 7.4 Hz, 4H), 2.73 (t, J = 7.4 Hz, 4H), 2.04 (p, J = 7.4 Hz, 4H). yV-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2-hydroxypropan-2- yl)furan-2-sulfonamide
Figure imgf000175_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 4-(2-hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C5 to give the titled compound as a white solid (2.5 g, 63%). 1H NMR (600 MHz, DMSO-d6): δ = 7.61 (br.s. , 1 H), 7.37 (d, J = 0.9 Hz, 1 H), 6.77 (s, 1 H), 6.58 (d, J = 0.9 Hz, 1 H), 2.74 (t, J = 7.3 Hz, 4H), 2.65 (t, J = 7.3 Hz, 4H), 1 .89 (tt, J= 7.3, 7.3 Hz, 4H), 1.34 (s, 6H). 13C NMR (101 Hz, DMSO-d6): δ = 157.4, 155.7, 142.2, 137.3, 136.7, 1 35.7, 132.4, 1 15.7, 109.3, 66.6, 32.6, 31.1 , 30.6, 25.1 . yV-((8-bromo-1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2- hydroxypropan-2-yl)furan-2-sulfonamide
Figure imgf000176_0001
4-Bromo-8-isocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 4-(2-hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C1 to give the titled compound as a white solid (40 mg, 7%). 1H NMR (400 MHz, CD3OD): δ = 7.68 (s, 1 H), 7.23 (s, 1 H), 2.91 (t, J= 7.6 Hz, 4H), 2.85 (t, J = 7.6 Hz, 4H), 2.1 1 (m, 4H), 1 .51 (s, 6H). LCMS (m/z): 482.9 [M-H]"; 97.64% (210 nm), 99% (254 nm). HPLC: 96.70% (210nm), 97.22% (254nm).
/V-((8-chloro-1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2- hydroxypropan-2-yl)furan-2-sulfonamide
Figure imgf000176_0002
4-Chloro-8-isocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 4-(2-hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C1 to give the titled compound as a white solid (50 mg, 16%). 1 H NMR (400 MHz, CD3OD) δ = 7.55 (s, 1 H), 7.02 (s, 1 H), 2.91 (t, J= 7.2 Hz, 4H), 2.85 (t, J = 7.2 Hz, 4H), 2.09 (m, 4H), 1 .5 (s, 6H). LCMS (m/z): 460.9 (M +Na)"; 95.16% (210 nm), 95.07% (254 nm). HPLC: 97.91 % (210 nm), 98.04% (254 nm).
4-(2-hydroxypropan-2-yl)-yV-((8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)furan-2-sulfonamide
Figure imgf000177_0001
4- lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 4-(2-hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C1 to give the titled compound as a white solid (15 mg, 3%). 1 H NMR (400 MHz, CD3OD): δ = 7.58 (s, 1 H), 7.07 (s, 1 H), 6.46 (s, 1 H), 2.82-2.73 (m, J = 7.5 Hz, 8H), 2.12 (s, 3H), 2.05-2.02 (m, 4H), 1.508 (s, 6H). LCMS( m/z): AM A O (M -1 )"; 99.59% (210 nm), 99.33% (254 nm). HPLC: 97.92% (210nm), 97.53% (254nm).
5- (N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl )sulfamoyl)furan-3- carboxylic acid
Figure imgf000177_0002
Ethyl 5-(N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)sulfamoyl)furan-3- carboxylate (0.1 g, 0.24 mmol) in THF (8 mL) at 0 °C was treated with a solution of LiOH (0.1 g, 2.4 mmol) in water (2 mL). The cooling bath was removed and the reaction mixture stirred for 3 h. The solution was acidified using 10% citric acid and immediately extracted using ethyl acetate (2 x 25 mL). The organics were washed using water (20 mL), brine (20 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by reversed phase HPLC to give the titled compound as a white solid (5.0 mg, 5%). 1H NMR (400 MHz, CD3OD): δ = 8.14 (s, 1 H), 7.28 (s, 1 H), 6.93 (s, 1 H), 2.85 (t, J = 7.6 Hz, 4H), 2.74 (t, J = 7.6 Hz, 4H), 2.04 (quin, J = 7.6 Hz, 4H).
Ethyl 5-(N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)furan-3-carboxylate
Figure imgf000178_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and ethyl 5-sulfamoylfuran-3-carboxylate were used in general method C3. The reaction mixture was quenched using water (50 ml_), extracted using ethyl acetate (2 x 25 ml_) and the organics washed with brine (25 ml_), dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 50% EtOAc-hexanes eluent to give the titled compound as a white solid (0.45 g, 63%). 1 H NMR (300 MHz, DMSO- d6) δ = 8.31 (s, 1 H), 7.59 (s, 1 H), 6.77 (s, 1 H), 4.22 (q, J= 7.2 Hz, 2H), 2.75 (t, J = 7.3 Hz, 4H), 2.65 (t, J = 7.3 Hz, 4H) 1 .90 (pent, J = 7.6 Hz, 4H), 1.26 (t, J = 7.2 Hz, 3H).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(prop-1-en-2- yl)furan-2-sulfonamide
Figure imgf000178_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 4-(prop-1 -en-2-yl)furan-2 -sulfonamide were used in general method C6 to give the titled compound as a white solid (85 mg, 51 %). 1 H NMR (400 MHz, CDCI3) δ 7.54 (s, 1 H), 7.28 (s, 1 H), 7.00 (s, 1 H), 5.26 (s, 1 H), 5.05 (s, 1 H), 2.86 (t, J = 7.4 Hz, 4H), 2.69 (t, J = 7.5 Hz, 4H), 2.09 - 1 .98 (m, 7H). 13C NMR (101 MHz, CDCI3) δ 144.4, 142.8, 137.8, 132.8, 129.2, 127.2, 1 19.4, 1 15.4, 1 13.6, 32.9, 30.5, 25.5, 20.9. HRMS (ESI) calcd. for C20H23N2O4S [M+H] 387.1373, found 387.1379.
4-(2-hydroxypropan-2-yl)-yV-((3,5,6,7-tetrahydro-2H-indeno[5,6-d]furan-8- yl)carbamoyl)furan-2-sulfonamide
Figure imgf000179_0001
8-isocyanato-3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan (prepared using general method A1 ) and ethyl 5-sulfamoylfuran-3-carboxylate were used in general method C3 to give ethyl 5-(N-((3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8- yl)carbamoyl)sulfamoyl)furan-3-carboxylate as a pale-brown solid (0.25 g, 50%). 1H NMR (300 MHz, DMSO-afe) δ 8.32 (s, 1 H), 7.17 (s, 1 H), 6.77 (d, J = 5.2 Hz, 1 H), 4.43 (t, J= 8.6 Hz, 2H), 4.23 (q, J= 7.1 Hz, 2H), 3.07 (t, J= 8.6 Hz, 2H), 2.71 (t, J = 7.3 Hz, 2H), 2.63 (t, J = 7.3 Hz, 2H), 1 .89 (p, J = 7.4 Hz, 2H), 1 .26 (t, J = 7.1 Hz, 3H).
Ethyl 5-(A/-((3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8- yl)carbamoyl)sulfamoyl)furan-3-carboxylate (0.25 g, 0.6 mmol) in anhydrous THF (10 mL) at 0 °C was treated with methyl magnesium chloride solution (3.0 M in Et20, 6 eq.) drop-wise over 5 minutes with vigorous stirring. The solution was then stirred at 0 °C for 30 min then at ambient temperature for 4 h before being quenched drop-wise with a solution of sat. ammonium chloride. The aqueous solution was extracted using EtOAc (2 x 25 mL), the combined organ ics washed with brine (20 mL), dried (Na2S04) and concentrated in vacuo. The crude product was triturated with diethyl ether then purified by reverse phase preparative HPLC to give the titled compound as a white solid (32 mg, 13%). 1H NMR (400 MHz, DMSO-afe) δ 7.58 (s, 1 H), 7.08 (s, 1 H), 6.84 (s, 1 H), 5.02 (s, 1 H), 4.47 (t, J= 8.6 Hz, 2H), 3.10 (t, J= 8.6 Hz, 2H), 2.73 (t, J= 7.3 Hz, 2H), 2.59 (t, J = 7.5 Hz, 2H), 1 .90 (d, J = 7.4 Hz, 2H), 1 .36 (s, 6H).
A^((4-bromo-3,5,6,7-tetrahydro-2H-indeno[5,6-d]furan-8-yl)carbamoyl)-4-(2- hydroxypropan-2-yl)furan-2-sulfonamide
Figure imgf000180_0001
4-bromo-8-isocyanato-3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan (prepared using general method A1 ) and 4-(prop-1 -en-2-yl)furan-2-sulfonamide were used in general method C1 to give the titled compound as a white solid (20 mg, 9%). 1H NMR (400 MHz, CD3OD) δ = 7.48 (d, J = 1.2 Hz, 1 H), 6.93 (d, J = 1 .2 Hz, 1 H), 4.60 (t, J = 8.7 Hz, 2H), 3.16 (t, J = 8.6 Hz, 2H), 2.85 (m , 4H), 2.03 (p, J = 7.5 Hz, 2H), 1.50 (s, 6H).
4-(2-hydroxypropan-2-yl)-yV-((3,5,6,7-tetrahydro-2H-indeno[5,6-d]furan-4- yl)carbamoyl)furan-2-sulfonamide
Figure imgf000180_0002
4-isocyanato-3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan (prepared using general method A1 ) and 4-(prop-1 -en-2-yl)furan-2 -sulfonamide were used in general method C1 to give the titled compound as a white solid (20 mg, 9%). 1 H NMR (400 MHz, CD3OD) δ = 7.58 (s, 1 H), 7.07 (s, 1 H), 6.46 (s, 1 H), 4.49 (d, J= 8.9 Hz, 2H), 3.05 (t, J = 8.7 Hz, 2H), 2.82 (t, J = 7.4 Hz, 2H), 2.70 (t, J = 7.4 Hz, 2H), 2.04 (p, J = 7.4 Hz, 2H), 1 .51 (d, J = 1 .9 Hz, 6H).
4-(2-hydroxypropan-2-yl)-A/-((2,3,6,7-tetrahydrobenzo[1 ,2-d:4,5-d']difuran-4- yl)carbamoyl)furan-2-sulfonamide
Figure imgf000181_0001
4-isocyanato-2,3,6,7-tetrahydrobenzo[1 ,2-£>:4,5-£>']difuran (prepared using general method A1 ) and 4-(2-hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C6 to give the titled compound as a white solid (285 mg, 96%). 1H NMR (600 MHz, DMSO-afe) δ 7.76 (s, 1 H), 7.66 (s, 1 H), 7.01 (s, 1 H), 6.45 (s, 1 H), 5.04 (s, 1 H), 4.46 (t, J= 8.6 Hz, 2H), 4.39 (t, J= 8.6 Hz, 2H), 3.08 (t, J = 8.6 Hz, 2H), 2.94 (t, J = 8.6 Hz, 2H), 1 .37 (s, 6H). yV-(benzo[1 ,2-d:4,5-d']difuran-4-ylcarbamoyl)-4-(2-hydroxypropan-2- yl)furan-2-sulfonamide
Figure imgf000181_0002
4-isocyanatobenzo[1 ,2-b:4,5-b']difuran (prepared using general method A1 ) and ethyl 5-sulfamoylfuran-3-carboxylate were used in general method C3 to give ethyl 5-(N-(benzo[1 ,2-b:4,5-b']difuran-4-ylcarbamoyl)sulfamoyl)furan-3- carboxylate as a white solid (0.05 g, 53%). 1H NMR (300 MHz, CD3OD) 5 = 8.25 (s, 1 H), 7.72 (d, J2.1 Hz, 1 H), 7.63 (d, J2.1 Hz, 1 H), 7.46 (s, 1 H), 7.27 (s, 1 H), 6.93 (s, 1 H), 6.89 (d, J 2.1 Hz, 1 H), 6.86 (d, J 2.1 Hz, 1 H), 4.30 (q, J 6.9 Hz, 2H), 1 .4 (t, J 6.9 Hz, 3H). Ethyl 5-(N-(benzo[1 ,2-b:4,5-b']difuran-4-ylcarbamoyl)sulfamoyl)furan-3- carboxylate (0.25 g, 0.6 mmol) in anhydrous THF (10 ml_) at 0 °C was treated with methyl magnesium chloride solution (3.0 M in Et20, 10 eq.) drop-wise over 10 minutes with vigorous stirring. The solution was then stirred at 0-10 °C for 3 h then quenched drop-wise with a solution of sat. ammonium chloride. The aqueous solution was extracted using EtOAc (2 x 20 ml_), the combined organ ics washed with brine (20 ml_), dried (Na2S04) and concentrated in vacuo. The crude product was triturated with diethyl ether then purified by reverse phase preparative HPLC to give the titled compound as a white solid (15 mg, 6%).1 H NMR (400 MHz, CD3OD) δ = 7.76 (d, J2.0 Hz, 1 H), 7.65 (d, J 2.4 Hz, 1 H), 7.55 (s, 1 H), 7.47 (s, 1 H), 7.05 (s, 1 H), 6.93 (d, J 2.0 Hz, 1 H), 6.89 (d, J 2.4 Hz, 1 H), 1 .5 (s, 6H). yV-(anthracen-9-ylcarbamoyl)-4-(2-hydroxypropan-2-yl)furan-2-sulfonamide
Figure imgf000182_0001
9-isocyanatoanthracene (prepared using general method B2) and 4-(2- hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C6 to give the titled compound as a white solid (24 mg, 23%). 1 H NMR (400 MHz, CD3OD) δ = 8.49 (s, 1 H), 8.07 - 7.98 (m, 4H), 7.75 (s, 1 H), 7.55 - 7.44 (m, 4H), 7.27 - 7.22 (m, 1 H), 1.49 (s, 6H). 13C NMR (101 MHz, CD3OD) δ = 153.8, 149.4, 141 .4, 136.6, 131 .7, 128.8, 128.2, 127.4, 126.4, 125.9, 124.9, 122.8, 1 15.2, 1 1 1 .1 , 67.2, 29.6.
4-(2-hydroxypropan-2-yl)-A-(quinolin-8-ylcarbamoyl)furan-2-sulfonamide
Figure imgf000183_0001
Phenyl chloroform ate (1 .5 eq) was added slowly to a solution of quinolin-8- amine (1 g, 6.9 mmol) in THF (10 mL) and triethylamine (2 eq.) to 0 °C. The solution was stirred at room temperature for 2 h or until completion. The solution was diluted using sat.aq. NaHC03 solution, extracted using ethyl acetate (2 x 50 mL), washed with water, brine then dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica using 10% EtOAc-hexanes to give phenyl quinolin-8- ylcarbamate (1.5 g, 83%) as a-white solid which was used directly in the next reaction step.
4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (0.2 g, 0.98 mmol) in THF (5 mL) at 0 °C was treated portion-wise with sodium hydride (3 eq.) and the suspension stirred at ambient temperature for 45 minutes (until effervescence ceased). The crude phenyl quinolin-8-ylcarbamate was dissolved in THF (5 mL) then added slowly to the reaction and the solution stirred at ambient temperature until completion, typically 4 h. The reaction was quenched with sat.aq. NH4CI, extracted with ethyl acetate (x2), washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified using reverse phase HPLC to give the titled compound, 4-(2-hydroxypropan-2-yl)-A/-(quinolin-8- ylcarbamoyl)furan-2-sulfonamide as a white solid (40 mg, 1 1 %). 1H NMR (400 MHz, DMSO-afe) δ 9.63 (s, 1 H), 8.89 (d, J = 4.3 Hz, 1 H), 8.37 (m, 2H), 7.80 - 6.76 (m, 5H), 5.09 (s, 1 H), 1 .38 (s, 6H).
4-(2-hydroxypropan-2-yl)-yV-((6-methoxyquinolin-8-yl)carbamoyl)furan-2- sulfonamide
Figure imgf000184_0001
4-(2-hydroxypropan-2-yl)-A/-((6-methoxyquinolin-8-yl)carbamoyl)furan-2- sulfonamide was synthesised using modification of the procedures used to make 4-(2-hydroxypropan-2-yl)-A/-(quinolin-8-ylcarbamoyl)furan-2-sulfonamide but using 6-methoxyquinolin-8-amine in place of quinolin-8-amine. The titled compound was obtained as an off-white solid (75 mg, 38%). 1H NMR (400 MHz, DMSO-afe) δ 8.79 (s, 1 H), 8.63 (m, 1 H), 8.17 (m, 1 H), 8.09 (d, J = 2.7 Hz, 1 H), 7.49 (dd, J = 8.3, 4.2 Hz, 1 H), 7.40 (s, 1 H), 6.79 (d, J = 2.8 Hz, 1 H), 6.69 (s, 1 H), 4.96 (s, 1 H), 3.84 (s, 3H), 1 .36 (s, 6H).
A^((2,3-dihydrobenzo[d][1 ,4]dioxin-5-yl)carbamoyl)-4-(2-hydroxypropan-2- yl)furan-2-sulfonamide
Figure imgf000184_0002
5-lsocyanato-2,3-dihydrobenzo[£>][1 ,4]dioxine (prepared using general method A1 ) and 4-(2-hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C6 to give the titled compound as a white solid (49 mg, 39%). 1 H NMR (600 MHz, Acetonitrile-ds) δ = 7.56 (dd, J= 8.4, 1 .5 Hz, 1 H), 7.45 (d, J= 1 .0 Hz, 1 H), 6.98 (d, J = 1 .0 Hz, 1 H), 6.7 (t, J = 8.4 Hz, 1 H), 6.48 (dd, J = 8.4, 1 .5 Hz, 1 H), 4.22 (m, 4H), 1 .43 (s, 6H).
A/-((2,3-dihydrobenzofuran-7-yl)carbamoyl)-4-(2-hydroxypropan-2-yl)furan- 2-sulfonamide
Figure imgf000185_0001
7-lsocyanato-2,3-dihydrobenzofuran (prepared using general method A1 ) and 4-(2-hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C6 to give the titled compound as a white solid (32 mg, 39%). 1 H NMR (600 MHz, Acetonitrile-tfs) δ 7.64 (d, J = 7.7 Hz, 1 H), 7.48 (d, J = 1.1 Hz, 1 H), 7.00 (d, J = 1 .1 Hz, 1 H), 6.89 (m, 1 H), 6.74 (t, J = 7.7 Hz, 1 H), 4.56 (t, J = 8.7 Hz, 1 H), 3.2 (t, J = 8.7 Hz, 1 H), 1 .43 (s, 6H). yV-((2,4-bis(trifluoromethyl)phenyl)carbamoyl)-4-(2-hydroxypropan-2- yl)furan-2-sulfonamide
Figure imgf000185_0002
1 -lsocyanato-2,4-bis(trifluoromethyl)benzene (prepared using general method A1 ) and 4-(2-hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C4 to give the titled compound as an off white solid (0.12 g, 33%). 1H NMR (400 MHz, DMSO-d6): δ = 8.59 (d, J = 8.8 Hz, 1 H), 7.87 (d, J = 9.2 Hz, 1 H), 7.81 (s, 1 H), 7.67 (s, 1 H), 7.43 (s, 1 H), 6.68 (s, 1 H), 4.94 (s, 1 H), 1 .36 (s, 6H). 13C NMR (100 MHz, DMSO-d6): δ = 156.0, 154.4, 142.5, 138.1 , 135.8, 129.9, 125.2, 124.9, 123.0, 122.5, 121 .3, 120.7, 120.4, 1 15.5, 1 15.2, 1 10.2, 66.5, 31 .0. LCMS, Purity: 90.47%, tr = 3.84 min, m/z 459.25 (M-H+). HRMS (FAB") calcd for CieHuFeNaOsS [Λ/-Η]" : 459.0528, found: 459.0512. yV-((2,5-bis(trifluoromethyl)phenyl)carbamoyl)-4-(2-hydroxypropan-2- yl)furan-2-sulfonamide
Figure imgf000186_0001
2-lsocyanato-1 ,4-bis(trifluoromethyl)benzene (prepared using general method A1 ) and 4-(2-hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C4 to give the titled compound as an off white solid (55 mg, 12%). 1H NMR (400 MHz, CD3OD): δ = 8.61 (s, 1 H), 7.75 (d, J = 7.6 Hz, 1 H), 7.48 (s, 1 H), 7.37 (d, J = 8.4 Hz, 1 H), 6.95 (s, 1 H), 1 .41 (s, 6H). 13C NMR (100 MHz, DMSO-d6): δ 156.4, 154.5, 139.7, 138.1 , 132.9, 127.2, 124.9, 122.3, 1 18.9, 1 17.6, 1 17.0, 1 10.0, 66.5, 31 .0. LCMS, Purity: 95.02%, tr = 2.09 min, m/z 558.94 (M-H+). HRMS (FAB") calcd for CieHuFeNaOsS [M- ]~ : 459.0528, found: 459.0224.
4-(2-hydroxypropan-2-yl)-yV-((2-methoxyphenyl)carbamoyl)furan-2- sulfonamide
Figure imgf000186_0002
1 -isocyanato-2-methoxybenzene (prepared using general method A2) and 4-(2- hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C2 to give the titled compound as an off white solid (30 mg, 38%). yV-((2,5-dimethoxyphenyl)carbamoyl)-4-(2-hydroxypropan-2-yl)furan-2- sulfonamide
Figure imgf000186_0003
2-isocyanato-1 ,4-dimethoxybenzene and 4-(2-hydroxypropan-2-yl)furan-2- sulfonamide were used in general method C2 to give the titled compound as an off white solid (52 mg, 55%).
N-((4-chloro-2,6-dimethylphenyl)carbamoyl)-4-(2-hydroxypropan-2- l)furan-2-sulfonamide
Figure imgf000187_0001
4-chloro-2,6-dimethylaniline, 1 (300 mg, 1 .92 mmol) was dissolved in THF (50 ml_) and cooled to 0 °C. NaH (100 mg, 2.49 mmol) was added in portions to the aforementioned solution under nitrogen atmosphere and stirred the mixture for 15 min. Phenyl chloroformate (0.33 ml_, 0.72 mmol) was added dropwise to the aforementioned solution at 0 °C. The reaction mixture was warmed to RT and stirred for 12 h. Upon completion, the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 30% EtOAc-hexanes eluant to give phenyl (4-chloro-2,6-dimethylphenyl)carbamate (0.2 g, 85%) as a white solid. 1 H NMR (300 MHz, CDCI3): δ = 7.41 -7.36 (m, 2H), 7.21 -7.19 (m, 2H), 7.1 1 -7.10 (m, 3H), 6.30 (bs, 1 H), 2.33 (s, 6H).
4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (133 mg, 0.64 mmol) was dissolved in anhydrous THF (5 ml_) and treated carefully with NaH (65 mg, 1 .63 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 45 min then treated with a solution of phenyl (4-chloro-2,6- dimethylphenyl)carbamate (200 mg, 0.73 mmol) in THF (3 ml_) under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 2 h. Upon completion, the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 50 ml_) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluant to give N-((4-chloro-2,6- dimethylphenyl)carbamoyl)-4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (20 mg, 31 %) as a white solid. 1 H NMR (400 MHz, CDCI3): δ = 7.83 (s, 1 H), 7.56 (s, 1 H), 7.19 (s, 1 H), 7.10-7.05 (m, 2H), 2.16 (s, 6H), 1 .55 (s, 6H). LCMS (m/z): 385.05 [M-H]", 94.12% (21 O nm). HPLC: 92.60% (21 Onm). HRMS calculated for Ci6H18Cli N205Si [M-H]" 385.0630, found 365.0621 .
A^((2,4-dimethyl-6-(trifluoromethyl) phenyl)carbamoyl)-4-(2- h droxypropan-2-yl)furan-2-sulfonamide
Figure imgf000188_0001
4-Chloro-2-methyl-6-(trifluoromethyl)aniline (230 mg, 1 .1 mmol) was dissolved in anhydrous THF (20 ml_) and treated with Et3N (0.17 ml_, 1 .32 mmol) at RT. The solution was treated with triphosgene (130 mg, 0.44 mmol) and the resulting mixture stirred at 60°C for 4 h then concentrated in vacuo. The residue obtained was stirred with n-pentane (20 ml_) for 10 min, filtered through a Celite pad and concentrated in vacuo to give 5-chloro-2-isocyanato-1 -methyl-3- (trifluoromethyl)benzene (0.2 g) as a white solid. The product was used in the next step without further purification.
4-(2-hydroxypropan-2-yl)furan-2-sulfonamide, 3 (150 mg, 0.731 mmol) was dissolved in anhydrous THF (50 ml_) and treated carefully with NaH (44 mg, 1 .096 mmol) at 0 °C under nitrogen atmosphere. The resulting reaction mixture was stirred at RT for 30 minutes and treated with solution of 5-chloro-2- isocyanato-1 -methyl-3-(trifluoromethyl)benzene (0.2 g) in THF (30 ml_) under nitrogen atmosphere. The resulting reaction mixture was stirred at RT for 2 h. Upon completion, the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 50 mL) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluent. The product was then triturated with diethyl ether and n-pentane to give A/-((2,4-dimethyl-6-(trifluoromethyl) phenyl)carbamoyl)-4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (15 mg, 5%) as a white solid. 1 H NMR (400 MHz, CD3OD): δ = 7.52-7.48 (m, 3H), 6.99 (s, 1 H), 2.19 (s, 3H), 1 .47 (s, 6H). 19F NMR (400 MHz, CD3OD): δ = -63.09. LCMS (m/z): 439.05 [M-H]"; 94.86% (210 nm), 96.92% (254 nm). HPLC: 98.90% (210nm). HRMS calculated for C16H15CI1 F3N2O5S1 [M-H]" 439.0348, found 439.0339.
yV-((4-chloro-2,6-diisopropylphenyl)carbamoyl)-4-(2-hydroxypropan-2- yl)furan-2-sulfonamid
Figure imgf000189_0001
5-chloro-2-isocyanato-1 ,3-diisopropylbenzene (prepared using general method A2) and 4-(2-hydroxypropan-2-yl)furan-2-sulfonamide were used in general method C2 to give the titled compound as a white solid (161 mg, 34%). 1H NMR (600 MHz, DMSO-afe) δ = 7.82 (s, 1 H), 7.61 (s, 1 H), 7.09 (s, 2H), 6.93 (s, 1 H), 5.04 (s, 1 H), 3.05 - 2.99 (m, 2H), 1 .35 (s, 6H), 1 .05 (d, J= 6.9 Hz, 12H). HRMS calculated for C20H26CI1 N2O5S1 [M-H]" 441 .1256, found 441 .1264.
/V-((4-chloro-2,6-dicyclopropylphenyl)carbamoyl)-4-(2-hydroxypropan-2- yl)furan-2-sulfonamide
Figure imgf000190_0001
4-Chloro-2,6-dicyclopropylaniline, 1 (250 mg, 1 .20 mmol) was dissolved in THF (50 mL) and cooled to 0 °C. NaH (72 mg, 1 .80 mmol) was added in portions and the resulting mixture stirred for 20 min under nitrogen atmosphere. Phenyl chloroformate (370 mg, 2.40 mmol) was added dropwise at 0 °C then the reaction mixture was warmed to RT and stirred for 12 h. Upon completion the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 8% EtOAc-hexanes eluant to give phenyl (4-chloro-2,6-dicyclopropyl phenyl)carbamate (0.2 g, 51 %) as a white solid. 1H NMR (300 MHz, CDCI3): δ = 7.38-7.35 (m, 2H), 7.21 -7.19 (m, 3H), 6.84-6.83 (m, 2H) , 2.06-2.04 (m, 2H), 1.04-1 .02 (m, 4H), 0.69-0.68 (m, 4H).
4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (75 mg, 0.365 mmol) was dissolved in anhydrous THF (50 mL) and treated carefully with NaH (36 mg, 0.914 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 30 min then treated with a solution of phenyl (4-chloro-2,6- dicyclopropyl phenyl)carbamate (135 mg, 0.402 mmol) in THF (3 mL) under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 4 h. Upon completion the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 50 mL) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 50-100% EtOAc-hexanes to give A/-((4-chloro-2,6- dicyclopropylphenyl)carbamoyl)-4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (10 mg, 6%) as a white solid. 1 H NMR (400 MHz, CD3OD): δ = 7.59 (s, 1 H), 7.13 (s, 1 H), 6.76 (s, 2H), 1 .88-1 .86 (m, 2H), 1.47 (s, 6H), 0.89-0.84 (m, 4H), 0.55-0.54 (m, 4H). LCMS (m/z): 436.95 [M-H]"; 96.29% (210 nm). HPLC: 98.29% (210nm). HRMS calculated for C20H22CI1 N2O5S1 [M-H]" 437.0943, found 437.0945. HRMS calculated for C20H22CI1 N2O5S1 [M-H]" 437.0943, found 437.0945.
4-(2-hydroxypropan-2-yl)-N-((5-methoxy-2,3-dihydro-1 H-inden-4- yl)carbamoyl)furan-2-sulfonamide
Figure imgf000191_0001
5-methoxy-2,3-dihydro-1 /-/-inden-4-amine (150 mg, 0.59 mmol) was dissolved in THF (15 mL) and cooled to 0 °C. NaH (35 mg, 0.89 mmol) was added to the aforementioned solution and stirred for 20 min. Phenyl chloroformate (150 mg, 0.932 mmol) was added dropwise at 0 °C then the solution allowed to warm to RT overnight. Upon completion, the reaction mixture was diluted with saturated NaHC03 and extracted with EtOAc (30 mL) The organic extract was washed with water, brine dried (Na2S04) and concentrated in vacuo to give phenyl (5- methoxy-2,3-dihydro-1 H-inden-4-yl)carbamate (100 mg, 59%) as a white solid. 1 H NMR (300 MHz, CDCI3): δ = 7.40-7.35 (m, 2H), 7.22-7.18 (m, 3H), 7.06 (d, J = 8.1 Hz, 1 H), 6.73 (d, J = 8.4 Hz, 1 H), 3.86 (s, 3H), 2.98-2.84 (m, 4H), 2.08 (t, J = 7.5Hz , 2H). LCMS (m/z): 284.3 [M+H]+
4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (87 mg, 0.424 mmol) was dissolved in anhydrous THF (5 mL) and treated carefully with NaH (44 mg, 1 .097 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 1 h then treated with a solution of phenyl (5-methoxy-2,3- dihydro-1 -/-inden-4-yl)carbamate (120 mg, 0.424 mmol) in THF (5 mL) under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 6 h. Upon completion, the reaction mixture was diluted with saturated NH4CI solution and extracted with EtOAc (2 x 30 ml_). The combined organic extract was washed with water, brine dried (Na2S04) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC [column: X bridge (150 mm x 19 mm particle size 5pm); flow: 15 mL/min; eluent: 10 mM ammonium bicarbonate in water (A) & MeCN (B); gradient: T/% B= 0/10, 2/10, 9/70]. The fractions were lyophilized to give 4-(2-hydroxypropan- 2-yl)-N-((5-methoxy-2,3-dihydro-1 H-inden-4-yl)carbamoyl) furan-2-sulfonamide (45 mg, 17%) as a white solid. 1 H NMR (400 MHz, CD3OD): δ = 7.67 (s, 1 H), 7.21 (s, 1 H), 7.04 (d, J = 8 Hz , 1 H), 6.77 (d, J = 8 Hz , 1 H), 3.79 (s, 3H), 2.84 (t, J = 7.2 Hz, 2H), 2.69 (t, J = 7.2 Hz, 2H), 2.01 -1 .97 (m, 2H), 1 .49 (s, 6H). LCMS (m/z): 393.10 [M-H]"; 98.97% (210 nm), 99.47% (254 nm). HPLC: 92.07% (210nm), 93.87% (254nm). HRMS calculated for Ci8H2iN206Si [M-H]+ 393.1 126, found 392.1 1 13.
N-((7-chloro-5-cyclopropyl-2,3-dihydro-1 H-inden-4-yl)carbamoyl)-4-(2- hydroxypropan-2-yl)furan-2-sulfonamide
Figure imgf000192_0001
7-chloro-5-cyclopropyl-2,3-dihydro-1 - -inden-4-amine, 6 (70mg, 0.33 mmol) was dissolved in THF (5 ml_) and cooled to 0 °C. NaH (20 mg, 0.505 mmol) was added to the aforementioned solution under nitrogen atmosphere and stirred for 15 min before phenyl chloroformate (100 mg, 0.674 mmol) was added dropwise at 0°C. The reaction mixture was warmed to RT and stirred for 12h. Upon completion the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using, 10% EtOAc-hexanes eluent to give phenyl (7-chloro-5-cyclopropyl-2,3-dihydro-1 - -inden-4-yl)carbamate (80 mg, 73%) as a brown solid. 1H NMR (300 MHz, CDCI3): δ = 7.39-7.37 (m, 3H), 7.25-7.24 (m, 2H), 6.85(s, 1 H), 3.0-2.94 (m, 4H), 2.12-2.10 (m, 2H), 1.34 (m, 1 H), 0.96-0.95 (m, 2H), 0.59-0.57 (m, 2H). LCMS (m/z): 328.30 [M+H]+.
4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (56 mg, 0.274 mmol) was dissolved in anhydrous THF (5 ml_) and treated carefully with NaH (27 mg, 0.685 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 15 min and was treated with a solution of phenyl (7-chloro-5- cyclopropyl-2,3-dihydro-1 /-/-inden-4-yl)carbamate (100 mg, 0.244 mmol) in THF (2 ml_) under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 3 h. Upon completion the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 30 ml_) and the combined organic extract washed with water, brine dried (Na2S04) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC [column: Gemini NX C18 (21.2 mm x 150 mm particle size 5pm); flow: 18 mL/min; eluent: 10 mM ammonium bicarbonate in water (A) & MeCN (B); gradient: T/% B= 0/20, 2/30, 10/50]. The fractions were lyophilized to give N-((7-chloro-5-cyclopropyl-2,3-dihydro-1 /-/-inden-4-yl)carbamoyl)-4-(2- hydroxypropan-2-yl)furan-2-sulfonamide (45 mg, 38%) as a white solid. 1H NMR (400 MHz, DMSO-afe): δ = 7.95 (d, J = 2.4 Hz, 1 H), 6.74 (s, 1 H), 6.69(d, J= 2.4 Hz, 1 H), 4.49 (m, 1 H), 2.84 (t, J= 7.6 Hz, 2H), 2.66 (t, J= 7.6 Hz, 2H), 1 .95 (m, 2H), 1 .78 (m, 1 H), 1 .41 (d, J = 6.4 Hz, 6H), 0.82 (m, 2H), 0.54 (m, 2H). LCMS (m/z): 437.0 [M-H]", 97.99% (210 nm). HPLC: 98.26% (210 nm). HRMS calculated for C20H22CI1 N2O5S1 [M-H]+ 437.0943, found 437.0927.
4-(2-hydroxypropan-2-yl)-N-((3-oxo-1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)furan-2-sulfonamide
Figure imgf000194_0001
Figure imgf000194_0002
A solution of 8-nitro-3,5,6,7-tetrahydro-s-indacen-1 (2H)-one (200 mg, 0.92 mmol) in MeOH (5 mL) was degassed with nitrogen for 5 minutes, 10% Pd/C (20 mg, 10% wt/wt) was added and the mixture stirred under hydrogen atmosphere at room temperature for 2 h. The reaction mixture was filtered through Celite and the filtrate was concentrated in vacuo to give 8-amino- 3,5,6,7-tetrahydro-s-indacen-1 (2H)-one as off an white solid (160 mg, 93%). 1 H NMR (600 MHz, DMSO-afe) δ 6.49 (s, 1 H), 6.34 (s, 2H), 2.90-2.84 (m, 2H), 2.80 (t, J = 7.5 Hz, 2H), 2.62 (t, J = 7.4 Hz, 2H), 2.56-2.51 (m, 2H), 2.04-1 .99 (m, 2H). 13C NMR (150 MHz, DMSO-afe): 206.6, 155.4, 153.7, 144.1 , 125.3, 1 18.6, 109.4, 36.8, 33.7, 28.6, 25.0, 24.9. LCMS (m/z): 188 [M+H]+. HRMS calculated for C2H14N10i[M+H]+ 188.1070, found 188.1077.
To di-f-butyldicarbonate (163 mg, 0.74 mmol) in anhydrous acetonitrile (1 mL) was added DMAP (26.1 mg, 0.21 mmol) at room temperature, stirred for 5 minutes, a solution of 8-amino-3,5,6,7-tetrahydro-s-indacen-1 (2H)-one (100 mg, 0.53 mmol) in acetonitrile was added. The reaction mixture was stirred for 30 minutes at room temperature. Reaction mixture was used directly in the next step without workup.
To 4-(2-hydroxypropan-2-yl) furan-2 -sulfonamide intermediate (100 mg, 0.48 mmol) in anhydrous THF (1 mL) was added NaH (18.3 mg, 0.48 mmol) at 0 °C and stirred for 30 minutes at ambient temperature under nitrogen atmosphere. Again cooled to 0 °C, 8-isocyanato-3,5,6,7-tetrahydro-s-indacen-1 (2H)-one (previous step reaction mixture) was added and stirred at ambient temperature for 16 h. To the reaction mixture added 0.5 mL of H20, loaded directly on C18 column for purification using aqueous 10 mM (NH4)HC03 solution and acetonitrile as mobile phase, to give 4-(2-hydroxypropan-2-yl)-N-((3-oxo- 1 ,2,3,5, 6,7-hexahydro-s-indacen-4-yl)carbamoyl) furan-2 -sulfonamide as a white solid (150 mg, 67%).1H N MR (600 MHz, DMSO-afe): δ 8.79 (s, 1 H), 7.37 (s, 1 H), 6.94 (s, 1 H), 6.61 (s, 1 H), 4.92 (s, 1 H), 2.92 (t, J = 5.6 Hz, 2H), 2.82 (t, J = 7.5 Hz, 2H), 2.75 (t, J = 7.5 Hz, 2H), 2.63-2.57 (m, 2H), 1 .97-1 .80 (m, 2H), 1 .34 (s, 6H); LCMS (m/z): 417 [M-H]-. HRMS calculated for C20H23N2O6S1 [M+H]+ 419.1271 , found 419.1291
To a solution of 4-(2-hydroxypropan-2-yl)-N-((3-oxo-1 ,2,3,5,6,7-hexahydro-s- indacen-4-yl)carbamoyl)furan-2-sulfonamide (70 mg, 0.16 mmol) in MeOH (2 mL) was added NaBH4 (63 mg, 1.67 mmol) at O °C under nitrogen atmosphere, resulting reaction mixture was stirred at room temperature for 3 h. Reaction mixture was quenched with H2O (2 mL, distilled out MeOH, aqueous layer was directly loaded on C18 column for purification using aqueous 10 mM (NH4)HCO3 solution and acetonitrile as mobile phase, to give N-((3-hydroxy- 1 , 2,3,5, 6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2-hydroxypropan-2- yl)furan-2-sulfonamide as off-white solid (60 mg, 86%). 1H NMR (600 MHz, DMSO-afe) δ 7.73 (bs, 1 H), 7.38 (s, 1 H), 6.81 (s, 1 H), 6.60 (s, 1 H), 5.63 (bs, 1 H), 4.92 (bs, 1 H), 4.87 (d, J = 6.0 Hz, 1 H), 3.00-2.84 (m, 2H), 2.77 (t, J = 7.4 Hz, 2H), 2.64-2.53 (m, 2H), 2.07-2.00 (m, 1 H), 1 .97-1 .92 (m, 1 H), 1 .91 -1.81 (m, 2H), 1 .35 (s, 6H); 13C NMR (150 MHz, DMSO-afe): 159.5, 156.0, 144.9, 143.3, 138.3, 137.7, 136.8, 136.0, 133.0, 72.6, 67.0, 35.4, 33.1 , 31.5, 31 .4, 31.0, 30.4, 25.5; LCMS (m/z): 419 [M-H]-; HRMS calculated for C20H23N2O6S1 [M-H]" 419.1282, found 419.1263. yV-((1-hydroxy-1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2- hydroxypropan-2-yl) furan-2-sulfonamide
Figure imgf000196_0001
A solution of 4-nitro-3,5,6,7-tetrahydro-s-indacen-1 (2H)-one (1 10 mg, 0.50 mmol) in MeOH (5 ml_) was degassed with nitrogen for 5 minutes, added 10% Pd/C (1 1 mg, 10% wt/wt), stirred under hydrogen atmosphere at room temperature for about 2 h. Reaction mixture was filtered through Celite pad, filtrate was concentrated to give 4-amino-3,5,6,7-tetrahydro-s-indacen-1 (2H)- one as an off white solid (75 mg, 80%). 1 H NMR (600 MHz, DMSO-afe) δ 6.72 (s, 1 H), 5.1 1 (s, 2H), 2.87 -2.74 (m, 4H), 2.70 (t, J = 7.4 Hz, 2H), 2.62-2.54 (m, 2H), 2.06-1 .99 (m, 2H); 13C NMR (150 MHz, DMSO-afe) δ 206.7, 144.6, 141 .7, 139.9, 136.9, 136.8, 104.2, 39.9, 36.7, 30.6, 30.5, 24.5, 23.7. LCMS (m/z): 188 [M+H]+; HRMS calculated for
Figure imgf000196_0002
[M+H]+ 188.1070, found 188.1074.
To di-t-butyldicarbonate (81 .6 mg, 0.37 mmol) in anhydrous acetonitrile (1 ml_) was added DMAP (13.0 mg, 0.04 mmol) at room temperature, stirred for 5 minutes, a solution of 4-amino-3,5,6,7-tetrahydro-s-indacen-1 (2H)-one (50 mg, 0.26 mmol) in acetonitrile (1 ml_) was added. The reaction mixture was stirred for 30 minutes at room temperature. Reaction mixture was used directly in the next step without workup. To 4-(2-hydroxypropan-2-yl) furan-2-sulfonamide intermediate (50 mg, 0.24 mmol) in anhydrous THF (1 mL) was added NaH (9.3 mg, 0.24 mmol) at 0 °C and stirred for 30 minutes at ambient temperature under nitrogen atmosphere. Again cooled to 0 °C, 4-isocyanato-3,5,6,7-tetrahydro-s-indacen-1 (2H)-one (previous step reaction mixture) was added and stirred at ambient temperature for 16 h. To the reaction mixture added 0.5 mL of H20, loaded directly on C18 column for purification using aqueous 10 mM (NH4)HC03 solution and acetonitrile as mobile phase, to give 4-(2-hydroxypropan-2-yl)-N-((1 -oxo- 1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl) carbamoyl) furan-2-sulfonamide (70 mg, 63%). 1 H N MR (600 MHz, DMSO-afe) δ 7.94 (s, 1 H), 7.38 (s, 1 H), 7.17 (s, 1 H), 6.60 (s, 1 H), 4.92 (s, 1 H), 2.94-2.89 (m, 2H), 2.85 (t, J = 7.4 Hz, 2H), 2.80 (t, J = 7.4 Hz, 2H), 2.56-2.52 (m, 2H), 2.00-1 .94 (m, 2H), 1.35 (s, 6H); LCMS (m/z): 417 [M-H]-; HRMS calculated for C20H21 N2O6S1 [M-H]" 417.1 126, found 417.1 1 13.
To a solution of 4-(2-hydroxypropan-2-yl)-N-((1 -oxo-1 ,2,3,5,6,7-hexahydro-s- indacen-4-yl)carbamoyl)furan-2-sulfonamide (50 mg, 0.1 1 mmol) in MeOH (2 mL) was added NaBH4 (45 mg, 1.19 mmol) at O °C under nitrogen atmosphere, resulting reaction mixture was stirred at room temperature for 3 h. Reaction mixture was quenched with H2O (1 mL), distilled out MeOH, aqueous layer was loaded directly on C18 column for purification using aqueous 10 mM (NH4)HCO3 solution and acetonitrile as mobile phase, to give N-((1 -hydroxy- 1 , 2,3,5, 6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2-hydroxypropan-2- yl)furan-2-sulfonamide (20 mg, 40%). 1H NMR (600 MHz, DMSO-tf6) δ 7.77 (s, 1 H), 7.51 (s, 1 H), 6.93 (s, 1 H), 6.79 (s, 1 H), 6.55 (s, 1 H), 5.05 (d, J = 5.8 Hz, 1 H), 4.99 (s, 1 H), 4.94 (q, J = 6.4 Hz, 1 H), 2.79 (t, J = 7.5 Hz, 2H), 2.70-2.61 (m, 3H), 2.52-2.49 (m, 1 H), 2.2-2.21 (m, 1 H), 1 .95-1 .90 (m, 2H), 1 .74-1 .59 (m, 1 H), 1 .36 (s, 6H); 13C NMR (150 MHz, DMSO-afe): 163.5, 146.0, 143.1 , 140.6, 138.5, 136.5, 136.2, 122.0, 1 16.5, 1 12.5, 108.5, 74.9, 67.0, 36.0, 33.0, 31 .5, 30.9, 27.8, 25.6; LCMS (m/z): 419 [M-H]-. HRMS calculated for C20H23N2O6S1 [M-H]" 419.1282, found 419.1265.
N-((4,6-d i met hyl pyr i m id i n-2-yl)carbamoyl)-4-(2-hydroxypropan-2-yl)f u ran-2- sulfonamide
Figure imgf000198_0001
A solution of 4,6-dimethylpyrimidin-2-amine (200 mg, 1.62 mmol) in THF (5 mL) was cooled to 0 °C and treated with NaH (130 mg, 3.24 mmol) under nitrogen atmosphere. The reaction mixture was stirred for 15 min and treated with phenyl chloroformate (380 mg, 2.43 mmol) at 0 °C under nitrogen atmosphere. The reaction mixture was warmed to RT and stirred for 12 h. Upon completion the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluant to give phenyl (4,6-dimethylpyrimidin-2-yl)carbamate (200 mg, 51 %) as a white solid. 1H NMR (300 MHz, CDCI3): δ = 7.91 (s, 1 H), 7.40-7.35 (m, 2H), 7.24-7.19 (m, 3H), 6.78(s, 1 H), 2.41 (s, 6H). LCMS (m/z): 244.20 [M+H]+.
4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (150 mg, 0.731 mmol) was dissolved in anhydrous THF (5 mL) and treated carefully with NaH (75 mg, 1 .83 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was warmed to 60 °C and stirred for 2 h. The solution was cooled to 0 °C and treated with a solution of phenyl (4,6-dimethylpyrimidin-2-yl)carbamate (195 mg, 0.804 mmol) in THF (5 mL) under nitrogen atmosphere at 0 °C. The reaction mixture was warmed to 50 °C for 4 h and then stirred at RT for 4 h. Upon completion the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 30 mL) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC [column: X-bridge (150 mm x 19 mm particle size 5pm); flow: 15 mL/min; eluent: 10 mM ammonium acetate in 0.1 % AcOH in water (A) & MeCN (B); gradient: 11% B= 0/15, 2/25, 8/40]. The fractions were lyophilized to give N-((4,6-dimethylpyrimidin-2-yl)carbamoyl)-4-(2- hydroxypropan-2-yl)furan-2-sulfonamide (25 mg, 7%) as a white solid. 1H NMR (400 MHz, CDCI3): δ = 8.18 (s, 1 H), 7.51 (d, J= 0.8 Hz, 1 H), 7.40 (d, J= 1 .2 Hz, 1 H), 6.78 (s, 1 H), 2.48 (s, 6H), 1 .57 (s, 6H). LCMS (m/z): 355.0 [M+H]+, 100% (210 nm), 100% (254 nm). HPLC: 96.49% (210 nm), 98.76% (254 nm). HRMS calculated for Ci4H17N 05Si [M-H]" 353.0925, found 353.0921 .
N-((4-cyclopropyl-6-methylpyrimidin-2-yl)carbamoyl)-4-(2-hydroxypropan- 2-yl)furan-2-sulfonamide
Figure imgf000199_0001
4-cyclopropyl-6-methylpyrimidin-2-amine (50mg, 0.33 mmol) was dissolved in THF (2 ml_) and cooled to O °C. NaH (16 mg, 0.40 mmol) was added carefully to aforementioned solution and stirred for 20 min. Phenyl chloroformate (80 mg, 0.503 mmol) was added dropwise at 0 °C. The reaction mixture was warmed to RT and stirred at RT for 12 h. Upon completion of reaction (TLC, 50% ethyl acetate-hexanes, Rf, 0.4), the reaction mixture was diluted with EtOAc and filtered through a celite pad. The filtrate was concentrated in vacuo and crude product was purified by column chromatography on silica gel (60-120 mesh) using 30% EtOAc-hexanes eluant to give phenyl (4-cyclopropyl-6- methylpyrimidin-2-yl)carbamate (40 mg, 44%) as an off white solid. 1 H NMR (300 MHz, CDCI3): δ = 8.06 (s, 1 H), 7.40-7.37 (m, 2H), 7.24-7.15 (m, 3H), 6.73 (s, 1 H), 2.4 (s, 3H), 1 .94-1 .86 (m, 1 H), 1 .15-1.1 (m, 2H), 1 .0-0.99 (m, 2H). LCMS (m/z): 270.3 [M+H]+
4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (150 mg, 0.731 mmol) was dissolved in anhydrous THF (5 mL) and treated carefully with NaH (73 mg, 1 .829 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 1 h and treated with a solution of phenyl (4-cyclopropyl-6- methylpyrimidin-2-yl)carbamate (190 mg, 0.731 mmol) in THF (5 mL) under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred at RT for 6 h. Upon completion, the reaction mixture was diluted with saturated NH4CI solution and extracted with EtOAc (2 x 30 mL). The combined organic extract was washed with water, brine dried (Na2S04) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC [column: X bridge (150 mm x 19 mm particle size 5pm); flow: 15 mL/min; eluent: 10 mM ammonium bicarbonate in water (A) & MeCN (B); gradient: T/% B= 0/15, 2/25, 8/40]. The fractions were lyophilized to give N-((4-cyclopropyl-6- methylpyrimidin-2-yl)carbamoyl)-4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (30 mg, 1 1 %) as a white solid. 1 H NMR (400 MHz, CD3OD): δ = 7.53 (s, 1 H), 7.07 (s, 1 H), 6.80 (s, 1 H), 2.35 (s, 3H), 1 .97-1 .93 (m, 1 H), 1 .42 (s, 6H), 1 .08- 1 .02 (m, 4H). LCMS (m/z): 381 .00 [M+H]+; 98.60% (210 nm), 99.49% (254 nm). HPLC: 98.05% (210nm), 99.01 % (254nm). HRMS calculated for Ci6H19N405Si [M-H]+ 379.1082, found 379.1082.
W-((4,6-di-ferf-butylpyrimidin-2-yl)carbamoyl)-4-(2-hydroxypropan-2- yl)furan-2-sulfonamide
Figure imgf000200_0001
4,6-di-tert-butylpyrimidin-2-amine (0.15 g, 0.72 mmol) was dissolved in THF (5 mL) and cooled to 0 °C. NaH (37 mg, 0.93 mmol) was added to aforementioned solution and resulting m ixture was stirred at 1 5 min under nitrogen atmosphere. Phenyl chloroformate (0.16 g, 1 .08 mmol) was added dropwise to the aforementioned solution at 0 °C. The reaction mixture was warmed to RT and stirred for 12 h. Upon completion of reaction (TLC, 10% ethyl acetate-hexanes, Rf, 0.5), the reaction mixture was concentrated in vacuo. The residue obtained was diluted with 10% IPA/CHCI3, filtered through a celite pad and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 4% EtOAc-hexanes eluant to give phenyl (4,6-di-terf- butylpyrimidin-2-yl)carbamate (0.1 g, 43%) as a white solid. 1 H NMR (300 MHz, CDCI3): δ = 7.65 (s, 1 H), 7.38 (m, 2H), 7.22 (m, 3H), 7.01 (s, 1 H), 1 .32 (s, 18H). LCMS (m/z): 327.80 [M+H]+
4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (65 mg, 0.305 mmol) was dissolved in anhydrous THF (8 mL) and treated carefully with NaH (30 mg, 0.764 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 45 minutes and treated with a solution of phenyl (4,6-di-terf- butylpyrimidin-2-yl)carbamate (100 mg, 0.305 mmol) in THF (5 mL) dropwise under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 3 h. Upon completion of reaction, (TLC, 50% ethyl acetate- hexanes, Rf, 0.5), the reaction m ixture was diluted with saturated NH4CI solution and extracted with EtOAc (2 x 20 mL). The combined organic extract was washed with water, brine dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluant to give A/-((4,6-di-te/t-butylpyrimidin-2- yl)carbamoyl)-4-(2-hydroxypropan-2-yl)furan-2-sulfonamide (0.07 g, 52%) as a white solid. 1 H NMR (400 MHz, DMSO-afe): δ =13.75 (s, 1 H), 10.71 (s, 1 H), 7.83 (s, 1 H), 7.37 (s, 1 H), 7.20 (s, 1 H), 5.17 (s, 1 H), 1 .39(s, 6H), 1 .31 (s, 18H). LCMS (m/z): 439.55 [M+H]+; 94.58% (210 nm), 97.94% (254 nm). HPLC: 98.51 % (210nm), 99.27% (254nm). HRMS calculated for C2oH29N405Si [M-H]+ 437.1864, found 437.1846. METHYL FURANS yV-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2-hydroxypropan-2- yl)-5-methylfuran-2-sulfonamide
Figure imgf000202_0001
4-isocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 4-(2-hydroxypropan-2-yl)-5-methylfuran-2-sulfonamide were used in general method C2 to give the titled compound as a white solid (52 mg, 51 %). yV-((2,6-diisopropylphenyl)carbamoyl)-4-(2-hydroxypropan-2-yl)-5- methylfuran-2-sulfonamide
Figure imgf000202_0002
2-lsocyanato-1 ,3-diisopropylbenzene (prepared using general method A1 ) and 4-(2-hydroxypropan-2-yl)-5-methylfuran-2-sulfonamide were used in general method C1 to give the titled compound as an off white solid (14 mg, 4%). 1H NMR (400 MHz, CD3OD): δ = 7.24 (t, J = 7.6 Hz, 1 H), 7.15 (d, J = 7.6 Hz, 2H), 7.04 (s, 1 H), 3.10 (sept., J= 6.8 Hz, 2H), 2.50 (s, 3H), 1.50 (s, 6H), 1.17 (d, J = 6.8 Hz, 12H).
DEUTERATED FURANS yV-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2-hydroxypropan-2- yl-1 ,1 ,1 ,3,3,3-d6)furan-2-sulfonamide
Figure imgf000203_0001
Λ/-((1 , 2,3,5, 67-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2-hydroxypropan-2- yl)furan-2-sulfonamide can be synthesized using 4-isocyanato-1 ,2,3,5,6,7- hexahydro-s-indacene (prepared using general method A1 ) and /6-4-(2- hydroxypropan-2-yl)-5-methylfuran-2-sulfonamide in general method C1.
Alternatively, ethyl 2-methyl-5-sulfamoylfuran-3-carboxylate (0.4 g, 0.96 mmol) in anhydrous THF (30 mL) at -10 °C was treated with c/3-methyl magnesium iodide solution (1.0 M in Et20, 10 eq.) drop-wise over 10 minutes with vigorous stirring. The solution was then stirred at ambient temperature for 12 h then cooled to 0 °C and quenched drop-wise with a solution of sat. ammonium chloride. The aqueous solution was extracted using EtOAc (2 x 20 mL), the combined organics washed with brine (20 mL), dried (Na2S04) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC to give the titled compound as a white solid (5 mg, 1 %). 1H NMR (300 MHz, CD3OD): δ = 7.50 (d, J = 1.2 Hz, 1 H), 6.95 (d, J = 1 .2 Hz, 1 H), 6.89 (s, 1 H), 2.83 (t, J = 7.2 Hz, 4H), 2.75 (t, J= 7.2 Hz, 4H), 2.02 (quin, J= 7.2 Hz, 4H). yV-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2-hydroxypropan-2- yl-1 ,1 ,1 ,3,3,3-d6)-5-methylfuran-2-sulfonamide
Figure imgf000203_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and /6-4-(2-hydroxypropan-2-yl)-5-methylfuran-2 -sulfonamide were used in general method C1 to give the titled com pound as a white solid (10 mg, 3%).1 H NMR (400 MHz, CD3OD) δ = 7.03 (s, 1 H), 6.95 (s, 1 H), 2.86 (t, J= 7.4 Hz, 4H), 2.73 (t, J = 7.4 Hz, 4H), 2.48 (s, 3H), 2.04 (p, J = 7.4 Hz, 4H).
4-(2-hydroxypropan-2-yl-1 ,1 ,1 ,3,3,3-d6)-5-methyl-N-((3,5,6,7-tetrahydro-2H- indeno[5,6- ?]furan-4-yl)carbamoyl)furan-2-sulfonamide
Figure imgf000204_0001
4-isocyanato-3,5,6,7-tetrahydro-2H-indeno[5,6-o]furan (prepared using general method A1 ) and /6-4-(2-hydroxypropan-2-yl)-5-methylfuran-2 -sulfonamide were used in general method C1 to give the titled compound as a white solid (20 mg, 5%). 1H NMR (400 MHz, CD3OD) δ = 7.13 (s, 1 H), 6.52 (s, 1 H), 4.51 (t, J = 8.6 Hz, 2H), 3.03 (t, J = 8.6 Hz, 2H), 2.84 (t, J = 7.4 Hz, 2H), 2.68 (t, J = 7.4 Hz, 2H), 2.50 (s, 3H), 2.05 (p, J = 7.4 Hz, 2H).
AA((4-bromo-3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan-8-yl)carbamoyl)-4-(2- hydroxypropan-2-yl-1 ,1 ,1 ,3,3,3-d6)-5-methylfuran-2-sulfonamide
Figure imgf000204_0002
4-Bromo-8-isocyanato-3,5,6,7-tetrahydro-2H-indeno[5,6-b]furan (prepared using general method A1 ) and /6-4-(2-hydroxypropan-2-yl)-5-methylfuran-2- sulfonamide were used in general method C1 to give the titled compound as a white solid (32 mg, 39%). 1 H NMR (400 MHz, DMSO-afe) δ = 8.01 (s, 1 H), 7.07 (s, 1 H), 5.04 (s, 1 H), 4.59 (t, J= 8.7 Hz, 2H), 3.14 (t, J= 8.7 Hz, 2H), 2.77 (t, J = 7.4 Hz, 2H), 2.69 (t, J = 7.4 Hz, 2H), 2.43 (s, 3H), 1 .98 (q, J = 7.4 Hz, 2H). THIOPHENES
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)thiophene-2- sulfonamide
Figure imgf000205_0001
4-lsocyanato-1,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and thiophene-2 -sulfonamide were used in general method C2 to give the titled compound as a white solid (11 mg, 11%).1H NMR (400 MHz, CD3OD): δ = 7.79 (d, J=4.0 Hz, 1H), 7.76 (d, J=4.0 Hz, 1H), 7.73 (t, J=4.0 Hz, 1H), 6.93 (s, 1H), 2.83 (t, J = 12 Hz, 4H), 2.66 (t, J= 12 Hz, 4H), 2.04-1.96 (m, 4H).13C NMR (100 MHz, CD3OD): δ 143.5, 143.2, 137.8, 132.7, 132.2, 126.6, 126.4, 118.2, 110.3, 32.5, 29.9, 25.1; LCMS Purity: >95%; LCMS (m/z): 363 [M +H]+; HRMS calculated for C17H18N2O3S2 (M+H)+ 363.0832, found 363.0819.
A^((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-5-methylthiophene-2- sulfonamide
Figure imgf000205_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) 5-methylthiophene-2-sulfonamide were used in general method C2 to give the titled compound as a white solid (12 mg, 18%).1 H NMR (400 MHz, DMSO-afe): δ = 7.88 (s, 1 H), 7.43 (d, J = 4.0 Hz, 1 H), 6.89 (s, 1 H), 6.82 (d, J = 4.0 Hz, 1H), 2.78 (t, J= 12 Hz, 4H), 2.61 (t, J= 12 Hz, 4H), 2.47 (s, 3H), 1.97- 1 .89 (m, 4H). 13C NMR (100 MHz, DMSO-afe): δ = 143.2, 142.9, 137.3, 130.6, 126.0, 125.6, 125.2, 1 17.5, 108.7, 32.69, 30.7, 25.5, 15.4; LCMS Purity: >95%; LCMS (m/z): 377 [M +H]+; HRMS calculated for C18H20N2O3S2, (M+H)+ 377.0988, found 377.0994.
THIAZOLES
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl )thiazole-2- sulfonamide
Figure imgf000206_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and thiazole-2 -sulfonamide were used in general method C2 to give the titled compound as a white solid (8 mg, 20%). 1 H NMR (400 MHz, CD3OD): δ = 7.02 (d, 1 H, J = 4.0 Hz), 6.99 (s, 1 H), 6.60 (d, 1 H, J = 4.0 Hz), 2.88 (t, 4H, J = 8.0 Hz), 2.76 (t, 4H, J = 8.0 Hz), 2.08-2.02 (m, 4H); 13C NMR (100 MHz, CD3OD): δ = 169.6, 144.2, 144.1 , 137.7, 137.5, 132.4, 1 18.1 , 106.9, 32.4, 29.9, 25.2.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-2-methylthiazole-5- sulfonamide
Figure imgf000206_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 2-methylthiazole-5-sulfonamide were used in general method C3 to give the titled compound as a white solid (35 mg, 65%); 1H NMR (600 MHz, DMSO-afe) 5 = 7.73 (s, 1 H), 7.53 (s, 1 H), 6.78 (s, 1 H), 2.75 (t, J = 7.4 Hz, 4H), 2.66 (t, J = 7.4 Hz, 4H), 2.59 (s, 3H), 1 .93-1 .88 (m, 4H); 13C NMR (150 MHz, DMSO-afe): 166.8, 158.3, 143.9, 142.0, 141 .4, 136.6, 132.3, 1 15.5, 32.5, 30.4, 25.0, 18.6; LCMS Purity: >95%; LCMS (m/z): 378 [M+H]+; HRMS calculated for
Figure imgf000207_0001
[M-H]+ 376.0795, found 376.0791 .
TRIAZOLES
1 -benzyl-N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 Η-Λ ,2,4- triazole-3-sulfonamide
Figure imgf000207_0002
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -benzyl-1 HA ,2,4-triazole-3-sulfonamide were used in general method C3 to give the titled compound as a white solid (40 mg, 15%) 1 H NMR (400 MHz, DMSO-d6): δ = 8.9 (s, 1 H), 8.0 (s, 1 H), 7.35-7.28 (m, 5H), 6.90 (s, 1 H), 5.48 (s, 2H), 2.77 (t, J = 7.2 Hz, 4H), 2.59 (t, J = 7.2 Hz, 4H), 1 .95- 1 .90 (m, 4H). LCMS (m/z): 438.10 (M +1 )+ 95.84% (210 nm), 97.84% (254 nm). HPLC: 95.99% (210nm), 95.31 % (254nm).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4H-1 ,2,4-triazole-3- sulfonamide
Figure imgf000208_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 4H-1 ,2,4-triazole-3-sulfonamide were used in general method C3 to give the titled compound as a white solid (31 mg, 62%); 1H NMR (600 MHz, DMSO-afe) δ = 9.83 (bs, 1 H), 7.99 (s, 1 H), 7.62 (s, 1 H), 6.77 (s, 1 H), 2.73 (t, J = 7.4 Hz, 4H), 2.64 (t, J = 7.4 Hz, 4H), 1 .91 -1 .86 (m, 4H); 13C NMR (150 MHz, DMSO-afe): 159.2, 149.8, 148.7, 142.5, 137.2, 132.8, 1 16.1 , 33.0, 30.9, 25.6; LCMS Purity: >95%; LCMS (m/z): 348 [M+H]+; HRMS calculated for C15H16N5O3S1 [M-H]" 346.0979, found 346.0983.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 -isopropyl-1 Η-Λ ,2,3- triazole-4-sulfonamide
Figure imgf000208_0002
1 ,2,3,5, 6,7-hexahydro-s-indacen-4-amine, 7 (100 mg, 0.578 mmol) was dissolved in anhydrous THF (5 mL) and treated with Et3N (70 mg, 0.693 mmol) at RT. The solution was treated with triphosgene (70 mg, 0.231 mmol) and resulting mixture was stirred at 70 °C for 3h. The reaction mixture was concentrated in vacuo. The residue obtained was stirred with n-pentane (20 mL) for 10 min and filtered through celite. The filtrate was concentrated in vacuo to give isocyanate as a white solid. In another 50 mL round bottom flask, 1 -isopropyl-1 H-1 ,2,3-triazole-4-sulfonamide (95 mg, 0.50 mmol) was dissolved in anhydrous THF (5 mL) and treated carefully with NaH (42 mg, 1 .05 mmol) at 0 °C under nitrogen. It was stirred at RT for 45 minutes and treated with aforementioned solution of isocyanate in THF under nitrogen. The resulting reaction mixture was stirred at RT for 5 h. Upon completion (TLC, 70% ethyl acetate-hexanes, Rf, 0.3), the reaction mixture was diluted with saturated NH4CI solution and extracted with EtOAc (2 x 25 mL). The combined organic extract was washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC [column: Gemini NX C18 (21 .5 mm x 150 mm particle size 5pm); flow: 15 mL/min; eluent: 10 mM ammonium bicarbonate in water (A) & MeCN (B); gradient: T/% B= 0/10, 2/20, 8/65]. The fractions were lyophilized to give N-((1 , 2, 3,5,6,7- hexahydro-s-indacen-4-yl)carbamoyl)-1 -isopropyl-1 HA ,2,3-triazole-4- sulfonamide (25 mg, 12%), as a white solid. 1H NMR (400 MHz, DMSO-afe): δ = 8.75 (s, 1 H), 7.91 (s, 1 H), 6.89 (s, 1 H), 4.9 (m, 1 H), 2.79 (t, J = 1.2 Hz, 4H), 2.60 (t, J= 7.2 Hz, 4H), 1 .96-1 .89 (m, 4H), 1 .5 (d, J =6.8 Hz , 6H). LCMS (m/z): 390.10 [M+H]+ ; 100% (210 nm) ,100% (254 nm). HPLC: 96.05% (210nm), 96.13% (254nm). HRMS calculated for C18H22N5O3S1 [M-H]" 388.1449, found 388.1457.
PYRAZOLES
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 -methyl-1 H-pyrazole- 5-sulfonamide
Figure imgf000209_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 1 -methyl-1 H-pyrazole-5-sulfonamide were used in general method C2 to give the titled compound as a white solid (8 mg) 20%. 1H NMR (400 MHz, DMSO-d6): δ = 7.95 (bs, 1 H), 7.45 (s, 1 H), 6.88 (s, 1 H), 6.66 (s, 1 H), 4.02 (s, 3H), 2.77 (t, J = 16 Hz, 4H), 2.60 (t, J = 16 Hz, 4H), 1 .96-1 .88 (m, 4H). 'X NMR (150 MHz, DMSO-tf6): δ = 143.1 , 142.9, 137.2, 125.2, 1 17.4, 1 10.0, 109.0, 108.7, 38.6, 33.0, 30.7, 25.5; LCMS Purity: >95%; LCMS (m/z): 361 [M +H] +; HRMS calculated for C17H20N4O3S (M+H) + 361 .13289, found 361 .13213.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl )-1-methyl-1 H- pyrazole-3-sulfonamide
Figure imgf000210_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -methyl-1 H-pyrazole-3-sulfonamide were used in general method C1 to give the titled compound as a white solid (40 mg, 8%). 1H NMR (400 MHz, DMSO-d6): δ = 10.8 (brs, 1 H), 8.02 (s, 1 H), 7.86 (s, 1 H), 6.92 (s, 1 H), 6.69 (s, 1 H), 3.91 (s, 3H), 2.80 (t, J= 7.2 Hz, 4H), 2.62 (t, J= 7.2 Hz, 4H), 1 .96 (t, J= 7.2 Hz ,4H). LCMS (m/z): 383.10 (M +Na)+; 96.00% (210 nm), 93.44% (254 nm). HPLC: 97.86% (210nm), 97.44% (254nm).
A^((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1-(trifluoromethyl)-1 H- pyrazole-3-sulfonamide
Figure imgf000210_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -(trifluoromethyl)-1 H-pyrazole-3-sulfonamide were used in general method C2 to give the titled compound as a white solid (5 mg, 1 %). 1H NMR (400 MHz, CD3OD) δ = 8.28 (d, J= 2.8 Hz, 1 H), 6.96 (d, J= 2.8, 1 H), 6.91 (s, 1 H), 2.84 (t, J = 7.4 Hz, 4H), 2.75 (t, J = 7.4 Hz, 4H), 2.03 (m, J = 7.4 Hz, 4H).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 -isopropyl-1 H- pyrazole-3-sulfonamide
Figure imgf000211_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -isopropyl-1 H-pyrazole-3-sulfonamide were used in general method C1 to give the titled compound as an off-white solid (40 mg, 9%). 1 H NMR (400 MHz, DMSO-d6): δ = 10.92 (s, 1 H), 8.02 (s, 1 H), 8.0 (s, 1 H), 6.94 (s, 1 H), 6.74 (s, 1 H), 4.67-4.59 (m, 1 H), 2.78 (t, J= 7.2 Hz, 4H), 2.58 (t, J = 7.2 Hz, 4H), 1 .95-1 .91 (m, 4H), 1 .44 (d, J = 6.8 Hz, 6H). LCMS (m/z): 387.1 (M - 1 )"; 97.14% (210 nm), 95.1 1 % (254 nm). HPLC: 95.57% (210 nm), 93.53% (254 nm).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 -isopropyl-1 H- pyrazole-4-sulfonamide
Figure imgf000211_0002
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -isopropyl-1 /-/-pyrazole-4-sulfonamide were used in general method C3 to give the titled compound as a white solid (40 mg, 10%) 1 H NMR (400 MHz, DMSO-d6): δ = 10.6 (s, 1 H), 8.44 (s, 1 H), 8.05 (s, 1 H), 7.86 (s, 1 H), 6.94 (s, 1 H), 4.63-4.57 (m, 1 H), 2.80 (t, J= 7.2 Hz, 4H), 2.57 (t, J= 7.6 Hz, 4H), 1 .94-1.89 (m, 4H), 1 .42 (d, J = 6.8 Hz 6H). LCMS (m/z): 389.20 (M +1 )+; 97.25% (210 nm), 94.22% (254 nm). HPLC: 97.13% (210nm), 95.06% (254nm).
1 -cyclopropyl-N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 H- pyrazole-3-sulfonamide
Figure imgf000212_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -cyclopropyl-1 H-pyrazole-3-sulfonamide were used in general method C3 to give the titled compound as a white solid (20 mg, 6%). 1H NMR (400 MHz, DMSO-afe) δ = 7.83 (s, 1 H), 7.8 (s, 1 H), 6.84 (s, 1 H), 6.48 (s, 1 H), 3.81 - 3.71 (m, 1 H), 2.77 (t, J = 7.4 Hz, 4H), 2.64 (t, J = 7.4 Hz, 4H), 2.02 - 1 .86 (m, 4H), 1 .09 - 0.93 (m, 4H).
1 -(ferf-butyl)-N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 H- pyrazole-3-sulfonamide
Figure imgf000212_0002
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -(ferf-butyl)-l H-pyrazole-3-sulfonamide were used in general method C3 to give the titled compound as a pale yellow solid (120 mg, 51 %). 1H NMR (400 MHz, DMSO-d6): δ = 10.85 (br.s., 1 H), 7.95 (s, 1 H), 7.88 (br.s., 1 H), 6.88 (s, 1 H), 6.63 (s, 1 H), 2.79 (t, J = 7.2 Hz, 4H), 2.61 (t, J = 7.2 Hz, 4H), 1 .96 (m, 4H), 1 .55 (s, 9H). LCMS (m/z): 403.15 (M +1 )+; 97.86% (210 nm), 96.50% (254 nm). HPLC: 96.45% (210nm), 95.89% (254nm).
N-((1 ,2,3,5,6 -hexahydro-s-indacen-4-yl)carbamoyl)-1-(1-methylpiperidin- 4-yl)-1 H-pyrazole-3-sulfonamide
Figure imgf000213_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -cyclohexyl-1 H-pyrazole-3-sulfonamide were used in general method C3 to give the titled compound as a white solid (20 mg, 6%). 1H N MR (400 MHz, DMSO-afe) δ = 10.8 (s, 1 H), 8.03 (s, 1 H), 7.99 (d, J = 2.4, 1 H), 6.95 (s, 1 H), 6.75 (d, J = 2.4 Hz, 1 H), 4.33 - 4.20 (m, 1 H), 2.79 (t, J = 7.4 Hz, 4H), 2.58 (t, J = 7.4 Hz, 4H), 2.05 - 1.88 (m, 6H), 1 .86 - 1 .63 (m, 6H), 1 .48 - 1 .33 (m, 2H).
A -{(1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1-phenyl-1 H-pyrazole- 3-sulfonamide
Figure imgf000213_0002
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -phenyl-1 H-pyrazole-3-sulfonamide were used in general method C1 to give the titled compound as an off-white solid (1 10 mg, 27%). 1H NMR (400 MHz, DMSO-d6): δ = 10.92 (s, 1 H), 8.61 (d, J = 2.4 Hz, 1 H), 7.95 (br.s., 1 H), 7.86 (d, J = 8.4 Hz, 2H), 7.56 (t, J = 7.6 Hz, 2H), 7.41 (t, J = 7.2 Hz, 1 H), 6.9 (d, J = 2.0 Hz, 1 H), 6.86 (s, 1 H), 2.77 (t, J = 7.2 Hz, 4H), 2.62 (t, J = 1.2 Hz, 4H), 1 .91 -1 .83 (m, 4H). LCMS (m/z): 421 .05 (M -1 )"; 96.62% (210 nm), 95.12% (254 nm). HPLC: 95.2% (210 nm), 95.77% (254 nm).
1 -benzyl-N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 H-pyrazole- 3-sulfonamide
Figure imgf000214_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and , 1 -benzyl-1 H-pyrazole-3-sulfonamide were used in general method C3 to give the titled compound as a white solid (85 mg, 34%). 1 H NMR (400 MHz, DMSO-d6): δ = 10.85 (s, 1 H), 8.05 (d, J= 2.4 Hz, 1 H), 7.99 (s, 1 H), 7.32-7.31 (m, 3H), 7.24-7.22(m, 2H), 6.93 (s, 1 H), 6.78 (d, J = 2.4 Hz, 1 H), 5.44 (s, 2H), 2.80 (t, J= 7.6 Hz, 4H), 2.57 (t, J = 7.2 Hz, 4H), 1 .96 (m, 4H). LCMS (m/z): 437.15 (M +1 )+; 97.70% (21 0 nm), 96.86% (254 nm). HPLC: 98.05% (210 nm), 97.56% (254 nm).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 -(1 -phenylethyl)-1 H- pyrazole-3-sulfonamide
Figure imgf000214_0002
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and , 1 -(1 -phenylethyl)-1 H-pyrazole-3-sulfonamide were used in general method C3 to give the titled compound as a white solid (0.13 g, 38%).1H N MR (400 MHz, DMSO-afe) δ = 7.94 (d, J=2.4Hz, 1H), 7.80 (s, 1H), 7.34- 7.18 (m, 5H), 6.85 (s, 1H), 6.62 (d, J=2.3 Hz, 1H), 5.68 (q, J=7.0 Hz, 1H), 2.76 (t, J= 7.4 Hz, 4H), 2.58 (t, J= 7.4 Hz, 4H), 1.90 (p, J = 7.4 Hz, 4H), 1.8 (d, J7.1 Hz, 3H).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1-(2-(piperidin-1- yl)ethyl)-1H-pyrazole-3-sulfonamide
Figure imgf000215_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and , 1 -(2-(piperidin-1 -yl)ethyl)-1 /-/-pyrazole-3-sulfonamide were used in general method C3 to give the titled compound as a white solid (110 mg, 25%).1H NMR (400 MHz, CD3OD): δ = 7.76 (d, J= 2.4 Hz , 1 H), 6.91 (s, 1 H), 6.73 (d, J= 2.4 Hz 1 H), 4.55 (t, J= 6.4 Hz, 2H), 3.41 (t, J= 6.0Hz, 2H), 3.02 (s, 4H), 2.86 (t, J= 7.2 Hz, 4H), 2.78 (t, J= 7.2 Hz, 4H), 2.06-1.99 (m, 4H), 1.74-1.70 (m,4H), 1.51 (d, J= 5.2 Hz,2H). LCMS (m/z): 458.20 (M+1)+; 100% (210 nm), 100% (254 nm). HPLC: 98.70% (210nm), 98.31% (254nm).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 ,5-dimethyl-1 H- pyrazole-3-sulfonamide
Figure imgf000215_0002
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 ,5-dimethyl-1 H-pyrazole-3-sulfonamide were used in general method C1 to give the titled compound as a white solid (15 mg, 4 %). 1 H NMR (400 MHz, DMSO-d6): δ = 10.7 (br.s. , 1 H), 7.98 (s, 1 H), 6.93 (s, 1 H), 6.52 (s, 1 H) , 3.79 (s, 3H), 2.80 (t, J= 7.2 Hz, 4H), 2.62 (t, J= 7.6 Hz, 4H), 2.28 (s, 3H), 1.98-1.93 (m, 4H). LCMS (m/z): 397.10 (M+Na)+; 97.75% (210 nm), 88.23% (254 nm). HPLC: 94.42% (210nm), 95.19% (254nm).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1-methyl-5- (trifluoromethyl)-1 H-pyrazole-3-sulfonamide
Figure imgf000216_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -methyl-5-(trifluoromethyl)-1 /-/-pyrazole-3-sulfonamide were used in general method C3 to give the titled compound as a white solid (200 mg, 48%). 1 H NMR (400 MHz, CD3OD): δ = 7.10 (s, 1 H), 6.87 (s, 1 H), 4.03 (s, 3H), 2.83 (t, J = 7.2 Hz, 4H), 2.74- (t, J = 7.2 Hz, 4H), 2.03-1 .99 (m, 4H). LCMS( m/z): 429.10 (M +1 )+; 97.73% (210 nm), 95.71 % (254 nm). HPLC: 94.95% (210nm), 93.52% (254nm). yV-((2,6-diisopropylphenyl)carbamoyl)-1-methyl-5-(trifluoromethyl)-1 H- pyrazole-3-sulfonamide
Figure imgf000216_0002
2-lsocyanato-1 ,3-diisopropylbenzene (prepared using general method A1 ) and 1 -methyl-5-(trifluoromethyl)-1 /-/-pyrazole-3-sulfonamide were used in general method C3 to give the titled compound as a white solid (70 mg, 39%). 1 H NMR (400 MHz, CD3OD): δ = 7.18-7.16 (m, 1 H), 7.10-7.08 (m, 3H), 4.03 (s, 3H), 3.17-3.13 (m, 2H), 1 .03 (d, J= 6.0 Hz, 12H). LCMS (m/z): 433.15 (M +1 )+; 99.73% (210 nm), 98.16% (254 nm). HPLC: 97.51 % (210 nm), 95.47% (254 nm).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1-isopropyl-5- (trifluoromethyl)-1 H-pyrazole-3-sulfonamide
Figure imgf000217_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -isopropyl-5-(trifluoromethyl)-1 - -pyrazole-3- sulfonamide were used in general method C3 to give the titled compound as a white solid (15 mg, 12%). 1 H NMR (400 MHz, DMSO-d6): δ = 8.54 (s, 1 H), 6.90 (s, 1 H), 6.77 (s, 1 H), 4.62-4.56 (m, 1 H), 2.76 (t, J= 7.2 Hz, 4H), 2.67 (t, J= 7.6 Hz, 4H), 1 .92-1.84 (m, 4H), 1.44 (d, J= 6.4 Hz, 6H). LCMS (m/z): 455.05 (M -1 )"; 96.13% (210 nm), 95.41 % (254 nm). HPLC: 95.71 % (210 nm), 95.12% (254 nm).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl )-5-isopropyl-1 -methyl- 1 H-pyrazole-3-sulfonamide
Figure imgf000217_0002
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 5-isopropyl-1 -methyl-1 H-pyrazole-3-sulfonamide were used in general method C3 to give the titled compound as a white solid (10 mg, 17%). 1H NMR (400 MHz, CD3OD): δ = 6.88 (s, 1 H), 6.50 (s, 1 H), 3.82 (s, 3H), 3.08 - 3.03 (m, 1 H), 2.83 (t, J = 1.2 Hz, 4H), 2.71 (t, J = 7.2 Hz, 4H), 2.04-1.96 (m, 4H), 1.21 (d, J = 6.8 Hz, 6H). LCMS( m/z): 403.20 (M +1 )+ ; 98.39% (210 nm), 94.19% (254 nm). HPLC: 95.62% (210nm), 93.00% (254nm). yV-((2,6-diisopropylphenyl)carbamoyl)-5-(2-hydroxypropan-2-yl)-1-methyl- 1 H-pyrazole-3-sulfonamide
Figure imgf000218_0001
2-lsocyanato-1 ,3-diisopropylbenzene (prepared using general method A1 ) and 5-(2-hydroxypropan-2-yl)-1 -methyl-1 /-/-pyrazole-3-sulfonamide were used in general method C1 to give the titled compound as a white solid (90 mg, 26%). 1 H NMR (400 MHz, CD3OD): δ = 7.25-7.24 (m, 1 H), 7.16-7.14 (m, 2H), 6.67(s, 1 H), 4.13 (s, 3H), 3.1 1 -3.08 (m, 2H), 1 .61 (s, 6H), 1 .16 (d, J = 6.8 Hz, 12H). LCMS (m/z): 423.20 (M +1 )+; 99.16% (210 nm), 97.19% (254 nm). HPLC: 98.16% (210nm), 97.09% (254nm).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl )-5-(2-hydroxypropan- 2-yl)-1 -methyl-1 H-pyrazole-3-sulf onamide.
Figure imgf000218_0002
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 5-(2-hydroxypropan-2-yl)-1 -methyl-1 - -pyrazole-3- sulfonamide were used in general method C1 to give the titled compound as a white solid (70 mg, 15%). 1H NMR (400 MHz, DMSO-afe) δ 8.01 (s, 1 H), 6.92 (s, 1 H), 6.53 (s, 1 H), 5.51 (s, 1 H), 4.02 (s, 3H), 2.79 (t, J= 7.4 Hz, 4H), 2.62 (t, J = 7.4 Hz, 4H), 1 .95 (p, J = 7.4 Hz, 4H), 1 .50 (s, 6H).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl )-5-(2-hydroxypropan- 2-yl)-1 -phenyl-1 H-pyrazole-3-sulf onamide
Figure imgf000219_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 5-(2-hydroxypropan-2-yl)-1 -phenyl-1 - -pyrazole-3- sulfonamide were used in general method C1 to give the titled compound as a white solid (10 mg, 2%). 1 H NMR (400 MHz, CD3OD): δ = 7.54 (s, 5H), 6.59 (s, 1 H), 6.91 (s, 1 H), 2.86 (t, J= 7.2 Hz, 4H), 2.69 (t, J= 7.2 Hz, 4H), 2.05-1 .96 (m, 4H), 1 .44 (s, 6H). LCMS (m/z): 481.20 (M -1 )"; 93.76% (210 nm), 93.24% (254 nm). HPLC: 95.86% (210 nm), 93.93% (254 nm).
1-benzyl-N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-5-(2- hydroxypropan-2-yl)-1 H-pyrazole-3-sulfonamide
Figure imgf000219_0002
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 1 -benzyl-5-(2-hydroxypropan-2-yl)-1 - -pyrazole-3- sulfonamide were used in general method C1 to give the titled compound as a white solid (40 mg, 7%). 1H NMR (400 MHz, CD3OD): δ = 7.20-7.14 (m, 5H), 6.95 (s 2H), 6.73 (s, 1 H), 5.77 (s, 2H), 2.86 (t, J = 7.2 Hz, 4H), 2.68 (t, J = 7.6 Hz, 4H), 2.01 -1.94 (m, 4H), 1.51 (s, 6H). LCMS (m/z): 494.7 (M +1 )+; 98.74% (210 nm), 96.05% (254 nm). HPLC: 95.1 1 % (210 nm), 95.08% (254 nm). yV-((4-chloro-2,6-diisopropylphenyl)carbamoyl)-5-(2-hydroxypropan-2-yl)-1- methyl-1 H-pyrazole-3-sulfonamide
Figure imgf000220_0001
5-chloro-2-isocyanato-1 ,3-diisopropylbenzene (prepared using general method A2) and 5-(2-hydroxypropan-2-yl)-1 -methyl-1 /-/-pyrazole-3-sulfonamide were used in general method C2 to give the titled compound as a white solid (83 mg, 17%). 1H NMR (600 MHz, DMSO-afe) δ = 7.81 (s, 1 H), 7.1 0 (s, 2H), 6.42 (s, 1 H), 5.45 (s, 1 H), 3.99 (s, 3H), 3.03 (hept, J= 7.0 Hz, 2H), 1.47 (s, 6H), 1 .05 (d, J = 1 .8 Hz, 12H). HRMS calculated for C20H28CI1 N4O4S1 [M-H]" 455.1525, found 455.1515. yV-((4-chloro-2,6-diisopropylphenyl)carbamoyl)-5-(2-hydroxypropan-2-yl)-1- phenyl-1 H-pyrazole-3-sulfonamide
Figure imgf000220_0002
5-chloro-2-isocyanato-1 ,3-diisopropylbenzene (prepared using general method A2) and 5-(2-hydroxypropan-2-yl)-1 -phenyl-1 /-/-pyrazole-3-sulfonamide were used in general method C2 to give the titled compound as a white solid (168 mg, 31 %). 1H N MR (600 MHz, DMSO-afe) δ = 7.87 (s, 1 H), 7.52 (s, 5H), 7.10 (s, 2H), 6.71 (s, 1 H), 5.42 (s, 1 H), 3.10 - 2.92 (m, 2H), 1.31 (s, 6H), 1 .02 (d, J = 7.0 Hz, 12H). HRMS calculated for C25H30CI1 N4O4S1 [M-H]" 517.1682, found 517.1671 .
N-((4-chloro-2,6-dimethylphenyl)carbamoyl)-1-isopropyl-1 H-pyrazole-3- sulfonamide
Figure imgf000221_0001
A solution of 4-chloro-2,6-dimethylaniline (50 mg, 0.321 mmol) in DCM (5 mL) was treated with Et3N (50 mg, 0.48 mmol) and cooled to 0 °C, Phenyl chloroformate (60 mg, 0.39 mmol) was added dropwise at 0 °C. The reaction mixture was warmed to RT and stirred for 12 h. Upon completion the reaction mixture was diluted with saturated NaHC03 solution, extracted with DCM (2 x 20 mL) and the combined organic extract washed with water, brine dried (Na2S04) and concentrated in vacuo. The residue obtained was washed with n- pentane and dried in vacuo to give phenyl (4-chloro-2,6- dimethylphenyl)carbamate (75 mg, 85%) as a brown solid. 1 H NMR (300 MHz, CDCI3): δ = 7.46-7.37 (m, 4H), 7.26-7.20 (m, 2H), 7.12-7.1 1 (m, 2H), 2.33(s, 6H). LCMS (m/z): 275.9 [M+H]+.
1 -isopropyl-1 /-/-pyrazole-3-sulfonamide (150 mg, 0.79 mmol) was dissolved in anhydrous THF (5 mL) and treated carefully with NaH (80 mg, 1 .98 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 30 min then treated with a solution of phenyl (4-chloro-2,6- dimethylphenyl)carbamate (240 mg, 0.87 mmol) in THF (3 mL) under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 3 h. Upon completion the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 30 mL) and the combined organic extract was washed with water, brine dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluant to give A/-((4-chloro-2,6- dimethylphenyl)carbamoyl)-1 -isopropyl-1 - -pyrazole-3-sulfonamide (90 mg, 31 %) as a white solid. 1H NMR (400 MHz, DMSO-afe): δ = 1 1 .05 (s, 1 H), 7.99 (d, J = 2.8 Hz, 1 H), 7.93 (s, 1 H), 7.13 (s, 2H), 6.73(d, J = 2.4 Hz, 1 H), 4.64-4.57 (m, 1 H), 2.03 (s, 6H), 1 .43 (d, J = 6.8 Hz, 6H). LCMS (m/z): 370.95 [M+H]+.; 97.62% (210 nm), 97.48% (254 nm). HPLC: 97.20% (210nm). HRMS calculated for Ci5H18CliN403Si [M-H]" 369.0794, found 369.0785.
N-((4-chloro-2,6-dimethoxyphenyl)carbamoyl)-1-isopropyl-1H-pyrazole-3- sulfonamide
Figure imgf000222_0001
A solution of 4-chloro-2,6-dimethoxyaniline (200 mg, 1 .06 mmol) in THF (8 mL) was cooled to 0 °C and treated with NaH (62 mg, 1 .59 mmol) under nitrogen atmosphere. The resulting mixture was stirred for 15 min. before phenylchloroformate (330 mg, 2.13 mmol) was added dropwise at 0°C. The reaction mixture was warmed to RT and stirred for 12h. Upon completion, the reaction mixture was diluted with EtOAc and filtered through a celite pad and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 10% EtOAc-hexanes eluant to give phenyl (4-chloro-2,6-dimethoxyphenyl)carbamate (0.2 g, 61 %) as a white solid. 1H NMR (300 MHz, CDCI3): δ = 7.35-7.33 (m, 2H), 7.20-7.19 (m, 3H), 6.61 (s, 2H), 3.83 (s, 6H). 1 -isopropyl-1 H-pyrazole-3-sulfonamide (100 mg, 0.53 mmol) was dissolved in anhydrous THF (5 ml_) and treated carefully with NaH (52 mg, 1 .32 mmol) at 0 °C under nitrogen atmosphere.). The resulting mixture was stirred at RT for 40 min then treated with a solution of phenyl (4-chloro-2,6- dimethoxyphenyl)carbamate (180 mg, 0.58 mmol) in THF (3 ml_) under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 12 h. Upon completion the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 50 ml_) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The combined organic extract was washed with water, brine, dried over anhydrous Na2S04 and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC [column: Gemini NX C18 (21 .2 mm x 150 mm particle size 5pm); flow: 20 mL/min; eluent: 10 mM ammonium bicarbonate in water (A) & MeCN (B); gradient: T/% B= 0/20, 2/20, 8/70]. The fractions were lyophilized to give N-((4-chloro-2,6-dimethoxyphenyl)carbamoyl)-1 -isopropyl- 1 H-pyrazole-3-sulfonamide (20 mg, 9%) as a white solid. 1 H NMR (400 MHz, DMSO-afe): δ = 7.90 (d, J = 2.0 Hz, 1 H), 7.28 (s, 1 H), 6.72 (s, 2H), 6.63 (d, J = 2.0 Hz, 1 H), 4.60-4.54 (m, 1 H), 3.70 (s, 6H), 1 .43(d, J = 6.8 Hz, 6H). LCMS (m/z): 403.0 [M+H]+.; 90.61 % (210nm). HPLC: 91 .63% (210nm). HRMS calculated for Ci5H18Cli N405Si [M-H]" 401 .0692, found 401 .0684. N-((4-chloro-2-methy1-6-(trifluoromethyl)phenyl)carbamoyl)-1-isopropyl-1 H- pyrazole-3-sulfonamide
Figure imgf000223_0001
4-Chloro-2-methyl-6-(trifluoromethyl)aniline (50 mg, 0.24 mmol) was dissolved in anhydrous THF (5 ml_) and treated with Et3N (30 mg, 0.29 mmol) at RT. The solution was treated with triphosgene (30 mg, 0.095 mmol) and resulting mixture was stirred at 60 °C for 4 h. The reaction mixture was concentrated in vacuo. The residue obtained was stirred with n-pentane (20 ml_) for 10 min, filtered through a celite pad and concentrated in vacuo to give the corresponding isocyanate as a white solid. In another 50 mL round bottom flask, 1 -isopropyl-1 H-pyrazole-3-sulfonamide (40 mg, 0.212 mmol) was dissolved in anhydrous THF (5 mL) and treated carefully with NaH (22 mg, 0.529 mmol) at 0 °C under nitrogen atmosphere. It was stirred at RT for 30 minutes. The aforementioned solution of isocyanate was added in THF under nitrogen atmosphere. The resulting reaction mixture was stirred at RT for 2 h. Upon completion of reaction the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 25 mL) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluant to give N-((4-chloro-2-methyl-6- (trifluoromethyl)phenyl)carbamoyl)-1 -isopropyl-1 /-/-pyrazole-3-sulfonamide. This was triturated with diethyl ether and n-pentane to give N-((4-chloro-2-methyl-6- (trifluoromethyl)phenyl)carbamoyl)-1 -isopropyl-1 H-pyrazole-3-sulfonamide (35 mg, 35%) as a white solid. 1 H NMR (400 MHz, DMSO-afe): δ = 1 1 .05 (s, 1 H), 8.09 (s, 1 H), 7.96 (s, 1 H), 7.70 (s, 1 H), 7.61 (s, 1 H), 6.67 (d, J = 0.4 Hz, 1 H), 4.62 (m, 1 H), 2.05 (s, 3H), 1 .43 (d, J= 6.8 Hz, 6H). 19F NMR (400 MHz, DMSO- afe): δ = -60.82. LCMS (m/z): 425.00 [M+H]+; 94.05% (210 nm). HPLC: 98.03% (210nm). HRMS calculated for CisHisC FsNUOsSi [M-H]" 423.051 1 , found 423.0513.
N-((4-chloro-2-methoxy-6-(trifluoromethyl)phenyl)carbamoyl)-1-isopropyl- 1 H-pyrazole-3-sulfonamide
Figure imgf000225_0001
4-Chloro-2-methoxy-6-(trifluoromethyl)aniline (50 mg, 0.22 mmol) was dissolved in anhydrous THF (2 ml_) and treated with Et3N (27 mg, 0.27 mmol) at RT. The solution was treated with triphosgene (32 mg, 0.1 1 mmol) and resulting mixture was stirred at 70 °C for 3 h. The reaction mixture was concentrated in vacuo and the residue obtained stirred with 5% EtOAc-hexanes (20 ml_) for 10 min, filtered through celite and concentrated in vacuo to give the desired isocyanate as a white solid. In another 50 mL round bottom flask, 1 -isopropyl-1 H-pyrazole- 3-sulfonamide (42 mg, 0.22 mmol) was dissolved in anhydrous THF (5 mL) and treated carefully with NaH (18 mg, 0.44 mmol) at 0 °C under nitrogen atmosphere. The mixture was stirred at 0 °C for 20 minutes and treated with aforementioned solution of isocyanate in THF under nitrogen atmosphere. The resulting reaction mixture was stirred at 0-10 °C for 2 h. Upon completion, the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 50 mL) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 50% EtOAc-hexanes eluant to give N-((4-chloro-2-methoxy-6-(trifluoromethyl)phenyl)carbamoyl)-1 - isopropyl-1 /-/-pyrazole-3-sulfonamide (10 mg, 10%) as a white solid. 1 H NMR (400 MHz, CDCIs): δ = 7.81 (s, 1 H), 7.53 (d, J= 2.4 Hz, 1 H), 7.22 (d, J= 1 .6 Hz, 1 H), 7.10 (s, 1 H), 6.82 (d, J = 2.0 Hz, 1 H), 4.64-4.57 (m, 1 H), 3.84 (s, 3H), 1 .54 (d, J= 6.8 Hz, 6H). 19F NMR (400 MHz, CDCI3): δ = -61 .55. LCMS (m/z): 441.05 [M+H]+.; 94.58% (210 nm). HPLC: 92.16% (210nm). HRMS calculated for Ci5H15Cli F3N4O4Si [M-H]" 439.0460, found 439.0478. N-((4-chloro-2,6-diethylphenyl)carbamoyl)-1-isopropyl-1 H-pyrazole-3- sulfonamide
Figure imgf000226_0001
A solution of 4-chloro-2,6-dicyclopropylaniline (100 mg, 0.546 mmol) in THF (5 mL) was cooled to 0 °C and treated with NaH (30 mg, 0.66 mmol) under nitrogen atmosphere and stirred for 15 min. Phenyl chloroformate (130 mg, 0.819 mmol) was added dropwise to the aforementioned solution at 0 °C. The reaction mixture was warmed to RT and stirred for 12 h. Upon completion the mixture was diluted with EtOAc, filtered through a celite pad and concentrated in vacuo. The crude product was was purified by column chromatography on silica gel (60-120 mesh) using 10% EtOAc-hexanes to give phenyl (4-chloro- 2,6-diethylphenyl)carbamate (0.15 g, 91 %) as a white solid. 1 H NMR (300 MHz, CDCI3): δ = 7.38-7.33 (m, 2H), 7.23-7.18 (m, 3H), 7.13 (m, 2H), 6.27 (s, 1 H), 2.75-2.64 (m, 4H), 1 .28-1.22 (m, 6H).
1 -isopropyl-1 /-/-pyrazole-3-sulfonamide (75 mg, 0.40 mmol) was dissolved in anhydrous THF (5 mL) and treated carefully with NaH (40 mg, 0.99 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 30 min then treated with a solution of give phenyl (4-chloro-2,6- diethylphenyl)carbamate (130 mg, 0.44 mmol) in THF (3 mL) under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 4 h. Upon completion, the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 30 mL) and the combined organic extract was washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was was purified by column chromatography on silica gel (60-120 mesh) using 30-40% EtOAc-hexanes eluant followed by trituration with diethyl ether and n-pentane to give N-((4-chloro-2,6- diethylphenyl)carbamoyl)-1 -isopropyl-1 --pyrazole-3-sulfonamide (40 mg, 24%) as a white solid. 1 H NMR (400 MHz, DMSO-afe): δ = 1 1 .05 (s, 1 H), 7.99 (d, J = 2.4 Hz, 1 H), 7.89 (s, 1 H), 7.12 (s, 2H), 6.72 (d, J = 2.4 Hz, 1 H), 4.62-4.59 (m, 1 H), 2.42 (q, J = 7.6 Hz, 4H), 1 .43 (d, J = 6.8 Hz, 6H), 1 .02 (t, J = 7.6 Hz, 6H). LCMS (m/z): 399.0 [M+H]+, 96.72% (210nm). HPLC: 97.13% (210nm). HRMS calculated for C17H22CI1 N4O3S1 [M-H]" 397.1 107, found 397.1090. yV-((4-chloro-2,6-diisopropylphenyl)carbamoyl)-1-isopropyl-1 H-pyrazole-3- sulfonamide
Figure imgf000227_0001
5-Chloro-2-isocyanato-1 ,3-diisopropylbenzene (prepared using general method A2) and 1 -isopropyl-1 H-pyrazole-3-sulfonamide were used in general method C2 to give the titled compound as a white solid (221 mg, 49%). 1H NMR (600 MHz, DMSO-dS) δ = 7.70 (d, J= 2.3 Hz, 1 H), 7.49 (s, 1 H), 7.00 (s, 2H), 6.36 (s, 1 H), 4.62 - 4.29 (m, 1 H), 3.1 1 (d, J = 6.4 Hz, 2H), 1.38 (d, J= 6.8 Hz, 6H), 1.01 (d, J = 6.8 Hz, 12H). 13C NMR (151 MHz, DMSO) δ = 160.96, 156.43, 150.19, 135.25, 131 .49, 128.53, 123.27, 105.21 , 54.23, 28.80, 24.15, 23.55. HRMS calculated for C19H26CI1 N4O3S1 [M-H]" 425.1420, found 425.1409.
N-((4-chloro-2,6-dicyclo propylphenyl)carbamoyl)-1 -isopropyl-1 H-pyrazole- 3-sulfonamide
Figure imgf000228_0001
A solution of 4-chloro-2,6-dicyclopropylaniline (150 mg, 0.724 mmol) in THF (5 ml_) was cooled to 0 °C. NaH (35 mg, 0.87 mmol) was added in portions to aforementioned solution and stirred for 20 min. Phenyl chloroformate (170 mg, 1 .08 mmol) was added dropwise to the aforementioned solution at 0 °C. The reaction mixture was warmed to RT and stirred for 12 h. Upon completion, the mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 15% EtOAc-hexanes eluant to give phenyl (4-chloro-2,6- dicyclopropylphenyl)carbamate (195 mg, 83%) as a white solid. 1H NMR (300 MHz, CDCI3): δ = 7.37-7.35 (m, 2H), 7.21 -7.19 (m, 3H), 6.84-6.83 (m, 2H), 2.08-2.04 (m, 2H), 1 .04-1 .02 (m, 4H), 0.69-0.68 (m, 4H). LCMS (m/z): 328.2 [M+H]+.
1 -isopropyl-1 /-/-pyrazole-3-sulfonamide (100 mg, 0.53 mmol) was dissolved in anhydrous THF (5 ml_) and treated carefully with NaH (53 mg, 1 .32 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 30 min then treated with a solution of phenyl (4-chloro-2,6- dicyclopropylphenyl)carbamate (190 mg, 0.582 mmol) in THF (3 ml_) under nitrogen atmosphere at 0 °C. The reaction mixture was warmed to RT and stirred for 4 h. Upon completion of reaction the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 30 ml_) and the combined organic extract washed with water, brine, dried (Na2SO4) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluant to give N-((4-chloro-2,6- dicyclo propylphenyl)carbamoyl)-1 -isopropyl-1 /-/-pyrazole-3-sulfonamide. This was triturated with diethyl ether and n-pentane to give (25 mg, 1 1 %) as a white solid. 1 H NMR (400 MHz, DMSO-afe): δ = 1 1 .05 (s, 1 H), 8.01 -7.98 (m, 2H), 6.74 (s, 3H), 4.59-4.56 (m, 1 H), 1.77 -1 .76 (m, 2H), 1 .41 (d, J = 6.8 Hz, 6H), 0.77- 0.75 (m, 4H), 0.56-0.55 (m, 4H). LCMS (m/z): 423.00 [M+H]+.; 93.58% (210 nm). HPLC: 92.87% (210nm). HRMS calculated for C19H22CI1 N4O3S1 [M-H]" 421 .1 107, found 421 .1 107.
/V-((7-chloro-5-cyclopropyl-2,3-dihydro-1 H-inden-4-yl)carbamoyl)-1- isopropyl-1 H-pyrazole-3-s lfonamide
Figure imgf000229_0001
7-chloro-5-cyclopropyl-2,3-dihydro-1 - -inden-4-amine, 6 (70mg, 0.33 mmol) was dissolved in THF (5 ml_) and cooled to 0 °C. NaH (20 mg, 0.505 mmol) was added to the aforementioned solution under nitrogen atmosphere and stirred for 15 min before phenyl chloroformate (100 mg, 0.674 mmol) was added dropwise at 0°C. The reaction mixture was warmed to RT and stirred for 12h. Upon completion the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using, 10% EtOAc-hexanes eluent to give phenyl (7-chloro-5-cyclopropyl-2,3-dihydro-1 - -inden-4-yl)carbamate (80 mg, 73%) as a brown solid. 1H NMR (300 MHz, CDCI3): δ = 7.39-7.37 (m, 3H), 7.25-7.24 (m, 2H), 6.85(s, 1 H), 3.0-2.94 (m, 4H), 2.12-2.10 (m, 2H), 1.34 (m, 1 H), 0.96-0.95 (m, 2H), 0.59-0.57 (m, 2H). LCMS (m/z): 328.30 [M+H]+.
1 -isopropyl-1 /-/-pyrazole-3-sulfonamide (41 mg, 0.219 mmol) was dissolved in anhydrous THF (3 ml_) and treated carefully with NaH (21 mg, 0.549 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 30 min and was treated with a solution of phenyl (7-chloro-5-cyclopropyl-2,3- dihydro-1 -/-inden-4-yl)carbamate (80 mg, 0.244 mmol) in THF (2 mL) under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 4 h. Upon completion the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 30 mL) and the combined organic extract was washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC [column: Gemini NX C18 (21 .2 mm x 150 mm particle size 5pm); flow: 20 mL/m in; eluent: 10 mM ammonium bicarbonate in water (A) & MeCN (B); gradient: T/% B= 0/20, 2/30, 8/70]. The fractions were lyophilized to give N-((7- chloro-5-cyclopropyl-2,3-dihydro-1 /-/-inden-4-yl)carbamoyl)-1 -isopropyl-1 H- pyrazole-3-sulfonamide (20 mg, 20%) as a white solid. 1 H NMR (400 MHz, DMSO-afe): δ = 7.95 (d, J = 2.4 Hz, 1 H), 6.74 (s, 1 H), 6.69(d, J = 2.4 Hz, 1 H), 4.60-4.55 (m, 1 H), 2.84 (t, J = 7.6 Hz ,2H), 2.66 (t, J = 7.6 Hz, 2H), 1 .95-1 .92 (m, 2H), 1 .79-1 .76 (m, 1 H), 1 .41 (d, J = 6.4 Hz, 6H), 0.82-0.78 (m, 2H), 0.54- 0.50 (m, 2H). LCMS (m/z): 421 .15 [M-H]"; 94.19% (210 nm). HPLC: 95.46% (210nm). HRMS calculated for Ci9H22Cli N403Si [M-H]" 421 .1 107, found 421 .1 1 1 0.
5-chloro-3-cyclopropyl-2-(3-((1 -isopropyl-1 H-pyrazol-3-yl)sulfonyl)ureido)- N,N-dimethylbenzamide
Figure imgf000230_0001
2-am ino-5-chloro-3-cyclopropyl-N, N-dimethylbenzamide (200 mg, 0.84 mmol) was dissolved in anhydrous THF (5 mL) and treated carefully with NaH (50 mg, 1 .26 mmol) at 0 °C under nitrogen atmosphere. The resulting reaction mixture was stirred for 15 min. then phenyl chloroformate (262 mg, 1 .68 mmol) was added dropwise at 0 °C. The reaction mixture was warmed to RT and stirred for 12 h. Upon completion the reaction mixture was diluted with EtOAc, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 15% EtOAc-hexanes eluant to give phenyl (4-chloro-2-cyclopropyl-6-(dimethylcarbamoyl) phenyl)carbamate (0.14, 47%) as dark brown liquid. 1H NMR (300 MHz, CDCI3): δ = 7.39-7.34 (m, 2H), 7.23-7.15 (m, 3H), 7.08-7.02 (m, 2H), 3.09(s, 3H), 2.96 (s, 3H), 2.05-2.0 (m, 1 H), 1 .05-1.02 (m, 2H), 0.71 -0.69 (m, 2H). LCMS (m/z): 358.60 [M+H]+.
1 -isopropyl-1 /-/-pyrazole-3-sulfonamide (57 mg, 0.30 mmol) was dissolved in anhydrous THF (5 ml_) and treated carefully with NaH (18 mg, 0.451 mmol) at O °C under nitrogen atmosphere. The resulting mixture was stirred at RT for 45 min then treated with a solution of phenyl (4-chloro-2-cyclopropyl-6- (dimethylcarbamoyl) phenyl)carbamate (120 mg, 0.335 mmol) in THF (3 ml_) under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 2 h. Upon completion, the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 30 ml_) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC [column: Gemini NX C18 1 10A AXIA (21 .2 mm x 150 mm particle size 5pm); flow: 18 mL/min; eluent: 10 mM ammonium bicarbonate in water (A) & MeCN (B); gradient: T/% B= 0/20, 2/20, 10/60]. The fractions were lyophilized to give 5-chloro-3-cyclopropyl-2-(3-((1 -isopropyl-1 - -pyrazol-3-yl)sulfonyl)ureido)-A/,A/- dimethylbenzamide (10 mg, 7%) as a white solid. 1H NMR (400 MHz, CD3OD): 5 = 7.77 (s, 1 H), 7.1 1 (d, J = 2.4 Hz, 1 H), 7.05 (d, J = 2.0 Hz, 1 H), 6.76 (s, 1 H), 4.62-4.53 (m, 1 H), 2.96 (s, 3H), 2.84 (s, 3H), 1 .88-1 .87(m, 1 H), 1 .50 (d, J= 6.8 Hz, 6H), 0.90-0.88 (m, 2H), 0.63-0.61 (m, 2H). LCMS (m/z): 454.0 [M+H]+.; 97.52% (210 nm). HPLC: 92.05% (210nm). HRMS calculated for C19H23CI1 N5O4S1 [M-H]" 452.1 165, found 452.1 180.
N-((4,6-dimethylpyrimidin-2-yl)carbamoyl)-1-isopropyl-1 H-pyrazole-3- sulfonamide
Figure imgf000232_0001
A solution of 4,6-dimethylpyrimidin-2-amine (300 mg, 2.43 mmol) in THF (10 mL) was cooled to 0 °C and treated with NaH (140 mg, 3.64 mmol) under nitrogen atmosphere. The resulting mixture was stirred for 15 min then treated with phenyl chloroformate (0.6 mL, 4.87 mmol) at 0°C. The reaction mixture was warmed to RT and stirred for 12 h. Upon completion, the reaction mixture was diluted with EtOAc (30 mL) filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 30% EtOAc-hexanes eluant to give phenyl (4,6- dimethylpyrimidin-2-yl)carbamate (250 mg, 42%) as a white solid. 1H NMR (300 MHz, CDCI3): δ = 8.14 (s, 1 H), 7.45-7.34 (m, 2H), 7.25-7.18 (m, 3H), 6.78 (s, 1 H), 2.46 (s, 6H). LCMS (m/z): 244.30 [M+H]+.
1 -isopropyl-1 /-/-pyrazole-3-sulfonamide (75 mg, 0.396 mmol) was dissolved in anhydrous THF (50 mL) and treated carefully with NaH (40mg, 0.99 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was warmed to RT and stirred for 30 min. The reaction mixture was cooled to 0°C then treated with a solution of phenyl (4,6-dimethylpyrimidin-2-yl)carbamate (100 mg, 0.436 mmol) in THF (5 mL) under nitrogen atmosphere at 0 °C. The reaction mixture was warmed to RT and stirred for 3 h. Upon completion, the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 50 mL) and the combined organic extract washed with water, brine, dried (Na2SO4) and concentrated in vacuo. The crude product was purified by reverse phase preparative HPLC [column: Gemini NX-bridge (150 mm x 21.2 mm particle size 5pm); flow: 15 mL/min; eluent: 10 mM ammonium bicarbonate in water (A) & MeCN (B); gradient: T/% B= 0/10, 2/20, 10/60]. The fractions were lyophilized to give N-((4,6-dimethylpyrimidin-2-yl)carbamoyl)-1 -isopropyl-1 - -pyrazole-3- sulfonamide (10 mg, 13%) as a white solid. 1H NMR (400 MHz, CDCI3): δ = 13.0 (s, 1 H), 7.49-7.46 (m, 2H) , 7.02 (d, J = 2.4 Hz, 1 H), 6.74 (s, 1 H), 4.62- 4.57 (m, 1 H), 2.45 (s, 6H), 1 .53 (d, J= 6.8 Hz, 6H). LCMS (m/z): 339.10 [M+H]+. 99.70% (210 nm), 100% (254 nm). HPLC: 97.22% (210nm). HRMS calculated for Ci3H17N603Si [M-H]" 337.1088, found 337.1099.
N-((4,6-di-ferf-butylpyrimidin-2-yl)carbamoyl)-1-isopropyl-1 H-pyrazole-3- sulfonamide
Figure imgf000233_0001
A solution of 4,6-di-ieri-butylpyrimidin-2-amine (0.15 g, 0.72 mmol) in THF (5 ml_) was cooled to 0 °C and treated with NaH (35 mg, 0.86 mmol) under nitrogen atmosphere. The resulting mixture was stirred for 15 min and phenyl chloroformate (0.17 g, 1 .08 mmol) was added dropwise to the aforementioned solution at 0 °C. The reaction mixture was warmed to RT and stirred for 12 h. Upon completion, the reaction mixture was concentrated in vacuo and the residue obtained diluted with ethyl acetate, filtered through celite and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 15% EtOAc-hexanes eluant to give phenyl (4,6-di-ieri-butylpyrimidin-2-yl)carbamate (140 mg, 59%) as a white solid. 1H NMR (400 MHz, CDCI3): δ = 7.95 (s, 1 H), 7.65-7.61 (m, 2H), 7.50-7.45 (m, 4H), 1 .32 (s, 18H). LCMS (m/z): 328.40 [M+H]+;
1 -lsopropyl-1 /-/-pyrazole-3-sulfonamide (50 mg, 0.264 mmol) was dissolved in anhydrous THF (40 ml_) and treated carefully with NaH (27 mg, 0.661 mmol) at 0 °C under nitrogen atmosphere. The resulting mixture was stirred for 30 minutes then treated with a solution of phenyl (4,6-di-terf-butylpyrimidin-2- yl)carbamate (95 mg, 0.29 mmol) in THF (5 ml_) under nitrogen atmosphere at 0 °C. The resulting reaction mixture was warmed to RT and stirred for 4 h. Upon completion, the reaction mixture was diluted with saturated NH4CI solution, extracted with EtOAc (2 x 50 ml_) and the combined organic extract washed with water, brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel (60-120 mesh) using 40% EtOAc-hexanes eluant to give N-((4,6-di-ieri-butylpyrimidin-2-yl)carbamoyl)-1 - isopropyl-1 /-/-pyrazole-3-sulfonamide (38 mg, 25%) as a white solid. 1 H NMR (400 MHz, DMSO-de): δ = 13.75 (s, 1 H), 10.71 (s, 1 H), 8.01 (d, J = 2.4 Hz, 1 H), 7.18 (s, 1 H), 6.81 (d, J = 2.4 Hz, 1 H), 4.61 -4.54 (m, 1 H), 1.38(d, J = 6.8 Hz, 6H), 1 .31 (s, 18H). LCMS (m/z): 423.50 [M+H]+; 99.88% (210 nm). HPLC: 98.49% (210nm). HRMS calculated for C19H29N6O3S1 [M-H]" 421 .2027, found 421 .2008.
PHENYL/BICYCLICS
A-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)benzenesulfonamide
Figure imgf000234_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and phenylsulfonamide were used in general method C1 to give the titled compound as a white solid (50 mg, 13%). 1 H NMR (400 MHz, CD3OD): δ = 8.05 (d, J = 7.6 Hz, 2H), 7.72 (t, J = 7.6 Hz, 1 H), 7.63 (t, J = 7.6 Hz, 2H), 6.96 (s, 1 H), 2.84 (t, J = 7.2 Hz, 4H), 2.59 (t, J = 7.2 Hz, 4H), 2.00 (quin, J = 7.2 Hz, 4H). 5-(Dimethylamino)-yV-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)naphthalene-1 -sulfonamide
Figure imgf000235_0001
A solution of 5-(dimethylamino)naphthalene-1 -sulfonamide (20 mg, 0.08 mmol) in THF (5.0 ml_) was treated with DIPEA (17 μΙ_, 0.09 mmol), stirred at ambient temperature for 15 min, then a solution of 4-isocyanato-1 ,2,3,5,6,7-hexahydro- s-indacene (prepared using general method A2) (19 mg, 0.09 mmol) in THF (1.0 ml_) was added drop wise. The reaction mixture was stirred at ambient temperature overnight then concentrated in vacuo. The crude product was purified by HPLC to give the titled compound as a pale-yellow solid (24 mg, 66%). 1 H NMR (400 MHz, CD3OD) δ = 8.62 (d, J = 8.4 Hz, 1 H), 8.36 (dd, J = 9.5, 8.1 Hz, 2H), 7.67 - 7.56 (m, 2H), 7.26 (d, J = 7.8 Hz, 1 H), 6.91 (s, 1 H), 2.91 (s, 6H), 2.79 (t, J = 7.4 Hz, 4H), 2.40 (t, J= 7.4 Hz, 4H), 1.92 (quin, J= 7.4 Hz, 4H). 13C NMR (101 MHz, CD3OD) δ = 152.5, 150.3, 144.4, 138.3, 131 .9, 131 .7, 131 .1 , 130.3, 130.1 , 129.9, 128.9, 127.2, 123.6, 1 19.4, 1 19.1 , 1 18.9, 1 15.7, 78.4, 78.0, 77.7, 49.6, 48.3, 45.6, 33.4, 33.2, 30.5, 29.3, 25.9, 25.8; HRMS (ESI) calcd. for C25H27N3O3S [M+H] 450.1846, found 450.1859.
/V-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-2,3-dihydro benzo[ ?] thiophene-6-sulf onamide 1 ,1 -dioxide
Figure imgf000235_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 2,3-dihydrobenzo[b]thiophene-6-sulfonamide 1 ,1 -dioxide were used in general method C2 to give the titled compound as a white solid (33 mg, 28%).1 H NMR (600 MHz, DMSO-d6): δ = 8.17 (bs, 1 H), 8.15 (s, 1 H), 8.13 (d, J = 9 Hz, 1H), 7.73 (d, J=12Hz, 1H), 6.89 (s, 1H),3.68 (t, J=9Hz, 2H), 3.43 (t, J = 6 Hz, 2H), 2.75 (t, J = 6 Hz, 4H), 2.55 (t, J = 6 Hz, 4H), 1.93-1.88 (m, 4H). 13C NMR (150 MHz, DMSO-d6): δ = 151.6, 143.3, 143.0, 142.7, 139.6, 137.6, 137.5, 132.2, 128.9, 120.1, 117.9, 50.9, 32.9, 30.6, 25.6, 25.4 LCMS (m/z): 447 [M +H] +
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-2- methoxybenzenesulfonamide
Figure imgf000236_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 2-methoxybenzenesulfonamide were used in general method C6 to give the titled compound as a white solid (30 mg, 48%).1H NMR (400 MHz, CD3OD) δ 7.96 (d, J= 7.9 Hz, 1H), 7.62 (t, J= 8.3 Hz, 1H), 7.14-7.05 (m, 2H), 6.97 (s, 1H), 4.00 (s, 3H), 2.83 (t, J=7.3 Hz, 4H), 2.56 (t, J=7.3 Hz, 4H), 2.09- 1.90 (m,4H).13CNMR(101 MHz, CD3OD) δ 155.9, 143.2, 136.6, 134.7, 129.5, 127.2, 126.6, 125.9, 119.5, 118.0, 111.6, 111.2, 55.3, 31.9, 29.3, 24.5. HRMS (ESI) calcd. for C20H23N2O4S [M+H] 387.1373, found 387.1378.
/V-(1,2,3,5,6,7-Hexahydros-indacen-4-ylcarbamoyl)-3- (trifluoromethyl)benzenesulfonamide
Figure imgf000236_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 3-(trifluoromethyl)benzenesulfonamide were used in general method C4 to give the titled compound as a white solid (0.015 g, 12%); Off white sticky. 1 H NMR (400 MHz, DMSO-d6): δ = 1 1 .01 (bs, 1 H), 8.34 (s, 1 H), 8.25-8.23 (m, 2H), 8.1 1 (d, J = 7.6 Hz, 1 H), 7.90 (t, J = 8.0 Hz, 1 H), 6.93 (s, 1 H), 2.77 (t, J = 7.2 Hz, 4H), 2.50 (m, 4H), 1 .90 (quin, J = 7.6 Hz, 4H). LCMS, Purity: 96.69%, m/z 425.1 (M+H+). HRMS (FAB+) calcd for C20H19F3N2O3S [M+H]+ : 425.1068, found: 425.1009.
/V-(1 ,2,3,5,6,7-hexahydros-indacen-4-ylcarbamoyl)-3- methoxybenzenesulfonamide
Figure imgf000237_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 3-methoxybenzenesulfonamide were used in general method C4 to give the titled compound as an off-white solid (0.025 g, 23%). 1H NMR (400 MHz, DMSO-d6): δ = 10.77 (bs, 1 H), 8.15 (s, 1 H), 7.56-7.45 (m, 3H), 7.27 (d, J = 8.0 Hz, 1 H), 6.93 (s, 1 H), 3.82 (s, 3H), 2.77 (t, J = 7.2 Hz, 4H), 2.53 (t, J = 7.6 Hz, 4H), 1 .92 (quin, J= 7.2 Hz, 4H). LCMS, Purity: 95.02%, tr= 3.77 min, m/z 387.28 (M+H+). HRMS (FAB+) calcd for C20H22N2O4S [M+H]+ : 387.1300, found: 387.1301.
/V-(1 ,2,3,5,6,7-Hexahydros-indacen-4-ylcarbamoyl)-3- (trifluoromethoxy)benzenesulfonamide
Figure imgf000237_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 3-(trifluoromethoxy)benzenesulfonamide were used in general method C4 to give the titled compound as an off-white solid (0.045 g, 43%). 1H NMR (400 MHz, DMSO-d6): δ = 8.04 (d, J = 8.0 Hz, 1 H), 7.95 (s, 1 H), 7.72 (t, J = 8.4 HZ, 1 H), 7.62(d, J = 8.4 Hz, 1 H), 6.95 (s, 1 H), 2.82 (t, J = 7.6 Hz, 4H), 2.59 (t, J= 7.6 Hz, 4H), 1 .99 (quin, J= 7.6 Hz, 4H). 13C NMR (100 MHz, DMSO- d6): δ = 149.2, 147.9, 143.1 , 142.2, 137.3, 131 .5, 128.5, 126.4, 125.9, 121 .2, 1 19.7, 1 18.6, 1 18.1 , 32.5, 29.4, 25.0. 19F NMR (233.33 MHz, DMSO-d6): -57.10 (s, 3F). LCMS, Purity: 95.56%, m/z 441 .20 (M+H+). HRMS (FAB+) calcd for C2oHi9F3N20 S [M+H]+ : 441.1018, found: 441 .1015.
3-(difluoromethoxy)-/V-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)benzenesulfonamide
Figure imgf000238_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 3-(difluoromethoxy)benzenesulfonamide were used in general method C5 to give the titled compound as an off-white solid (0.056 g, 50%). 1H NMR (600 MHz, Acetonitrile-tfs) δ = 7.85 (d, J= 8.0 Hz, 1 H), 7.75 (t, J= 2.1 Hz, 1 H), 7.60 (t, J= 8.0 Hz, 1 H), 7.53 (s, 1 H), 7.43 (dd, J= 8.0, 2.1 Hz, 1 H), 6.95 (s, 1 H), 2.81 (t, J = 7.5 Hz, 4H), 2.55 (t, J = 7.5 Hz, 4H), 1 .95 (quin, J = 7.5 Hz, 4H).
Λ 1-(1 ,2,3,5,6,7-Hexahydros-indacen-4-ylcarbamoyl)benzene-1 ,3- disulfonamide
Figure imgf000238_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and benzene-1 ,3-disulfonamide were used in general method C4 to give the titled compound as a white solid (0.080 g, 12%).1H NMR (400 MHz, DMSO-d6): δ = 11.02 (bs, 1H), 8.36 (s, 1H), 8.24 (s, 1H), 8.15 (d, J= 7.8 Hz, 1H), 8.11 (d, J=8.0 Hz, 1H), 7.84(t, J=8.0 Hz, 1H), 7.63 (s, 2H), 6.93 (s, 1H), 2.77 (t, J= 7.2 Hz, 4H), 2.54 (t, J= 7.6 Hz, 4H), 1.92 (quin, J= 7.2 Hz, 4H).13C NMR (100 MHz, DMSO-d6): δ = 149.0, 144.9, 143.1, 140.9, 137.3, 130.4, 130.2, 128.5, 124.4, 118.1, 32.4, 30.0, 25.0. LCMS, Purity: 98.63%, m/z 436.03 (M+H+). HRMS (FAB+) calcd for C19H21N3O5S2 [M+H]+ : 436.0923, found: 436.0919.
Λ1-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-A^,A^- dimethylbenzene-1 ,3-disulfonamide
Figure imgf000239_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1) and Λ/1,Λ/1 -dimethylbenzene-1 ,3-disulfonamide were used in general method C1 to give the titled compound as a white solid (0.019 g, 5%). 1H NMR (400 MHz, CD3OD) δ 8.41 (t, J= 1.4 Hz, 1H), 8.32 (dt, J=7.9, 1.4 Hz, 1 H), 8.08 (dt, J= 7.9, 1.4 Hz, 1 H), 7.87 (t, J= 7.9 Hz, 1 H), 6.95 (s, 1 H), 2.84 (t, J= 7.4 Hz, 4H), 2.73 (s, 6H), 2.61 (t, J= 7.4 Hz, 4H), 2.00 (p, J= 7.4 Hz, 4H).
3-(N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)sulfamoyl)benzoic acid
Figure imgf000239_0002
Methyl 3-(Λ/-(1 , 2, 3, 5, 6, 7-hexahydro-s-indacen-4- ylcarbamoyl)sulfamoyl)benzoate (0.25 g, 0.603 mmol) was dissolved in a mixture of tetrahydrofuran:methanol:water (9 mL, 1 : 1 : 1 ) and the mixture was cooled to 0 °C. Lithium hydroxide monohydrate (0.75 g, 1.81 mmol, 3 eq) was added and the mixture stirred at ambient temperature for 3h. Upon completion, the reaction mixture was poured into chilled water and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. The product was purified by reverse phase preparative HPLC to afford the titled compound as a white solid (0.017 g, 3%). 1H NMR (400 MHz, DMSO-d6): δ = 13.26 (bs, 1 H), 8.43 (s, 1 H), 8.13-8.08 (m, 2H), 7.99 (bs, 1 H), 7.67 (t, J = 8.0 Hz, 1 H), 6.87 (s, 1 H), 6.52 (s, 1 H), 2.75 (t, J = 7.2 Hz, 4H), 2.55(t, J = 7.6 Hz, 4H), 1 .89 (quin, J = 7.6 Hz, 4H). LCMS, Purity: 96%, m/z 400.98 (M+H+). HRMS (FAB+) calcd for C2oH2oN205S [M+H]+: 401 .1093, found: 401 .4514.
3-(N-(1 , 2, 3, 5, 6, 7-Hexahydro-s-indacen-4- ylcarbamoyl)sulfamoyl)benzamide
Figure imgf000240_0001
3-(N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)sulfamoyl)benzoic acid (0.06 g, 0.074 mmol) was dissolved in anhydrous A/,A/-dimethylformamide (4 mL) and the solution cooled to 0 °C. Diisopropylethylamine (3.0 eq) and HATU (2.0 eq) were added and the mixture stirred at 0 °C for 15 min. Ammonium chloride (3.0 eq) was added and the mixture stirred at ambient temperature for 5 h. Upon completion the reaction mixture was poured into brine (20 mL) and extracted with ethyl acetate (2 x 10 mL). The combined organic extracts were washed with brine (10 mL), dried (Na2S04) and concentrated in vacuo. The crude residue was purified by reverse phase preparative HPLC to afford the titled compound as a white solid (0.01 1 g, 37%). 1H NMR (400 MHz, DMSO-d6): δ = 8.23 (d, J = 9.2 Hz, 2H), 8.02 (s, 1 H), 7.89 (d, J = 7.6 Hz, 1 H), 7.84 (d, J = 7.6 Hz, 1 H), 7.42 (t, J = 8.0 Hz, 1 H), 7.38 (s, 1 H), 7.33 (s, 1 H). 6.74 (s, 1 H), 2.73 (t, J = 6.8 Hz, 4H), 2.62 (t, J = 6.8 Hz, 4H), 1 .87 (quin, J = 7.6 Hz, 4H). LCMS, Purity: 93%, m/z 400.05 (M+H+). HRMS (FAB+) calcd for C20H21 N3O4S [M+H]+: 400.1253, found: 400.1378. yV-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-3-(2-hydroxypropan-2- yl)benzenesulfonamide
Figure imgf000241_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) was added directly to methyl 3-sulfamoylbenzoate (0.447 g, 2.07 mmol, 1 .20 equiv) at ambient temperature and the mixture was stirred overnight. The reaction mixture was poured into chilled water and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. The residue obtained was purified by column chromatography on silica gel using 0-10% gradient of methanol in dichloromethane to give methyl 3-(A/-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)benzoate as a light-brown solid (0.36 g, 50%).
Methyl 3-(A/-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)benzoate (0.06 g, 0.144 mmol) was dissolved in anhydrous THF and the solution cooled to 0 °C. Methyl magnesium bromide (3 M solution in diethyl ether, 0.14 mL, 0.42 mmol, 3.0 eq) was added and the mixture stirred at ambient temperature for 4 h. Upon completion, saturated aqueous ammonium chloride was added to the reaction mixture and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. Purification of the crude residue by reverse phase preparative HPLC gave the titled compound as an off-white solid (0.015 g, 25%).1H NMR (400 MHz, DMSO-d6): δ = 8.16 (s, 1H), 7.91 (s, 1H), 7.62 (d, J= 7.2 Hz, 1H), 7.59-7.48 (m, 2H), 7.32 (t, J= 7.6 Hz, 1H), 6.78 (s, 1H), 5.10 (s, 1H), 2.74 (t, J= 7.2 Hz, 4H), 2.60 (t, J= 6.8 Hz, 4H), 1.88 (quin, J = 7.6 Hz, 4H), 1.42(s, 6H). LCMS, Purity: 91 %, m/z 415.05 (M+H+).
3-Azido-N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)benzenesulfonamide
Figure imgf000242_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 3-azidobenzenesulfonamide were used in general method C6 to give the titled compound as an off white solid (70 mg, 50%).1H NMR (600 MHz, DMSO-de) δ = 8.22 (s, 1H), 7.72 (m, J= 5.2 Hz, H), 7.65 (t, J= 8.0 Hz, 1H), 7.59 (s, 1H), 7.46 - 7.42 (m, 1H), 6.93 (s, 1H), 2.77 (t, J= 7.4 Hz, 4H), 2.53 (t, J= 7.4 Hz, 4H), 1.92 (m, 4H).13C NMR (101 MHz, CD3OD) δ 151.2, 144.9, 144.6, 142.9, 142.5, 138.9, 131.5, 131.4, 128.6, 124.9, 124.6, 124.6, 119.8, 119.2, 118.9, 111.9, 33.7,33.6, 31.1,29.7,26.3. HRMS (ESI) calcd. for C19H20N5O3S [M+H] 398.1281, found 398.1272.
N-((1 ,2,3,5,6,7-Hexahydro-s-indacen-4-yl)carbamoyl)-3-(4-phenyl-1 H-1 ,2,3- triazol-1-yl) benzenesulfonamide
Figure imgf000242_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 3-(4-phenyl-1 H-1 ,2,3-triazol-1 -yl)benzenesulfonamide were used in general method C6 to give the titled compound as a pale yellow solid (10 mg, 49%). 1 H N MR (400 MHz, CD3OD) δ = 8.89 (s, 1 H), 8.55 (s, 1 H), 8.21 (d, J = 8.2 Hz, 1 H), 8.13 (d, J = 7.8 Hz, 1 H), 7.92 (d, J = 7.6 Hz, 2H), 7.79 (t, J = 8.0 Hz, 1 H), 7.68 (s, 1 H), 7.48 (t, J = 7.6 Hz, 2H), 7.39 (t, J = 7.4 Hz, 1 H), 6.92 (s, 1 H), 2.82 (t, J = 7.4 Hz, 4H), 2.70 - 2.63 (m, 4H), 1 .98 (m, 4H). 13C NMR (151 MHz, CD3OD) 5 = 148.8, 143.9, 143.6, 137.7, 137.2, 137.0, 130.6, 130.3, 129.7, 129.6, 128.8, 128.5, 127.6, 127.4, 126.7, 125.6, 124.5, 124.0, 1 19.3, 1 18.7, 1 10.8, 32.7, 32.6, 30.2, 28.7, 25.3. HRMS (ESI) calcd. for C27H26N5O3S [M+H] 500.1751 , found 500.1735.
N-(3-(N-(( ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)phenyl)pent-4-ynamide
Figure imgf000243_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and A/-(3-sulfamoylphenyl)pent-4-ynamide were used in general method C6 to give the titled compound as a white solid (1 16 mg, 61 %). 1 H NMR (400 MHz, CD3OD) δ = 8.18 (s, 1 H), 7.81 (d, J = 8.3 Hz, 1 H), 7.68 (d, J = 8.3 Hz, 1 H), 7.43 (dd, J = 8.3, 7.8 Hz, 1 H), 6.87 (s, 1 H), 2.79 (t, J = 7.2 Hz, 4H), 2.67 - 2.60 (m, 4H), 2.60 - 2.48 (m, 4H), 2.28 - 2.22 (m, 1 H), 2.04 - 1 .89 (m, 4H). 13C NMR (101 MHz, CD3OD) δ = 170.9, 143.3, 143.0, 138.8, 137.7, 128.7, 128.3, 126.4, 122.8, 122.0, 1 17.9, 1 17.7, 82.0, 68.9, 35.4, 32.4, 29.9, 25.1 , 13.9. HRMS (ESI) calcd. for C24H26N3O4S [M+H] 452.1639, found 452.1658.
3-(1-(3-aminopropyl)-1 Η-Λ ,2,3-triazol-4-yl)-AA(3-(A/-((1 ,2,3,5,6,7-hexahydro-s- indacen-4-yl)carbamoyl)sulfamoyl)phenyl)propanamide
Figure imgf000244_0001
A/-(3-(A/-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)phenyl)pent-4-ynamide and 3-azidopropan-1 -amine were used in general method F to give the titled compound as a white solid (6 mg, 43%). 1H NMR (600 MHz, CD3OD) δ = 7.85 (s, 1 H), 7.55 (t, J= 3.8 Hz, 2H), 7.50 (d, J = 8.0 Hz, 2H), 7.29 (t, J = 7.9 Hz, 1 H), 6.78 (s, 1 H), 4.26 (t, J = 6.4 Hz, 2H), 3.00 (t, J = 6.6 Hz, 2H), 2.71 (t, J = 7.3 Hz, 4H), 2.64 - 2.50 (m, 8H), 1 .94-2.02 (m, 2H), 1 .92 - 1.83 (m, 4H). 13C NMR (151 MHz, CD3OD) δ = 173.0, 147.4, 146.8, 144.7, 144.6, 139.5, 139.2, 131 .6, 130.0, 129.8, 124.2, 123.9, 123.2, 1 19.5, 1 18.6, 48.3, 37.7, 34.0, 31 .6, 26.7, 26.6, 22.9. HRMS (ESI) calcd. for C27H34N7O4S [M+H] 552.2387, found 552.2368.
N-(3-(N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)phenyl)-3-(1-(3-((7-nitrobenzo[c][1 ,2,5]oxadiazol-4- yl)amino)propyl)-1 H-1 ,2,3-triazol-4-yl)propanamide
Figure imgf000244_0002
A/-(2-Azidopropyl)-7-nitrobenzo[c][1 ,2,5]oxadiazol-4-amine was synthesized by the methods contained in Chun Li, Etienne Henry, Naresh Kumar Mani, Jie Tang, Jean-Claude Brochon, Eric Deprez, and Juan Xie Eur. J. Org. Chem. 2010, 2395-2405. To a solution of 4-chloro-7-nitrobenzo[c][1 ,2,5]oxadiazole (300 mg, 1.5 mmol) in THF (10 ml_) was added 3-azidopropyl amine (160 mg, 1 .65 mmol) and CS2CO3 (480 mg, 1 .5 mmol). The reaction mixture was stirred at 50 °C for 4 h. The reaction mixture was partitioned between EtOAc (50 mL) concentrated in vacuo. The residue was purified by column chromatography on silica gel using 30% EtOAc-petroleum ether eluent to afford A/-(2-Azidopropyl)- 7-nitrobenzo[c][1 ,2,5]oxadiazol-4-amine (240 mg, 76%). 1 H NMR (400 MHz, CDCI3): δ = 8.50 (d, J = 8.8 Hz, 1 H), 6.57 (s, 1 H, NH), 6.23 (d, J = 8.8 Hz, 1 H), 3.66 (q, J = 6.8 Hz, 2 H),3.59 (J = 6.0 Hz, 2 H), 2.00-2.1 6 (m, 2H). 13C NMR (101 MHz, CDCI3) δ 144.2, 144.0, 143.8, 136.7, 123.7, 98.8, 49.1 , 41 .6, 27.6. HRMS (ESI): calcd. for C9H10N7O3 264.0840; found 264.071 1 .
A/-(3-(A/-((1 ,2,3,5,6,7-Hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)phenyl)pent-4-ynamide (10 mg, 0.022 mmol) and A/-(2- azidopropyl)-7-nitrobenzo[c][1 ,2,5]oxadiazol-4-amine (7.0 mg, 0.026 mmol), 10 mol% THPTA, 5 mol% CuS04, 10 mol% sodium ascorbate in DMSO (500 uL) were stirred at room temperature for 12 h. The reaction mixture was subjected to purification using reverse phase (Reveleris flash column chromatography, 4 g, 18 mL/min. , mobile phase; 10 mmol aqu. NH4C03, MeCN) and freeze dried to give the product as a white solid (7.0 mg, 44 %). 1 H NMR (600 MHz, CD3OD) δ = 8.46 (d, J = 8.7 Hz, 1 H), 8.18 (s, 1 H), 7.79 (d, J = 8.4 Hz, 1 H), 7.67 (d, J = 7.9 Hz, 1 H), 7.61 (s, 1 H), 6.94 (s, 1 H), 6.15 (d, J = 9.0 Hz, 1 H), 4.46 (t, J = 6.7 Hz, 2H), 3.09 (t, J = 7.0 Hz, 2H), 2.82 (t, J = 7.4 Hz, 4H), 2.77 (t, J = 7.0 Hz, 2H), 2.70 - 2.56 (m, 6H), 2.37 - 2.26 (m, 2H), 1 .99 (q, J = 7.3 Hz, 4H). 13C NMR (151 MHz, CD3OD) δ = 172.9, 147.9, 145.4, 140.5, 139.0, 138.4, 130.6, 129.1 , 128.0, 125.6, 124.2, 123.6, 120.3, 1 19.6, 1 12.4, 70.6, 48.9, 37.2, 34.3, 34.2, 31 .7, 30.2, 26.8, 22.3; HRMS (ESI) calcd. for C33H34Nio07S [M-H] 713.2260, found 713.2290. yV-(3-(4-(3-((3-(yV-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)phenyl) amino)-3-oxopropyl)-1 Η-Λ ,2,3-triazol-1 - yl)propyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1 H-thieno[3,4-d]imidazol-4- yl)pentanamide
Figure imgf000246_0001
To a solution of biotin (0.4 g, 1 .63 mmol) and 3-azidopropylamine (0.2 g, 1 .96 mmol) in dry DMF (10.0 ml) was added HBTU (0.93 g, 2.45 mmol) followed by DIPEA (428 uL, 2.45 mmol). The reaction mixture was stirred at RT for 12 h. The reaction was monitored by LCMS and after the completion of reaction, it was diluted with EtOAc (50 mL) washed with H20 (25 mL), brine (25 mL). The organic layer was separated; dried (MgS04) and evaporated to give the crude product. The crude product was purified by column chromatography on silica gel using 50% EtOAc-Hexane eluent to isolate A/-(3-azidopropyl)-5- ((3aS,4S,6aR)-2-oxohexahydro-1 H-thieno[3,4-d]imidazol-4-yl)pentanamide as a white solid (0.13 g, 24%). H NMR (400 MHz, CD3OD) δ = 4.52 (dd, J = 7.9, 5.0 Hz, 1 H), 4.32 (dd, J = 7.9, 4.5 Hz, 1 H), 3.36 (t, J = 6.7 Hz, 2H), 3.28 (d, J= 6.8 Hz, 2H), 3.21 - 3.14 (m, 1 H), 2.93 (dd, J = 12.8, 5.0 Hz, 1 H), 2.75 (d, J = 12.8 Hz, 1 H), 2.20 (t, J = 7.3 Hz, 2H), 1.78 (q, J = 6.8 Hz, 2H), 1 .74 - 1 .57 (m, 4H), 1 .45 (q, J = 7.5 Hz, 2H). 13C NMR (101 MHz, CD3OD) δ = 173.5, 163.4, 61 .0, 59.3, 54.7, 48.2, 39.4, 35.8, 34.8, 27.7, 27.5, 27.2, 24.6.
A/-(3-(A/-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)phenyl)pent-4-ynamide (1 .0 mmol) and N-(3- azidopropyl)-5-((3aS,4S,6aR)-2-oxohexahydro-1 H-thieno[3,4-d]imidazol-4- yl)pentanamide (2.0 mmol), 10 mol% THPTA, 5 mol% CuS0 , 10 mol% sodium ascorbate in DMSO were stirred at room temperature for 12 h. The reaction mixture was purified using reverse phase column chromatography to give the titled compound as a white solid (8.0 mg, 31 %); 1H NMR (600 MHz, CD3OD) δ = 8.26 (s, 1 H), 7.83 - 7.68 (m, 3H), 7.50 - 7.43 (m, 1 H), 6.92 (s, 1 H), 4.48 (dd, J = 8.0, 4.8 Hz, 1H), 4.41 -4.22 (m, 3H), 3.18 (dd, J = 6.9, 3.5 Hz, 1H), 3.14 (td, J= 6.7, 1.7 Hz, 2H), 3.12 - 3.06 (m, 2H), 2.90 (dd, J= 12.8, 4.9 Hz, 1H), 2.81 (t, J= 7.7 Hz, 4H), 2.77 (d, J= 7.1 Hz, 1H), 2.71 (s, 1H), 2.62 (t, J= 7.3 Hz, 4H), 2.19 (td, J= 7.4, 1.7 Hz, 2H), 2.05 - 2.01 (m, 2H), 2.00 - 1.95 (m, 4H), 1.76-1.57 (m, 4H), 1.43 (q, J=7.6, 7.1 Hz, 2H).13C NMR(151 MHz, CD3OD) 5 = 174.8, 174.8, 171.6, 171.5, 164.5, 146.2, 143.6, 139.1, 137.7, 129.1, 129.1, 128.7, 128.1, 126.5, 123.7, 122.9, 122.4, 122.2, 120.9, 118.4, 118.4, 118.3, 118.2, 118.2, 117.2, 110.5, 69.0,61.9, 60.2, 55.6, 39.8, 36.1, 36.0, 35.8, 35.4, 35.4, 32.6, 32.6, 30.0, 29.7, 29.7, 28.6, 28.3, 28.0, 25.3, 25.2, 25.2, 20.9. HRMS (ESI) calcd. for
Figure imgf000247_0001
778.3163, found 778.3145.
N-((1 -(3-(N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)phenyl)-1H-1,2,3-triazol-4-yl)methyl)-5-
((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)pentanamide
Figure imgf000247_0002
5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-N-((1-(3- sulfamoylphenyl)-1 H-1 ,2,3-triazol-4-yl)methyl)pentanamide was synthesized using 5-((3aS,4S,6aR)-2-oxohexahydro-1H-thieno[3,4-d]imidazol-4-yl)-N-(prop- 2-yn-1 -yl)pentanamide (1.0 mmol) and 3-azidobenzenesulfonamide (2.0 mmol), 10 mol% THPTA, 5 mol% CuS04, 10 mol% NaAsc in DMSO were stirred at room temperature for 12 h. The formation of product was observed in LCMS. After completion of the reaction, the reaction mixture was subjected to HPLC purification (Reveleris flash column chromatography, 4 g, 18 mL/min., mobile phase; 10 mmol aq. NH C03, MeCN) to isolate 5-((3aS,4S,6aR)-2- oxohexahydro-1 H-thieno[3,4-d]imidazol-4-yl)-N-((1 -(3-sulfamoylphenyl)-1 H- 1 ,2,3-triazol-4-yl)methyl)pentanamid as a white solid (24 mg, 47%) which was used directly.
To a solution of 5-((3aS,4S,6aR)-2-oxohexahydro-1 H-thieno[3,4-d]imidazol-4- yl)-N-((1 -(3-sulfamoylphenyl)-1 H-1 ,2,3-triazol-4-yl)methyl)pentanamide (15 mg, 0.031 mmol) in THF (5.0 mL) under a nitrogen atmosphere was added DIPEA (605 μΙ_, 0.037 mmol). This mixture was stirred at room temperature for 15 min. A solution of 4-isocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) (705 mg, 0.037 mmol) in THF was added drop-wise. The reaction mixture was stirred at room temperature overnight then the solvent was removed in vacuo to give crude compound which was purified by reversed phase column chromatography using 10 mM aq. (NH4)2C03 and MeCN mobile phase to isolate the titled compound as a white solid (5.2 mg, 24%). 1 H NMR (600 MHz, CD3OD) δ = 8.53 (d, J= 2.4 Hz, 1 H), 8.50 (d, J= 7.5 Hz, 1 H), 8.22 - 8.1 1 (m, 2H), 7.86 - 7.78 (m, 1 H), 6.99 (s, 1 H), 4.62 (s, 2H), 4.57 - 4.49 (m, 1 H), 4.38 - 4.31 (m, 1 H), 3.27 - 3.20 (m, 1 H), 3.00 - 2.84 (m, 4H), 2.78 - 2.70 (m, 4H), 2.36 (t, J = 7.2 Hz, 2H), 2.06 (q, J = 7.4 Hz, 4H), 1 .83 - 1 .73 (m, 3H), 1 .71 - 1 .63 (m, 1 H), 1.54 - 1.43 (m, 3H). 13C NMR (151 MHz, CD3OD) δ = 174.6, 164.5, 146.1 , 145.4, 143.6, 137.7, 137.2, 130.5, 125.9, 123.4, 121 .1 , 1 17.8, 1 10.6, 61 .8, 60.2, 55.5, 47.7, 47.6, 39.8, 3.2, 34.3, 32.6, 30.14, 28.2, 27.9, 25.3, 25.2. HRMS (ESI) calcd. for C32H39N8O5S2 [M+H] 679.2479, found 679.2456.
A-(quinolin-6-ylcarbamoyl)-3-(3-(trifluoromethyl)-3H-diazirin-3- yl)benzenesulfonamide
Figure imgf000248_0001
Phenyl chloroformate (1 eq) was added to a solution of quinolin-6-amine (0.1 g, 0.69 mmol) in THF (50 mL) and triethylamine (1.5 eq.) to 0 °C. The solution was diluted using water, extracted using ethyl acetate (x2), washed with water, brine then dried (Na2S04) and concentrated in vacuo. The crude product was triturated with pentane to give phenyl quinolin-6-ylcarbamate as an off-white solid which was used directly in the next reaction step.
3-(3-(trifluoromethyl)-3H-diazirin-3-yl)benzenesulfonamide (0.185 g, 0.69 mmol) in THF (30 ml_) at 0 °C was treated portion-wise with sodium hydride (3 eq.) and the suspension stirred for 30 minutes (until effervescence ceased). The crude phenyl quinolin-6-ylcarbamate was dissolved in THF (20 ml_) then added slowly to the reaction and stirring continued at ambient temperature until completion, typically 2 h. The reaction was quenched with sat.aq. NH4CI, extracted with ethyl acetate (x2), washed with water (100 ml_), brine (100 ml_), dried (Na2S04) and concentrated in vacuo. The crude product was triturated using diethyl ether then pentane to give the titled compound, A/-(quinolin-6-ylcarbamoyl)-3-(3- (trifluoromethyl)-3 - -diazirin-3-yl)benzenesulfonamide as a white solid (10 mg, 3%). 1 H NMR (400 MHz, DMSO-afe) δ = 8.88 (s, 1 H), 8.59 (d, J = 3.5 Hz, 1 H), 8.1 1 (s, 1 H), 8.04 (d, J = 8.3 Hz, 1 H), 7.96 (d, J = 7.9 Hz, 1 H), 7.74 (d, J = 9.1 Hz, 1 H), 7.70 - 7.60 (m, 2H), 7.56 (t, J = 7.8 Hz, 1 H), 7.34 (dd, J = 8.3, 4.2 Hz, 1 H), 7.30 (d, J = 8.0 Hz, 1 H). 19F NMR (376 MHz, DMSO-afe) δ -64.49. yV-(quinolin-5-ylcarbamoyl)-3-(3-(trifluoromethyl)-3H-diazirin-3- yl)benzenesulfonamide
Figure imgf000249_0001
A/-(quinolin-5-ylcarbamoyl)-3-(3-(trifluoromethyl)-3 - -diazirin-3- yl)benzenesulfonamide was synthesised using modification of the procedures used to make N-(quinolin-6-ylcarbamoyl)-3-(3-(trifluoromethyl)-3H-diazirin-3- yl)benzenesulfonamide but using quinolin-5-amine in place of quinolin-6-amine. The titled compound was obtained as an off-white solid (10 mg, 3%). 1H NMR (400 MHz, DMSO-afe) δ = 8.79 (d, J= 4.1 Hz, 1 H), 8.61 (s, 1 H), 8.57 (d, J= 8.7 Hz, 1 H), 7.95 (d, J = 7.8 Hz, 1 H), 7.88 (d, J = 7.2 Hz, 1 H), 7.70 (s, 1 H), 7.59 - 7.50 (m, 2H), 7.40 (dd, J = 8.7, 4.1 Hz, 1 H), 7.29 (d, J = 7.9 Hz, 1 H). 19F NMR (376 MHz, DMSO-afe) δ -64.51 .
A^((6-methoxyquinolin-8-yl)carbamoyl)-3-(3-(trifluoromethyl)-3H-diazirin-3- yl)benzenesulfonamide
Figure imgf000250_0001
A/-((6-methoxyquinolin-8-yl)carbamoyl)-3-(3-(trifluoromethyl)-3H-diazinn-3- yl)benzenesulfonamide was synthesised using modification of the procedures used to make A/-(quinolin-6-ylcarbamoyl)-3-(3-(trifluoromethyl)-3 - -diazirin-3- yl)benzenesulfonamide but using 6-methoxyquinolin-8-amine in place of quinolin-6-amine. The titled compound was obtained as an off-white solid (35 mg, 20%). 1H NMR (400 MHz, CD3OD) δ 8.64 (dd, J = 4.2, 1 .6 Hz, 1 H), 8.18 - 8.02 (m, 3H), 7.88 (s, 1 H), 7.60 (t, J= 7.9 Hz, 1 H), 7.50 - 7.36 (m, 2H), 6.79 (d, J = 2.6 Hz, 1 H), 3.88 (s, 3H). 19F NMR (376 MHz, CD3OD) δ -67.04.
A-(quinolin-8-ylcarbamoyl)-3-(3-(trifluoromethyl)-3H-diazirin-3- yl)benzenesulfonamide
Figure imgf000250_0002
A/-(Quinolin-8-ylcarbamoyl)-3-(3-(trifluoromethyl)-3H-diazirin-3- yl)benzenesulfonamide was synthesised using modification of the procedures used to make A/-(quinolin-6-ylcarbamoyl)-3-(3-(trifluoromethyl)-3 - -diazirin-3- yl)benzenesulfonamide but using quinolin-8-amine in place of quinolin-6-amine. The titled compound was obtained as a white solid (20 mg, 16%). 1H NMR (400 MHz, CD3OD) δ = 8.82 (dd, J = 4.3, 1.6 Hz, 1 H), 8.35 (dd, J = 7.4, 1 .8 Hz, 1 H), 8.21 (dd, J = 8.3, 1 .7 Hz, 1 H), 8.12 (d, J = 7.9 Hz, 1 H), 7.88 (s, 1 H), 7.59 (t, J = 7.9 Hz, 1 H), 7.52 - 7.38 (m, 4H). yV-((2,3,6,7-tetrahydrobenzo[1 ,2-b:4,5-b']difuran-4-yl)carbamoyl)-3-(3- (trifluoromethyl)-3H-diazirin-3-yl)benzenesulfonamide
Figure imgf000251_0001
4-isocyanato-2,3,6,7-tetrahydrobenzo[1 ,2-£>:4,5-£>']difuran (prepared using general method A1 ) and 3-(3-(trifluoromethyl)-3H-diazirin-3- yl)benzenesulfonamide were used in general method C1 to give the titled compound as a white solid (0.01 g, 2%). 1H NMR (400 MHz, CD3OD) δ = 8.06 (dt, J = 7.9, 1 .3 Hz, 1 H), 7.80 (s, 1 H), 7.57 (t, J = 7.9 Hz, 1 H), 7.44 (d, J = 7.9 Hz, 1 H), 6.39 (s, 1 H), 4.49 (t, J = 8.6 Hz, 2H), 4.42 (t, J= 8.6 Hz, 2H), 3.09 (t, J = 8.4 Hz, 2H), 3.02 (t, J = 8.6 Hz, 2H). 19F NMR (376 MHz, CD3OD) δ -67.06.
4-chloro-N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)benzenesulfonamide
Figure imgf000251_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 4-chlorobenzenesulfonamide were used in general method C2 to give the titled compound as a white solid (48 mg, 43%). 1 H NMR (400 MHz, DMSO-d6): δ = 8.13 (s, 1 H), 7.93 (d, J = 8.0 Hz, 2H), 7.68 (d, J = 12 Hz, 2H), 6.92 (s, 1 H), 2.77 (t, J= 8.0 Hz, 4H), 2.54 (t, J= 8.0 Hz, 4H), 1.95-1.88 (m, 4H); 13C NMR (100 MHz, DMSO-d6): δ = 150.1 , 143.4, 139.9, 138.1 , 137.6, 129.6, 129.4, 129.2, 32.8, 30.5, 25.9; LCMS purity: >95%; LCMS (m/z): 391 [M +H]+; HRMS calculated for C19H19CIN2O3S [M+H]+: 391 .0878, found: 391.0895.
/V-(1 ,2,3,5,6,7-hexahydro-s-indacen-4-ylcarbamoyl)-4- methylbenzenesulfonamide
Figure imgf000252_0001
4- lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 4-methylbenzenesulfonamide were used in general method C4 to give the titled compound as a white solid (0.045 g, 27%). 1H NMR (400 MHz, DMSO-d6): δ = 10.70 (br.s, 1 H), 8.08 (s, 1 H), 7.82 (d, J= 8 Hz, 2H), 7.41 (d, J = 8.0 Hz, 2H), 6.92 (s, 1 H), 2.79-2.68 (m, 4H), 2.58-2.50 (m, 4H), 2.39 (s, 3H), 1 .97-1 .87 (m, 4H). 13C NMR (100 MHz, DMSO-d6): δ = 149.0, 143.6, 143.0, 137.1 , 129.4, 128.6, 127.3, 1 1 7.9, 32.4, 30.0, 25.0, 21 .0. LCMS, Purity: 95.08%, m/z 371 .07 (M+H+). HRMS (FAB+) calcd for C20H22N2O3S [M+H]+ : 371 .1351 , found: 371 .1419.
5- Chloro-/V-(4-(A -(1 ,2,3,5,6,7-hexahydro-s-indacen-4- ylcarbamoyl)sulfamoyl)phenethyl)-2-methoxybenzamide
Figure imgf000252_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 5-chloro-2-methoxy-N-(4-sulfamoylphenethyl)benzamide were used in general method C4 to give the titled compound as a white solid (45 mg, 10%). 1 H NMR (400 MHz, DMSO-d6): δ = 10.73 (s, 1 H), 8.27 (t, J= 5.2 Hz, 1 H), 8.09 (s, 1 H), 7.89 (d, J = 8.4 Hz, 2H), 7.65 (d, J = 2.4 Hz, 1 H), 7.50 (d, J = 8.4 Hz, 2H), 7.49 (d, J = 2.4 Hz, 1 H), 7.13 (d, J = 9.2 Hz, 1 H), 6.92 (s, 1 H), 3.78 (s, 3H), 3.54 (q, J = 6.4 Hz, 2H), 2.94 (t, J = 6.8 Hz, 2H), 2.75 (t, J = 7.2 Hz, 4H), 2.50 (m, 4H), 1 .89 (quin, J = 7.6 Hz, 4H). 13C NMR (100 MHz, DMSO-d6): δ = 163.6, 155.7, 145.3, 143.6, 143.0, 142.4, 142.1 , 139.6, 137.1 , 131.5, 129.5, 129.2, 127.4, 125.7, 124.8, 124.3, 1 17.9, 1 14.1 , 108.3, 56.2, 34.7, 32.6, 32.4, 30.0, 28.9, 24.9. LCMS, Purity: 90.06%, tr = 3.38 min, m/z 566.37 (M-H+). HRMS (FAB+) calcd for C29H30CIN3O5S [M+H]+ : 568.1595, found: 568.1589. yV-(4-(yV-(1 ,2,3,5,6,7-Hexahydros-indacen-4- ylcarbamoyl)sulfamoyl)phenethyl)-5-methylpyrazine-2-carboxamide
Figure imgf000253_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 5-methyl-N-(4-sulfamoylphenethyl)pyrazine-2-carboxamide were used in general method C4 to give the titled compound as an off-white solid (0.02 g, 4%). 1H NMR (400 MHz, DMSO-d6): δ = 10.71 (s, 1 H), 9.02 (s, 1 H), 8.96 (t, J = 6 Hz, 1 H), 8.59 (s, 1 H), 8.07 (s, 1 H), 7.85 (d, J = 8.4 Hz, 2H), 7.47 (d, J = 8.0 Hz, 2H), 6.92 (s, 1 H), 3.57 (q, J = 6.8 Hz, 2H), 2.97 (t, J = 7.4 Hz, 2H), 2.82-2.73 (m, 4H), 2.53 (s, 3H), 2.57-2.50 (m, 4H), 1 .97-1 .84 (m, 4H). LCMS, Purity: 88.15%, m/z 520.28 (M+H+). HRMS (FAB+) calcd for C27H29N5O4S [M+H]+ : 520.1940, found: 520.1977. 3-(4-(N-((1 ,2,3,5,6,7-Hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)phenyl)-N-(prop-2-yn-1-yl)propanamide
Figure imgf000254_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and N-(prop-2-yn-1 -yl)-3-(4-sulfamoylphenyl)propanamide were used in general method C6 to give the titled compound as a white solid (120 mg, 68%). 1H NMR (400 MHz, CD3OD) δ = 7.91 (d, J= 7.8 Hz, 2H), 7.39 (d, J = 7.8 Hz, 2H), 6.98 (s, 1 H), 3.95 (d, J= 2.9 Hz, 2H), 3.03 (t, J= 7.7 Hz, 2H), 2.85 (t, J = 7.4 Hz, 4H), 2.62 (t, J= 6.9 Hz, 4H), 2.55 - 2.46 (m, 2H), 2.25 (t, J= 2.6 Hz, 1 H), 2.02 (m, 4H). 13C NMR (101 MHz, CD3OD) δ = 172.0, 147.2, 144.1 , 143.8, 137.5, 129.0, 128.8, 128.1 , 127.4, 126.5, 1 18.9, 79.2, 71 .0, 36.8, 32.8, 32.8, 31 .2, 30.7, 28.8, 28.7, 25.4, 25.3. HRMS (ESI) calcd. for C25H28N3O4S [M+H] 466.1795, found 466.1794.
A^(4-(A^(1 ,2,3,5,6,7-Hexahydros-indacen-4- ylcarbamoyl)sulfamoyl)phenethyl)-2-(methyl(7- nitrobenzo[c][1 ,2,5]oxadiazol-4-yl)amino)acetamide
Figure imgf000254_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 2-(methyl(7-nitrobenzo[c][1 ,2,5]oxadiazol-4-yl)amino)-A/-(4- sulfamoylphenethyl)acetamide were used in general method C4 to give the titled compound as an orange solid (0.003 g, 1%).1H NMR (400 MHz, DMSO- d6): δ = 10.74 (s, 1H), 8.51 (d, J= 8.8 Hz, 1H), 8.31 (t, J= 7.6 Hz, 1H), 8.09- 7.96 (m, 1H), 7.82 (d, J= 8.4Hz, 2H), 7.41 (d, J= 7.6 Hz, 2H), 6.89 (s, 1H), 6.42-6.32 (m, 1H), 4.74(bs, 2H), 3.44-3.30 (m, 5H), 2.80 (t, J= 7.6 Hz, 2H), 2.73-2.69 (m, 4H), 2.61 -2.50 (m, 4H), 1.92-1.88 (m, 4H). LCMS, Purity: 92.20%, m/z 632.35 (M-H+).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2-((7- nitrobenzo[c][1,2,5]oxadiazol-4-yl)amino)ethyl)benzenesulfonamide
Figure imgf000255_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1) and 4-(2-((7-nitrobenzo[c][1 ,2,5]oxadiazol-4- yl)amino)ethyl)benzenesulfonamide were used in general method C4 to give the titled compound as a yellow solid (0.047 g, 15%). 1H NMR (400 MHz, DMSO-d6): δ = 10.69 (bs, 1H), 9.55 (s, 1H), 8.50 (d, J= 8.8 Hz, 1H), 8.09 (s, 1 H), 7.87 (d, J= 8.0 Hz, 2H), 7.56 (d, J= 8.0 Hz, 2H), 6.92 (s, 1 H), 6.50 (d, J = 8.8 Hz, 1H), 3.76 (bs, 2H), 3.11 (t, J= 6.8 Hz, 2H), 2.76 (t, J= 7.6 Hz, 4H), 2.53(t, J=6.8Hz, 4H), 1.90 (quin, J= 7.6 Hz, 4H).13C NMR (100 MHz, DMSO- d6): 5 = 149.1, 144.8, 144.3, 142.4, 138.3, 137.8, 137.1, 129.3, 128.6, 127.3, 125.7, 121.0, 117.9, 108.3, 99.5, 44.1, 33.2, 32.5, 30.1, 28.9, 25.0. LCMS, Purity: 96.50%, tr = 2.29 min, m/z 563.20 (M+H+). HRMS (FAB+) calcd for CayHaeNeOeS [M+H]+ : 563.1635, found: 563.1641.
2-(7-(Dimethylamino)-2-oxo-2H-chromen-4-yl)-yV-(4-(A/-(1,2,3,5,6,7- hexahydro-s-indacen-4-ylcarbamoyl)sulfamoyl)phenethyl)acetamide
Figure imgf000256_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 2-(7-(dimethylamino)-2-oxo-2H-chromen-4-yl)-N-(4- sulfamoylphenethyl)acetamide were used in general method C4 to give the titled compound as a pale-yellow solid (0.008 g, 0.44%). 1 H NMR (400 MHz, DMSO-d6): δ = 9.53 (s, 1 H), 8.29 (t, J = 4.8 Hz, 1 H), 8.22 (s, 1 H), 7.68 (d, J = 8.0 Hz, 2H), 7.46 (d, J = 8.8 Hz, 1 H), 8.42 (s, 1 H), 7.16 (d, J = 8.0 Hz, 2H), 6.74-6.70 (m, 2H), 6.54 (d J= 2.4 Hz, 1 H), 5.99 (s, 1 H), 3.56-3.52 (m, 2H), 3.48 (t, J= 6.0 Hz, 2H), 3.31 -3.24 (m, 2H), 2.76-2.70 (m, 4H), 3.02 (s, 6H), 2.63 (t, J = 7.2 Hz, 4H), 1 .88 (quin, J = 7.6 Hz, 4H). LCMS, Purity: 92.26%, m/z 629.40 (MH+). iV-((1 , 2,3,5,6 ,7-hexahydro-s-indacen-4-yl)carbamoyl)benzo[d|[1 ,3]dioxole-5- sulfonamide
Figure imgf000256_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and benzo[ ][1 ,3]dioxole-5-sulfonamide were used in general method C2 to give the titled compound as a white solid (28 mg, 27%). 1 H NMR (400 MHz, DMSO-d6): δ = 8.04 (br.s, 1 H), 7.47 (d, J= 8.0 Hz, 1 H), 7.38 (s, 1 H), 7.09 (d, J = 4.0 Hz, 1 H), 6.91 (s, 1 H), 6.16 (s, 2H) 2.77 (t, J = 8.0 Hz, 4H), 2.56 (t, J= 8.0 Hz, 4H), 1 .96-1 .89 (m, 4H); LCMS Purity: >95%; LCMS (m/z): 401 [M +H]+; HRMS calculated for C20H20N2O5S [M+H]+ 401 .1 166, found 401 .1 182. N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-6,7-dihydro-5H- pyrrolo[1 ,2-a]imidazole-2-sulfonamide
Figure imgf000257_0001
1 ,2,3,5,6,7-Hexahydro-s-indacen-4-amine (70 mg, 0.40 mmol) was dissolved in anhydrous THF (5 mL) and treated with Et3N (49 mg, 0.49 mmol) at RT. The solution was treated with triphosgene (48 mg, 0.161 mmol) and resulting mixture was stirred at 70 °C for 2 h. The reaction mixture was concentrated in vacuo. The residue obtained was stirred with 5% EtOAc-hexanes (20 mL) for 10 min, filtered through a celite pad and concentrated in vacuo to give the corresponding isocyanate as a white solid. In a separate flask, 6,7-dihydro-5 - - pyrrolo[1 ,2-a]imidazole-2-sulfonamide (1 15 mg, 0.61 mmol) was dissolved in anhydrous THF (5 mL) and treated carefully with NaH (25 mg, 0.61 mmol) at 0 °C under nitrogen atmosphere and stirred for 20 minutes. The aforementioned isocyanate in THF was added to reaction mixture under nitrogen atmosphere. The reaction mixture was warmed to RT, stirred for 4 h then concentrated in vacuo. The residue obtained was diluted with 10 mM ammonium bicarbonate in water (20 mL), acetonitrile (20 mL), ethyl acetate (10 mL) and solid formed was removed by filtration and washed with diethyl ether to give N-((1 ,2,3,5,6,7- hexahydro-s-indacen-4-yl)carbamoyl)-6,7-dihydro-5 - -pyrrolo[1 ,2-a]imidazole-2- sulfonamide (50 mg, 32%) as a white solid. 1H NMR (400 MHz, CD3OD): δ = 7.29 (s, 1 H), 6.85 (s, 1 H), 4.23(t, J= 7.2 Hz, 1 H), 2.86-2.79 (m, 6H), 2.72 (t, J = 7.2 Hz, 4H), 2.65-2.60 (m, 2H), 2.02-1 .95 (m, 4H). LCMS (m/z): 387.10 [M+H]+ ; 95.53% (210 nm). HPLC: 94.43% (210nm). HRMS calculated for C19H21 N4O3S1 [M-H]" 385.1340, found 385.1331.
4-Acetyl-/V-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)benzenesulfonamide
Figure imgf000258_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 4-acetylbenzenesulfonamide were used in general method C2 to give the titled compound as a white solid (31 mg, 16%). 1H NMR (600 MHz, DMSO-afe) δ = 1 1 .03 (bs, 1 H) 8.08 (d, J = 8.5 Hz, 2H), 7.99 (d, J = 8.5 Hz, 2H), 7.03 (bs, 1 H), 6.87 (s, 1 H), 2.75 (t, J = 7.4 Hz, 4H), 2.62 (s, 3H), 2.56 (t, J = 7.4 Hz, 4H), 1 .90 (p, J = 7.4 Hz, 4H). HRMS calculated for C21 H21 N2O4S1 [M-H]" 397.1 128, found 397.1225.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4- nitrobenzenesulfonamide
Figure imgf000258_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 4-nitrobenzenesulfonamide were used in general method C2 to give the titled compound as a pale yellow solid (148 mg, 60%). 1H NMR (600 MHz, DMSO-afe) δ = 10.00 (bs, 1 H), 8.21 (d, J = 9.0 Hz, , 2H), 7.97 (d, J = 9.0 Hz, 2H), 7.45 (s, 1 H), 6.75 (s, 1 H), 2.73 (t, J = 7.4 Hz, 4H), 2.61 (t, J = 7.4 Hz, 4H), 1 .87 (p, J = 7.4 Hz, 4H). HRMS calculated for
Ci9H18N305Si [M-H]" 400.0973, found 400.0979. 4-Amino-N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)benzenesulfonamide
Figure imgf000259_0001
Λ/-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4- nitrobenzenesulfonamide dissolved in a solution of ethyl acetate/ DMF (4: 1 , 25 mL/mmol) was stirred at room temperature for 1 h under hydrogen atmosphere with a catalytic amount of Pd/C (0.1 mol%) to afford the titled compound as a white solid (16 mg, 43%). 1 H NMR (600 MHz, DMSO-afe) δ = 7.95 (s, 1 H), 7.54 (d, J = 8.8 Hz, 2H), 6.91 (s, 1 H), 6.59 (d, J = 8.8 Hz, 2H), 6.05 (s, 2H), 2.77 (t, J = 7.4 Hz, 4H), 2.55 (t, J = 7.4 Hz, 4H), 1 .93 (q, J = 7.4 Hz, 4H). HRMS calculated for C19H20N3O3S1 [M-H]" 370.1231 , found
370.1225.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-2,3-dihydro-1 H- indene-5-sulfonamide
Figure imgf000259_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 2,3-dihydro-1 /-/-indene-5-sulfonamide were used in general method C2 to give the titled compound as a white solid (48 mg, 12%). 1 H
NMR (600 MHz, DMSO-afe) δ = 10.68 (bs, 1 H), 8.02 (s, 1 H), 7.75 (d, J = 1 .7 Hz, 1 H), 7.68 (dd, J = 7.9, 1 .7 Hz, 1 H), 7.41 (d, J = 7.9 Hz, 1 H), 6.90 (s, 1 H), 2.91 (t, J = 7.5 Hz, 4H), 2.76 (t, J = 7.4 Hz, 4H), 2.53 (t, J = 7.4 Hz, 4H), 2.05 (p, J = 7.5 Hz, 2H), 1 .91 (p, J = 7.4 Hz, 4H). HRMS calculated for
C22H23N2O3S1 [M-H]" 395.1435, found 395.1430.
A-((4-chlorophenyl)carbamoyl)-2,3-dihydro-1 H-indene-5-sulfonamide
Figure imgf000260_0001
1 -Chloro-4-isocyanatobenzene (prepared using general method B1 ) and 2,3- dihydro-1 /-/-indene-5-sulfonamide were used in general method C2 to give the titled compound as a white solid (60 mg, 32%). 1H NMR (600 MHz, DMSO-afe) δ 10.90 (bs, 1 H), 8.90 (s, 1 H), 7.74 (d, J = 1 .8 Hz, 1 H) 7.68 (dd, J = 7.9, 1.7 Hz, 1 H), 7.41 - 7.35 (m, 3H), 7.26 (dt, 2H), 2.91 (m, 4H), 2.05 (p, J = 7.5 Hz, 2H). HRMS calculated for C16H14CI1 N2O3S1 [M-H]" 349.0419, found HRMS 349.0418.
A-((4-chloro-2,6-diisopropylphenyl)carbamoyl)quinoline-8-sulfonamide
Figure imgf000260_0002
5-Chloro-2-isocyanato-1 ,3-diisopropylbenzene (prepared using general method A2) and quinoline-8-sulfonamide were used in general method C3 to give the titled compound as a white solid (75 mg, 71 %). 1H NMR (600 MHz, CD3OD): δ = 9.13 (dd, J = 4.2, 1 .6 Hz, 1 H), 8.57-8.49 (m, 2H), 8.26 (d, J = 8.2 Hz, 1 H), 7.77-7.67 (m, 2H), 6.99 (s, 2H), 2.65-2.60, (m, 2H), 0.85 (d, 12H); 13C NMR (150 MHz, CD3OD) 5 = 151 .2, 149.0, 143.3, 136.8, 136.7, 133.8, 133.5, 132.3, 129.4, 129.1 , 125.3, 123.0, 122.1 , 109.1 , 28.3, 22.5; LCMS Purity: >95%; LCMS (m/z): 446 [M+H]+; HRMS calculated for C22H25CI1 N3O3S1 [M+H]+ 446.1300, found 446.1314.
A-((4-chloro-2,6-diisopropylphenyl)carbamoyl)isoquinoline-5-sulfonamide
Figure imgf000261_0001
5-Chloro-2-isocyanato-1 ,3-diisopropylbenzene (prepared using general method A2) and isoquinoline-5-sulfonamide (prepared using general method E3) were used in general method C3 to give the titled compound as a white solid (70 mg, 67%). 1 H NMR (600 MHz, CD3OD): δ = 9.41 (s, 1 H), 8.82 (s, 1 H), 8.59 (d, J = 7.3 Hz, 2H), 8.35 (d, J= 8.2 Hz, 1 H), 7.79 (t, J= 7.6 Hz, 1 H), 6.96 (s, 2H), 2.74- 2.70 (m, 2H), 0.96 (s, 6H), 0.85 (d, 12H); 13C NMR (150 MHz, CD3OD) δ = 156.3, 152.5, 149.1 , 143.8, 137.2, 133.9, 133.1 , 1 32.6, 131 .5, 130.4, 126.3, 124.8, 122.8, 122.1 , 28.3, 22.4; LCMS Purity: >95%; LCMS (m/z): 446 [M+H]+; HRMS calculated for C22H25CI1 N3O3S1 [M+H]+ 446.1300, found 446.1319.
A-((4-chloro-2,6-diisopropylphenyl)carbamoyl)quinoline-5-sulfonamide
Figure imgf000261_0002
5-Chloro-2-isocyanato-1 ,3-diisopropylbenzene (prepared using general method A2) and quinoline-5-sulfonamide (prepared using general method E3) were used in general method C3 to give the titled compound as a white solid (31 mg, 60%). 1H NMR (600 MHz, CD3OD): δ = 9.53 (d, J= 8.9 Hz, 1 H), 8.94 (d, J= 3.8 Hz, 1 H), 8.35 (dd, J = 7.3, 1 .2 Hz, 1 H), 8.1 5 (d, J = 8.5 Hz, 1 H), 7.79 (dd, J = 8.5, 7.3 Hz, 1 H), 7.69 (dd, J = 8.7, 4.3 Hz, 1 H), 2.81 -2.76 (m, 2H), 0.85 (d, 12H); 13C NMR (150 MHz, CD3OD) δ = 161 .4, 151 .3,150.7, 149.1 , 142.2, 137.4, 134.0, 133.0, 132.8, 129.9, 129.4, 126.0, 124.1 , 122.9, 29.6, 24.0; LCMS Purity: >95%; LCMS (m/z): 446 [M+H]+; HRMS calculated for C22H25CI1 N3O3S1 [M+H]+ 446.1300, found 446.1317.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)quinoline-8- sulfonamide
Figure imgf000262_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 4H-quinoline-8-sulfonamide were used in general method C3 to give the titled compound as a white solid (60 mg, 51 %); 1 H NMR (600 MHz, DMSO-cfe) 5 = 9.1 1 (d, J = 2.7 Hz, 1 H), 8.56 (d, J= 8.3 Hz, 1 H), 8.40 (d, J = 7.4 Hz, 1 H), 8.32 (d, J= 8.2 Hz, 1 H), 8.17 (s, 1 H), 7.76 (t, J= 7.7 Hz, 1 H), 7.73 (dd, J = 8.4, 4.2 Hz, 1 H), 6.82 (s, 1 H), 2.67 (t, J = 7.4 Hz, 4H), 2.26 (t, J = 7.4 Hz, 4H), 1 .79 (p, J= 7.5 Hz, 4H); 13C NMR (150 MHz, DMSO-tf6): δ = 151.8, 151 .7, 143.3, 143.2, 137.5, 137.1 , 134.5, 133.4, 132.8, 129.9, 126.0, 122.8, 1 18.0, 108.7, 32.7, 30.2, 25.3. LCMS (m/z): 408 [M+H]+. HRMS calculated for C22H22N3O3S1 [M+H]+ 408.1376, found 408.1371 .
A-((4-chloro-2,6-diisopropylphenyl)carbamoyl)quinoline-3-sulfonamide
Figure imgf000262_0002
5-Chloro-2-isocyanato-1 ,3-diisopropylbenzene (prepared using general method A2) and quinoline-3-sulfonamide (prepared using general method E3) were used in general method C3 to give the titled compound as a white solid (30 mg, 57%). 1 H N MR (600 MHz, DMSO-afe): 5 1H NMR (600 MHz, DMSO-afe) δ = 9.24 (s, 1 H), 8.92 (s, 1 H), 8.20 (d, J = 8.3 Hz, 1 H), 8.12 (d, J = 8.5 Hz, 1 H), 7.98 (s, 1 H), 7.93 (t, J= 7.7 Hz, 1 H), 7.74 (t, J= 7.6 Hz, 1 H), 7.02 (s, 2H), 2.81 -2.78 (m, 2H), 0.84 (d, 12H); 13C NMR (150 MHz, DMSO-afe): δ = 153.7, 149.3, 148.7, 147.6, 141 .5, 136.8, 132.6, 132.4, 131 .5, 129.9, 129.2, 128.4, 126.4, 123.3, 28.5, 23.5; LCMS Purity: >95%; LCMS (m/z): 446 [M+H]+; HRMS calculated for C22H25CI1 N3O3S1 [M+H]+ 446.1300, found 446.1315.
A-((4-chloro-2,6-diisopropylphenyl)carbamoyl)quinoxaline-5-sulfonamide
Figure imgf000263_0001
5-Chloro-2-isocyanato-1 ,3-diisopropylbenzene (prepared using general method A2) and quinoxaline-5-sulfonamide (prepared using general method E3) were used in general method C3 to give the titled compound as a white solid (39 mg, 75%); 1 H NMR (600 MHz, DMSO-afe) δ = 9.18 (d, J = 3.7 Hz, 2H), 8.46 (d, J = 7.3 Hz, 1 H), 8.38 (dd, J= 8.1 , 2.7 Hz, 1 H), 8.04-7.95 (m, 1 H), 7.83 (s, 1 H), 6.99 (s, 2H), 2.55-2.49 (m, 2H), 0.74 (d, 12H); 13C NMR (150 MHz, DMSO-afe): 149.2, 147.1 , 146.8, 146.2, 142.7, 140.5, 138.5, 138.2, 1 34.2, 133.4, 132.6, 129.6, 123.4, 28.4, 22.7; LCMS Purity: >95%; LCMS (m/z): 447 [M+H]+; HRMS calculated for C21 H24CI1 N4O3S1 [M+H]+ 447.1252, found 447.1266.
A-((4-chloro-2, 6-diisopropylphenyl)carbamoyl)naphthalene-2-sulfonamide
Figure imgf000263_0002
5-chloro-2-isocyanato-1 , 3-diisopropylbenzene (prepared using general method A2) and naphthalene-2 -sulfonamide (prepared using general method E3) were used in general method C3 to give the titled compound as a white solid (35 mg, 67%). 1H NMR (600 MHz, CD3OD): δ = 8.55 (s, 1 H), 8.05-7.92 (m, 4H), 7.64- 7.58 (m 2H), 6.99 (s, 2H), 2.94-2.89 (m, 2H), 0.94 (bs, 12H); 13C NMR (150 MHz, CD3OD) 5 159.9, 150.6, 142.2, 135.9, 134.0, 133.7, 132.6, 130.2, 129.5, 129.1 , 128.8, 128.6, 128.1 , 124.3, 124.2, 29.7, 24.0; LCMS Purity: >95%; LCMS (m/z): 445 [M+H]+; HRMS calculated for C23H26CI1 N2O3S1 [M+H]+ 445.1347, found 445.1349.
A-((4-chloro-2,6-diisopropylphenyl)carbamoyl)-6-methoxynaphthalene-2- sulfonamide
Figure imgf000264_0001
5- chloro-2-isocyanato-1 , 3-diisopropylbenzene (prepared using general method A2) and 6-methoxynaphthalene-2-sulfonamide (prepared using general method E3) were used in general method C3 to give the titled compound as a white solid (39 mg, 70%); 1 H NMR (600 MHz, DMSO-afe) δ = 8.23 (s, 1 H), 7.86 (d, J = 9.0 Hz, 1 H), 7.78 (dd, J = 9.1 , 6.4 Hz, 2H), 7.49 (s, 1 H), 7.35 (d, J = 2.6 Hz, 1 H), 7.20 (dd, J = 8.9, 2.6 Hz, 1 H), 6.96 (s, 2H), 3.08-2.98 (m, 2H), 0.93 (bs, 12H); 13C NMR (150 MHz, DMSO-afe): δ = 158.0, 149.6, 135.2, 134.4, 131 .0, 130.5, 128.1 , 127.5, 127.3, 126.5, 124.7, 124.6, 122.7, 1 19.4, 106.2, 55.7, 28.3, 23.4; LCMS Purity: >95%; LCMS (m/z): 475 [M+H]+; HRMS calculated for C24H28CI1 N2O4S1 [M+H]+ 475.1453, found 475.1474.
6- chloro-yV-((4-chloro-2,6-diisopropylphenyl)carbamoyl)naphthalene-2- sulfonamide
Figure imgf000264_0002
5-Chloro-2-isocyanato-1 , 3-diisopropylbenzene (prepared using general method A2) and 6-chloronaphthalene-2-sulfonamide (prepared using general method E3) were used in general method C3 to give the titled compound as a white solid (34 mg, 61 %); 1 H NMR (600 MHz, DMSO-afe) δ = 8.30 (s, 1 H), 8.06 (d, J = 2.2 Hz, 1 H), 8.01 (d, J = 8.8 Hz, 1 H), 7.93-7.85 (m, 2H), 7.55 (dd, J = 8.7, 2.2 Hz, 1 H), 7.40 (s, 1 H), 6.95 (s, 2H), 3.09-2.97 (m, 2H), 0.92 (bs, 12H); 13C NMR (150 MHz, DMSO-afe): δ = 160.2, 149.6, 145.8, 134.8, 134.2, 131 .7, 131 .1 , 130.8, 130.7, 127.2, 126.8, 126.6, 125.8, 125.7, 122.7, 28.3, 23.6; LCMS Purity: >95%; LCMS (m/z): 479 [M+H]+; HRMS calculated for C23H25CI2N2O3S1 [M+H]+ 479.0957, found 479.0937. yV-((4-chloro-2,6-diisopropylphenyl)carbamoyl)-5,6,7,8- tetrahydronaphthalene-2-sulfonamide
Figure imgf000265_0001
5-chloro-2-isocyanato-1 , 3-diisopropylbenzene (prepared using general method A2) and 5,6,7,8-tetrahydronaphthalene-2-sulfonamide (prepared using general method E3) were used in general method C1 to give the titled compound as a white solid (8 mg, 38%); 1H NMR (600 MHz, DMSO-afe) δ = 7.85 (s, 1 H), 7.58 (s, 1 H), 7.57 (d, J = 8.0 Hz, 1 H), 7.22 (d, J = 7.2 Hz, 1 H), 7.08 (s, 2H), 2.85- 2.81 (m, 2H), 2.78-2.74 (m, 4H), 1.74 (t, J = 3.3 Hz, 4H), 0.98 (bs, 12H); 13C NMR (150 MHz, DMSO-afe): δ = 149.3, 137.7, 137.5, 132.4, 129.8, 129.5, 129.7, 126.3, 124.3, 123.4, 122.9, 29.3, 29.2, 28.5, 23.4, 22.8, 22.7; LCMS Purity: >95%; LCMS (m/z): 449 [M+H]+; HRMS calculated for C23H30CI1 N2O3S1 [M+H]+ 449.1660, found 449.1664. yV-((4-chloro-2,6-diisopropylphenyl)carbamoyl)thieno[3,2-b]pyridine-6- sulfonamide
Figure imgf000266_0001
5-chloro-2-isocyanato-1 , 3-diisopropylbenzene (prepared using general method A2) and thieno[3,2-b]pyridine-6-sulfonamide were used in general method C3 to give the titled compound as a white solid (35 mg, 66%); 1 H NMR (600 MHz, DMSO-afe) δ = 9.07 (d, J = 2.0 Hz, 1 H), 9.05 (s, 1 H), 8.45 (d, J = 5.5 Hz, 1 H), 8.01 (s, 1 H), 7.70 (d, J = 5.5 Hz, 1 H), 7.05 (s, 2H), 2.79-2.75 (m, 2H), 0.87 (d, 12H); 13C NMR (150 MHz, DMSO-afe): δ = 158.2, 152.7, 149.3, 145.8, 138.2, 132.7, 132.3, 132.2, 131.6, 131 .1 , 124.7, 123.4, 28.5, 22.9; LCMS Purity: >95%; LCMS (m/z): 452 [M+H]+; HRMS calculated for C20H23CI1 N3O3S2 [M+H]+ 452.0864, found 452.0884. yV-((4-chloro-2,6-diisopropylphenyl)carbamoyl)-3-ethylisoxazolo[5,4- b]pyridine-5-sulfonamide
Figure imgf000266_0002
5-Chloro-2-isocyanato-1 , 3-diisopropylbenzene (prepared using general method A2) and thieno[3,2-b]pyridine-6-sulfonamide were used in general method C3 to give the titled compound as a white solid (38 mg, 64%); 1 H NMR (600 MHz, DMSO-afe) 5 = 9.1 1 (s, 1 H), 8.98 (s, 1 H), 8.14 (s, 1 H), 7.08 (s, 2H), 3.09 (q, J = 7.5 Hz, 2H), 2.82-2.77 (m, 2H), 1 .34 (t, J= 7.5 Hz, 3H), 1.02-0.90 (d, 12H). 13C NMR (150 MHz, DMSO-afe): δ = 170.2, 161 .9, 150.5, 149.3, 134.3, 134.2, 132.8, 131 .0, 123.5, 1 12.7, 109.9, 28.5, 23.0, 19.2, 1 1 .9; LCMS Purity: >95%; LCMS (m/z): 465 [M+H]+; HRMS calculated for C21 H26CI1 N4O4S1 [M+H]+ 465.1358, found 465.1354.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)benzofuran-2- sulfonamide
Figure imgf000267_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and benzofuran-2-sulfonamide were used in general method C3 to give the titled compound as a white solid (60 mg, 52%); 1 H NMR (400 MHz, DMSO-de): δ = 8.00 (bs, 1 H), 7.77 (d, J= 8 Hz, 1 H), 7.69 (d, J= 8 Hz, 1 H), 7.51 (s, 1 H), 7.49 (d, J = 8 Hz, 1 H), 7.36 (t, J = 8 Hz, 1 H), 7.08 (bs, 1 H), 6.87 (s, 1 H), 2.75 (t, J = 8 Hz, 4H), 2.59 (t, J = 8 Hz, 4H), 1 .92-1 .85 (m, 4H). 13C NMR (150 MHz, DMSO-de): δ = 154.9, 143.2, 137.6, 130.3, 127.5, 126.7, 124.4, 123.3, 1 17.7, 1 12.3, 1 10.0, 109.4, 107.4, 32.9, 30.6, 25.5. LCMS (m/z): 397 [M+H] +. HRMS calculated for C2i H2i N204Si [M+H]+ 397.1217, found 397.1215.
A-((4-chloro-2,6-diisopropylphenyl)carbamoyl)benzofuran-2-sulfonamide
Figure imgf000267_0002
5-chloro-2-isocyanato-1 , 3-diisopropylbenzene (prepared using general method A2) and benzofuran-2 -sulfonamide (prepared using general method E3) were used in general method C3 to give the titled compound as a white solid (25 mg, 49%). 1H NMR (600 MHz, DMSO-afe) δ = 7.85 (s, 1 H), 7.73 (d, J= 7.8 Hz, 1 H), 7.66 (d, J= 8.4 Hz, 1 H), 7.47 (s, 1 H), 7.34 (t, J= 7.5 Hz, 1 H), 7.17 (s, 1 H), 7.04 (s, 2H), 2.99-2.95 (m, 2H), 0.94 (bs, 12H); 13C NMR (150 MHz, DMSO-afe): δ = 154.9, 149.5, 132.6, 132.3, 132.0, 127.2, 126.9, 126.8, 123.3, 123.2, 1 12.1 , 1 12.0, 109.8, 28.5, 23.3; LCMS Purity: >95%; LCMS (m/z): 435 [M+H]+; HRMS calculated for C2i H24C N20 Si [M+H]+ 435.1 140, found 435.1 140. yV-((4-chloro-2,6-diisopropylphenyl)carbamoyl)benzo[b]thiophene-2- sulfonamide
Figure imgf000268_0001
5-chloro-2-isocyanato-1 , 3-diisopropylbenzene (prepared using general method A2) and benzo[b]thiophene-2-sulfonamide (prepared using general method E3) were used in general method C3 to give the titled compound as a white solid (38 mg, 72%); 1 H NMR (600 MHz, DMSO-afe) δ 8.03 (d, J = 7.9 Hz, 1 H), 7.93 (d, J = 8.0 Hz, 2H), 7.80 (s, 1 H), 7.46 (dt, J = 15.4, 7.0 Hz, 2H), 7.04 (s, 2H), 3.05- 2.83 (m, 2H), 0.94 (bs, 12H); 13C NMR (150 MHz, DMSO-afe): δ 155.3, 149.5, 141 .1 , 138.0, 132.4, 132.0, 126.8, 125.7, 125.4, 123.1 , 123.0, 122.9, 109.7, 28.5, 23.3; LCMS Purity: >95%; LCMS (m/z): 451 [M+H]+; HRMS calculated for C21 H24CI1 N2O3S2 [M+H]+ 451 .091 1 , found. 451 .0900.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4-(2-(7-methoxy-4,4- dimethyl-1 ,3-dioxo-3,4-dihydroisoquinolin-2(1 H)- yl)ethyl)benzenesulfonamide
Figure imgf000268_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 4-(2-(7-methoxy-4,4-dimethyl-1 ,3-dioxo-3,4- dihydroisoquinolin-2(1 -/)-yl)ethyl)benzenesulfonamide were used in general method C2 to give the titled compound as a white solid (85 mg, 52%). 1 H NMR (600 MHz, DMSO-afe) δ = 10.72 (bs, 1 H), 7.91 (s, 1 H), 7.80 (d, J = 8.1 Hz, 2H), 7.58 (d, J = 8.7 Hz, 1 H), 7.53 (d, J = 2.9 Hz, 1 H), 7.39 (d, J = 7.9 Hz, 2H), 7.29 (dd, J = 8.7, 2.9 Hz, 1 H), 6.88 (s, 1 H), 4.13 (t, J = 7.5 Hz, 2H), 3.83 (s, 3H), 2.93 (t, J = 7.5 Hz, 2H), 2.76 (t, J = 7.4 Hz, 4H), 2.55 (t, J = 7.4 Hz, 4H), 1.90 (p, J = 7.4 Hz, 4H), 1 .42 (s, 6H). HRMS calculated for
C33H34N3O6S1 [M-H]" 600.2174, found 600.2183.
N-(4-(N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)phenethyl)-5-methylisoxazole-3-carboxamide
Figure imgf000269_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 5-methyl-A/-(4-sulfamoylphenethyl)isoxazole-3-carboxamide were used in general method C2 to give the titled compound as a white solid (14 mg, 62%). 1 H N MR (600 MHz, DMSO-afe) δ 8.78 (t, J = 5.8 Hz, 1 H), 7.80 (s, 1 H), 7.74 (d, J = 8.3 Hz, 2H), 7.41 (d, J = 8.3 Hz, 2H), 6.85 (s, 1 H), 6.50 (q, J = 1.0 Hz, 1 H), 3.49 (m, 2H), 2.91 (t, J = 7.0 Hz, 2H), 2.75 (t, J = 7.4 Hz, 4H), 2.56 (t, J = 7.4 Hz, 4H), 2.45 (d, J = 0.9 Hz, 3H), 1 .89 (p, J = 7.4 Hz, 4H). HRMS calculated for C26H27N4O5S1 [M-H]" 507.1708, found 507.1709. 3-Ethyl-yV-(4-(yV-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)phenethyl)-4-methyl-2-oxo-2,5-dihydro-1 H-pyrrole- 1-carboxamide
Figure imgf000269_0002
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 3-ethyl-4-methyl-2-oxo-A/-(4-sulfamoylphenethyl)-2,5- dihydro-1 H-pyrrole-1-carboxamide were used in general method C2 to give the titled compound as a white solid (78 mg, 50%).1H NMR (600 MHz, DMSO-afe) δ = 10.78 (bs, 1H), 8.38 (t, J= 7.6 Hz, 1H), 7.97 (s, 1H), 7.82 (d, J = 8.2 Hz, 2H), 7.42 (d, J= 8.2 Hz, 2H), 6.88 (s, 1H), 4.16 (s, 2H), 3.48 (q, J = 6.7 Hz, 2H), 2.88 (t, J= 7.2 Hz, 2H), 2.75 (t, J= 7.4 Hz, 4H), 2.53 (t, J= 7.4 Hz, 4H), 2.18 (q, J= 7.5 Hz, 2H), 2.01 (s, 3H), 1.90 (p, J= 7.4 Hz, 4H), 0.97 (t, J= 7.5 Hz, 3H). HRMS calculated for C29H33N4O5S1 [M-H]" 549.2177, found 549.2169.
5-Chloro-/V-(4-(N-((1,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)sulfamoyl)phenethyl)-2-methoxybenzamide
Figure imgf000270_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 5-chloro-2-methoxy-A/-(4-sulfamoylphenethyl)benzamide were used in general method C2 to give the titled compound as a white solid (325 mg, 70%).1H NMR (600 MHz, DMSO-afe) δ = 10.83 (bs, 1H), 8.27 (t, J = 6.0 Hz, 1 H), 7.96 (s, 1 H), 7.84 (d, J = 8.0 Hz, 2H), 7.66 (d, J = 2.8 Hz, 1 H), 7.50 (dd, J= 8.9 Hz, 2.8 Hz, 1H, 7.44 (t, J= 8.0 Hz, 2H), 7.13 (d, J= 8.9 Hz, 1H), 6.87 (s, 1H), 3.78 (s, 3H), 3.53 (q, J= 6.6 Hz, 2H), 2.91 (t, J= 7.2 Hz, 2H), 2.74 (t, J= 7.4 Hz, 4H), 2.53 (t, J= 7.4 Hz, 4H), 1.88 (p, J= 7.3 Hz, 4H). HRMS calculated for C29H29CI1N3O5S1 [M-H]" 566.1522, found
566.1543.
13C NMR (100 MHz, DMSO-d6): δ = 163.6, 155.7, 145.3, 143.6, 143.0, 142.4, 142.1, 139.6, 137.1, 131.5, 129.5, 129.2, 127.4, 125.7, 124.8, 124.3, 117.9, 114.1, 108.3, 56.2, 34.7, 32.6, 32.4, 30.0, 28.9, 24.9. LCMS, Purity: 90.06%, tr = 3.38 min, m/z 566.37 (M-H+). HRMS (FAB+) calcd for
C29H30CIN3O5S [M+H]+ : 568.1595, found: 568.1589.
PYRIDINES
2, 3, 5, 6, 7-hexahydro-s-indacen-4-ylcarbamoyl)-4-(2-hydroxypropan- 2-yl)pyridine-2-sulfonamide
Figure imgf000271_0001
To a solution of 1 ,2,3,5,6,7-hexahydros-indacen-4-amine (0.20 g, 1 .15 mmol) in anhydrous THF (5 ml_), triethylamine (0.35 g, 3.47 mmol, 3.0 eq) was added followed by triphosgene (0.265 g, 0.86 mmol, 0.5 eq) at 0 °C and the mixture was stirred at ambient temperature for 3 h. The mixture was cooled to 0 °C, methyl 2-sulfamoylisonicotinate (0.27 g, 1 .27 mmol, 1 .1 eq) added and stirring continued at ambient temperature overnight. Upon completion the reaction mixture was poured into brine and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. The crude product was purified by column chromatography on silica gel using 20-50% gradient of EtOAc-hexanes eluent to give methyl 2-(Λ/-(1 , 2, 3, 5, 6, 7-hexahydro-s-indacen-4-ylcarbamoyl)sulfamoyl)isonicotinate as a light brown solid (0.31 g, 65%).
Methyl 2-(A/-(1 , 2, 3, 5, 6, 7-hexahydro-s-indacen-4- ylcarbamoyl)sulfamoyl)isonicotinate (0.30 g, 0.72 mmol) was dissolved in anhydrous THF (8 ml_) and the solution cooled to 0 °C. Methyl magnesium bromide (3 M solution in diethyl ether, 0.96 ml_, 2.88 mmol, 4.0 eq) was added at 0 °C under nitrogen atmosphere and stirring continued at ambient temperature for 3 h. Upon completion the reaction mixture was poured into saturated aqueous ammonium chloride and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried (Na2S04) and concentrated in vacuo. The crude residue was purified by reverse phase prep HPLC to afford the titled compound as a white solid (0.016 g, 5%). 1 H NMR (400 MHz, CD3OD): δ = 8.45 (br.s, 1 H), 8.16 (s, 1 H), 7.55 (br.s, 1 H), 6.87 (s, 1 H), 2.80 (t, J = 7,2 Hz, 4H), 2.66 (t, J = 7.2 Hz, 4H), 1 .96 (quin, J = 7.6 Hz, 4H), 1 .53 (s, 6H). LCMS, Purity: 98%, m/z416.09 (M+H+). HRMS (FAB+) calcd for C21 H25N3O4S [M+H]+: 416.1566, found: 416.1556.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-6,7-dihydro-5H- cyclopenta[ ?]pyridine-3-sulfonamide
Figure imgf000272_0001
4-lsocyanato-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A2) and 6,7-dihydro-5H-cyclopenta[b]pyridine-3-sulfonamide were used in general method C2 to give the titled compound as a white solid (12 mg, 27%). 1H NMR (600 MHz, DMSO-d6): δ = 8.71 (s, 1 H), 8.01 (s, 1 H), 7.96 (bs, 1 H), 6.87 (s, 1 H), 2.97-2.93 (m, 4H), 2.75 (t, J = 6 Hz, 4H), 2.55 (t, J = 6 Hz, 4H), 2.1 1 -2.07 (m, 2H), 1.93-1 .88 (m, 4H). 13C NMR (150 MHz, DMSO-d6): δ 169.6, 146.4, 144.9, 143.2, 137.4, 137.2, 131.0, 129.6, 1 17.7, 108.7, 34.0, 32.9, 30.6, 30.3, 25.4, 23.2. LCMS (m/z): 398 [M +H] +.
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)pyridine-2- sulfonamide
Figure imgf000272_0002
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and pyridine-2-sulfonamide were used in general method C1 to give the titled compound as a white solid (40 mg, 10%). 1H NMR (400 MHz, DMSO-d6): δ = 8.5 (d, J = 4.0 Hz, 1 H), 7.88 (t, J = 7.6 Hz, 1 H), 7.81 (t, J = 7.6 Hz, 1 H), 7.59 (s, 1 H), 7.4 (t, J = 5.8 Hz, 1 H), 6.76 (s, 1 H), 2.73 (t, J = 7.2 Hz, 4H), 2.61 (t, J = 7.2 Hz, 4H), 1 .88 (quin, J = 7.2 Hz, 4H).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)pyridine-3- sulfonamide
Figure imgf000273_0001
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and pyridine-3-sulfonamide were used in general method C1 to give the titled compound as a white solid (12 mg, 3%). 1H NMR (400 MHz, CD3OD): δ = 9.08 (s, 1 H), 8.65 (d, J = 4.4 Hz, 1 H), 8.36 (d, J = 8.0 Hz, 1 H), 7.56 (dd, J= 8.0, 4.8 Hz, 1 H), 6.88 (s, 1 H), 2.82 (t, J= 7.2 Hz, 4H), 2.69 (t, J = 7.2 Hz, 4H), 2.0 (quin, J = 7.2 Hz, 4H).
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4- (trifluoromethyl)pyridine-2-sulfonamide
Figure imgf000273_0002
4-lsocyanato-8-methyl-1 ,2,3,5,6,7-hexahydro-s-indacene (prepared using general method A1 ) and 4-(trifluoromethyl)pyridine-2-sulfonamide were used in general method C1 to give the titled compound as a white solid (16 mg, 3%). 1H NMR (400 MHz, CD3OD): δ = 8.47 (s, 1 H), 8.22 (s, 1 H), 7.42 (s, 1 H), 6.98 (s, 1 H), 2.82 (t, J = 7.2 Hz, 4H), 2.66 (t, J = 7.2 Hz, 4H), 1 .95 (quin, J = 7.2 Hz, 4H); 19F NMR (233.33 MHz, DMSO-d6): -63.48 (s, 3F). LINKER
N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamothioyl)-4-(2- hydroxypropan-2-yl)furan-2-sulfonamide
Figure imgf000274_0001
To s solution of 1 ,2, 3,5,6, 7-hexahydro-s-indacen-4-amine (0.10 g, 0.58 mmol) in anhydrous DCM (2.0 ml_) was added 1 , 1 '-thiocarbonyldiimidazole (1.1 eq) and the reaction stirred for 4 h at ambient temperature. The solvent was removed in vacuo then the residue taken up in acetone (2.0 ml_) and potassium carbonate (2.5 eq) was added followed by 4-(2-hydroxypropan-2-yl)furan-2- sulfonamide (1.2 eq). The reaction mixture was heated at reflux overnight, concentrated in vacuo then neutralized using 10 % citric acid (10 ml_) and immediately extracted using ethyl acetate (2 x 10 ml_), dried (MgS04) and concentrated in vacuo. The crude product was purified using column chromatography on silica with MeOH/DCM eluent followed by HPLC to give the titled compound as an off white solid (13 mg, 4%). 1 H NMR (400 MHz, DMSO- d6): δ = 12.0 (bs, 1 H), 9.72(s, 1 H), 7.88 (s, 1 H), 7.43 (s, 1 H), 7.01 (s, 1 H), 5.15 (br.s., 1 H), 2.81 (t, J = 6.8 Hz, 4H), 2.59 (t, J = 6.8 Hz, 4H), 1 .95 (quin, J = 7.6 Hz, 4H), 1 .39 (s, 6H). 13C NMR (100 MHz, DMSO-d6): δ = 176.9, 143.1 , 142.7, 138.7, 137.1 , 130.4, 1 19.3, 1 17.7, 66.6, 32.4, 30.9, 29.9, 24.9. LCMS: Purity = 95.08%, tr = 3.45 min, m/z 421 .30 (M+H+).
Biological Testing Methodology NLRP3 inhibition assays
The following assays can be used to determine inhibitory activity of test compounds on the NLRP3 inflammasome using common stimuli such as adenosine triphosphate, nigericin, LeuLeu-OMe or monosodium urate crystals (MSU).
Cell culture
To generate HMDM, human monocytes are isolated from buffy coat blood using Ficoll-Plaque Plus (GE Healthcare) and density centrifugation. CD14÷ cell selection is performed using MACS magnetic beads (Miltenyi Biotec). Isolated CD14+ monocytes are differentiated in culture for 7 days with 10 ng/ml human CSF-1 (Miltenyi Biotec) in Iscove's modified Dulbecco's medium (IMDM) containing L-glutamine supplemented with 10% FBS and 1 % penicillin/streptomycin (Life Technologies) as described by Croker et al 2013 Immunol Cell Biol 91:625.
Mouse bone marrow-derived macrophages (BMDM) were derived from bone marrow progenitors isolated from the femurs and tibias of C57BL/6 mice. Bones were flushed with medium, and bone marrow cells were cultured for 7 days in RPMI 1640 medium supplemented with 10% heat inactivated FCS, 2 mM GlutaMAX (Life Technologies), 50 U/ml penicillin-streptomycin (Life Technologies) and 150 ng/ml recombinant human M-CSF (endotoxin-free, expressed and purified by The University of Queensland Protein Expression Facility).
NLRP3 inflammasome activation assays
HMDM are seeded at 1 x 105/ml. The following day the overnight medium is replaced and cells are stimulated with Escherichia coli serotype 01 1 1 :B4 (Sigma Aldrich) for 3 h. Medium is removed and replaced with serum free medium (SFM) containing test compound 30 min prior to NLRP3 stimulation. Cells are then stimulated with: adenosine 5' -triphosphate disodium salt hydrate (5 mM 1 h), nigericin (10 μΜ 1 h), LeuLeu-OMe (1 mM 2 h) or MSU (200 μg/ml 15 h). ATP can be sourced from Sigma Aldrich, nigericin and MSU from Invivogen and LeuLeu-Ome from Chem-lmpex International.
BMDM are seeded at 1 x 105/ml. The following day the overnight medium is replaced and cells are stimulated with Ultrapure lipopolysaccharide from Escherichia coli K12 strain (InvivoGen) for 3 h. Medium is removed and replaced with serum free medium (SFM) containing test compound 30 min prior to NLRP3 stimulation. Cells are then stimulated with: adenosine 5'-triphosphate disodium salt hydrate (1 .25-5 mM 1 h), nigericin (5 μΜ 1 h), LeuLeu-OMe (1 mM 2 h) or MSU (200 pg/ml 15 h). ATP can be sourced from Sigma Aldrich, nigericin and MSU from Invivogen and LeuLeu-Ome from Chem-lmpex International.
Measurement of IL-1 B. IL-18. TNFa and cell death
For ELISA and cell death assays cells are seeded in 96 well plates. Supernatants are removed and analysed using ELISA kits according to the manufacturer's instructions (DuoSet® R&D Systems, ReadySetGo!® eBioscience, BD OptEIA™, or Perkin Elmer AlphaLISA®). Cell death is assessed by measurement of LDH release relative to a 100% cell lysis control using the CytoTox96® non-radioactive cytotoxicity assay (Promega).
Murine studies on compound levels in blood plasma and brain
General experimental: Carbutamide was purchased from Sigma Aldrich (Catalogue No. 381578). Acetonitrile was Chromasolv® HPLC grade (Sigma Aldrich, Sydney, Australia), the formic acid was AR grade 99%-100% Normapur (VWR International Pty Ltd, Brisbane, Australia), DMSO was ReagentPlus® grade (D5879, Sigma Aldrich, Sydney, Australia) and the H20 Milli-Q was filtered. The HPLC vial and polypropylene inserts from Agilent Technologies (Melbourne, Australia), while the 1.5 mL Eppendorf tubes Protein LoBind Tubes were from VWR International Pty Ltd (Brisbane, Australia).
Preparation of precipitation solution: 100 mL ACN and 5 μί of 10 mM carbutamide in DMSO (ACN with 1 35 ng/m L carbutamide MS internal standard).
Preparation of standard curve in plasma: A 1 mg/mL of test compound in 10 mM NH4HC03 was prepared and diluted 1 0-fold to give a 1 00,000 ng/mL stock solution. A series of 1 0-fold dilutions of the 1 00,000 ng/mL stock solution with 1 0 mM NH4HC03 gave concentrations of 10,000, 1 ,000, 1 00 and 1 0 ng/mL. The 1 00,000 ng/mL stock solution was diluted to 3: 7 with 1 0 mM NH HC03 to give a concentration of 30,000 ng/mL and a series of 1 0-fold dilutions gave concentrations of 3,000, 300, 30 and 3 ng/mL.
20 μί of test compound-containing solution and 160 μί precipitation solution were added to 20 μί of mouse plasma in a low binding Eppendorf tube. The samples were vortexed, allowed to stand at 4 °C for 1 0 mins and centrifuged at 14, 000 x gfor 8 min. 1 50 μί of the supernatant was transferred to an HPLC vial insert. The samples were stored at 4 °C until analysis.
Preparation of standard curve in brain homogenate: The sample solutions prepared for the plasma standard curve were used for the brain homogenate standard curve.
The mouse brain homogenate from the saline control was thawed and vortexed for 3 min or until homogenous, sonicated for 1 min. When the foam settled, 50 μί of mouse brain homogenate was transferred into an Eppendorf tube, followed by 50 μί of test compound in 1 0 mM NH HC03, 1 50 μί of H20 and 500 μί of ice cold precipitation solution with vortexing after every addition. The standards were allowed to stand at 4 °C for 1 0 mins and then centrifuged at 14, 000 x g for 8 min. 200 μί of the supernatant was transferred to HPLC vial insert ensuring that no air bubbles were present and the samples stored at 4 °C until analysis.
Dosing of mice and transcardial perfusion Dosing: Oral gavage at 20 mg/kg Time point: 2 hour
Prepare stock compounds for dosing at 4 mg/ml in sterile PBS. Mice were weighed and dosed by oral gavage at 20 mg/kg for each compound. After 2 hours mice were anesthetized using a combination of Zoletil (50 mg/kg) and Xylazine (10 mg/kg) and blood was collected by cardiac puncture into tubes containing 20 μΙ_ of 100 mM EDTA. The blood was centrifuged at 2000 x g for 15 minutes at 4 °C to collect plasma.
Preparation of plasma samples for analysis: 20 μΙ_ of NH4HCO3 and 160 μΙ_ precipitation solution were added to 20 μΙ_ of mouse plasma in a low binding Eppendorf tube. The samples were vortexed, allowed to stand at 4 °C for 10 mins and centrifuged at 14,000 χ g for 8 min. 150 μΙ_ of the supernatant was transferred to an HPLC vial insert ensuring that no air bubbles were present. The samples were stored at 4 °C until analysis.
Brain homogenate preparation: The brains of mice were perfused with PBS for 5 minutes then dissected and weighed. Brain homogenate was prepared by homogenizing total brain (0.5 g) with 4 volumes (2 ml) of deionized water and stored at -20 °C before analysis. The homogenate was thawed, vortexed for 3 min or until homogenous, and sonicated for 1 min. When the foam settled, 50 μΙ_ of mouse brain homogenate was transferred into an Eppendorf tube, followed by 50 μΙ_ of 10 mM NH HC03, 150 μΙ_ of H20 and 500 μΙ_ of ice cold precipitation solution with vortexing after every addition. 200 μΙ_ of the supernatant was transferred to HPLC vial insert ensuring that no air bubbles were present and the samples stored at 4 °C until analysis.
Preparation of brain samples for analysis: 50 μΙ_ of mouse brain was transferred into an Eppendorf tube, followed by 50 μί of 10 mM NH4HC03, 150 μΙ_ of H20 and 500 μΙ_ of ice cold precipitation solution with vortexing after every addition. The solutions were allowed to stand at 4 °C for 10 mins and then centrifuged at 14,000 χ g for 8 min. 200 μΙ_ of the supernatant was transferred to HPLC vial insert ensuring that no air bubbles were present and the samples stored at 4 °C until analysis.
LC-MS/MS: The samples were analysed on an AB Sciex 4000QTrap MS with 2 Shimadzu Nexera LC-30AD Solvent Delivery Units, Shimadzu Nexera SIL- 30AC Auto-Sampler, Shimadzu Prominence DGU-20A5 Degasser, Shimadzu Prominence CBM-20A System Controller and Shimadzu Prominence CTO-20A Column Oven. The column oven was set to 40 °C, while the Autosampler was set to 15 °C. 2 μΙ_ injections were made and MS analyses were undertaken in Selected Reaction Monitoring (SRM) mode using Turbo Spray (-)-ESI with Low Resolution Q1 and Low Resolution Q3. MS parameters: CUR: 30.00, IS: - 4300.00, TEM: 500.00, GS1 : 50.00, GS2: 50.00, ihe: ON, CAD: High, DP - 60.00, EP -10.00, CXP -15.00. MCC950 SRM: Q1 403.2 to Q3 204.3 Da, dwell 150 msec, CE -27 and carbutamide (IS) SRM: Q1 270.0 to Q3 171 .0 Da, dwell 100 msec, CE -25. HPLC Column: Waters Atlantis® T3 5 pm 2.1 χ 50mm with Atlantis® T3 5 pm 2.1 χ 10 mm guard column. Flow rates and solvent: 0.35 ml/min, solvent A: 0.1 % formic acid in H2O, solvent B: 0.1 % formic acid in ACN; isocratic2% B from 0^2 mins, gradient 2% 100% B from 2^5 mins, isocratic 100% from 5- 9 mins, gradient 100%- 2% B from 9- 9.1 mins and isocratic 2% B from 9.1 ^13 mins. The peak areas from the SRM data for carbutamide and test compound were analysed using the AB Sciex's Analyst software using the Quantitation Wizard. The peak area was plotted against the ng/mL concentration in 20 μί 3 to 30,000 ng/mL test compound solutions and the lower and upper range of linear response was determined. These data were then plotted in Microsoft Excel and the linear response equation used to determine the test compound concentration in the 20 μί plasma solutions. Similarly, for the brain homogenate samples, the peak areas of the 50 μί 3 to 3,000 ng/mL test compound solutions were used to determine the test compound concentration in the 50 μΙ_ brain homogenate solutions.
RESULTS
The full series of tPSA and biological results are provided in the tables below, however select data is presented below for certain compounds of the invention.
Figure imgf000280_0001
MCC7401 MCC8173 MCC8223 MCC8219 IC50 108 nM ICS0 1 nM IC5024 nM IC50 50 nM PSA 84 PSA 84 PSA 75 PSA 75
Figure imgf000281_0001
Figure imgf000282_0001
Figure imgf000283_0001
Figure imgf000284_0001
Figure imgf000285_0001
Figure imgf000286_0001
Table 1 : Topological Polar Surface Area (tPSA) and molecular weight of select compounds.
Figure imgf000287_0001
Figure imgf000288_0001
Figure imgf000289_0001
Figure imgf000290_0001
Figure imgf000292_0001
Figure imgf000293_0001
Figure imgf000294_0001
Figure imgf000295_0001
Figure imgf000296_0001
Figure imgf000297_0001
Figure imgf000298_0001
Figure imgf000299_0001
Figure imgf000300_0001
Figure imgf000301_0001
Figure imgf000302_0001
Figure imgf000303_0001
Figure imgf000304_0001
Figure imgf000305_0001
Figure imgf000306_0001
Figure imgf000307_0001
Figure imgf000308_0001
Figure imgf000309_0001
Figure imgf000310_0001
Figure imgf000311_0001
Figure imgf000312_0001
Figure imgf000313_0001
Figure imgf000314_0001
Figure imgf000315_0001
Figure imgf000316_0001
Figure imgf000317_0001
Table 2: Compound HRMS characterisation data; Inhibition of IL-1 p release IC50 in nM cell based assay using either HMDM or BMDM (<100 nM = '++++' / <1 μΜ = '+++' / <10 μΜ = '++' / <50 μΜ = '+'); Inhibition of IL-18 release IC50 in nM cell based assay using HMDM (<100 nM = '++++' / <1 μΜ = '+++' / <10 μΜ = '++' / <50 μΜ = '+'). "ND" = not determined.
Figure imgf000317_0002
Table 3: Plasma levels of select test compounds at 2 hour timepoint after oral gavage at 20 mg/Kg
Figure imgf000318_0001
Brain 184 1339
Concentration
(ng/g)*
Plasma 17490 66260
Concentration
(ng/mL)
Brain/Plasma 0.0117 0.0203
Ratio3
tPSA 104.7 84.5
Table 4: Properties of sulfonylureas, including increased BBB penetration, with and without hydroxylalkyl group on furan ring.
Plasma Concentration Post-Dosing
A single dose pharmacokinetic study of N-((1 ,2,3,5,6,7-hexahydro-s-indacen-4- yl)carbamoyl)-1 -isopropyl-1 H-pyrazole-3-sulfonamide (MCC7840 and being a compound of the first aspect) in comparison to N-((1 ,2,3,5,6,7-hexahydro-s- indacen-4-yl)carbamoyl)-4-(2-hydroxypropan-2-yl)furan-2 -sulfonamide (MCC950) using an iv dose of 4 mg/Kg and po dose of 20 mg/Kg clearly indicated an extended half-life, increased maximum concentration (Cmax) and area under the curve (AUC) for the pyrazole derivative in comparison to the furan. This is advantageous leading to comparatively lower doses or less frequent administration.
The procedure followed was: Male C57BL/6 mice were used at 7-9 weeks age with 3 animals per group. Mice were dosed with test compound using single intravenous bolus or oral gavage. Blood samples were taken via submandibular or saphenous vein for analysis of plasma concentrations of compound by LC- MS/MS at the following timepoints: IV (3 mice): 0.083, 0.25, 0.5, 1 , 2, 4, 8 and 24 hours post dosing, PO (3 mice): 0.25, 0.5, 1 , 2, 4, 8 and 24 hours post dosing. LC-MS/MS method for the quantitative determination of test compound in corresponding biological matrix was developed. PK parameters were calculated using Phoenix WinNonlin 6.3. The results are shown graphically in FIGs 1 A to 1 C (MCC950) and FIGs 2A to 2C (MCC7840).
The relevant compound structures are shown below and tables 5-8 contain the relevant data:
Figure imgf000319_0001
Figure imgf000320_0001
Bioavailability of MCC_000950_016 in Mouse (ng/mL)
MCC_000950_016
PO
PO Time (h) M4 M5 M6 Mean PO SD CV (%)
0.250 21900 29000 48900 33267 + 13997 42.1
0.500 20400 34100 35800 30100 + 8443 28.1
1 .00 19300 33700 37000 30000 + 9412 31.4
2.00 18500 22500 26200 22400 + 3851 17.2
4.00 10200 13000 10500 11233 + 1537 13.7
8.00 4330 2360 4670 3787 + 1247 32.9
24.0 60.7 17.4 39.3 39.1 + 21 .7 55.3
PK Parameters M4 M5 M6 Mean PO SD CV (%)
Rsq_adj 0.999 0.996 0.996 -- + -- --
No. points used for T /2 4.00 5.00 6.00 ND + -- --
Cmax (ng/mL) 21900 34100 48900 34967 + 13521 38.7
Tmax (h) 0.250 0.500 0.250 0.333 + 0.144 43.3
T1/2 (h) 2.67 2.1 1 2.37 2.39 + 0.282 11.8
Tiast (h) 24.0 24.0 24.0 24.0 + -- --
AUCo-iast (ng h/mL) 108135 123399 144734 125422 + 18383 14.7
AUCo.inf (ng.h/mL) 108369 123452 144868 125563 + 18341 14.6
MRTo-iast (h) 4.26 3.07 3.47 3.60 + 0.603 16.8
M To-inf (h) 4.31 3.08 3.49 3.63 + 0.624 17.2
AUCExtra (%) 0.216 0.0429 0.0929 0.117 + 0.0892 76.0
AUMCExtra (%) 1 .40 0.376 0.730 0.835 + 0.519 62.1
Bioavailability (%)a 71 .0 + -- --
Tables 5 and 6: PK and bioavailability data for N-((1 ,2,3,5,6,7-hexahydro-s- indacen-4-yl)carbamoyl)-4-(2-hydroxypropan-2-yl)furan-2 -sulfonamide (MCC950) Bioavailability of MCC_007840_002 in Mouse (ng/mL)
MCC 007840 002
IV
IV Time (h) M1 M2 M3 Mean IV SD CV (%)
0.0833 47800 41900 38600 42767 + 4661 10.9
0.250 28100 29300 29300 28900 + 693 2.40
0.500 25200 25200 24500 24967 + 404 1.62
1 .00 19900 18900 17200 18667 + 1365 7.31
2.00 13300 14700 19900 15967 + 3478 21.8
4.00 6520 8550 8590 7887 + 1184 15.0
8.00 3490 3360 4440 3763 + 590 15.7
24.0 149 122 130 134 + 13.9 10.4
PK Parameters M1 M2 M3 Mean IV SD CV (%)
Rsq_adj 0.998 0.999 0.996 -- + -- --
No. points used for T /2 3.00 3.00 3.00 3.00 + -- --
Co (ng/mL) 62333 50100 44301 52245 + 9205 17.6
T1/2 (h) 3.62 3.29 3.26 3.39 + 0.204 6.01
Vdss (L/kg) 0.170 0.158 0.151 0.160 + 0.00989 6.20
CI (mL/m in/kg) 0.659 0.633 0.571 0.621 + 0.0455 7.33
Tiast (h) 24.0 24.0 24.0 24.0 + -- --
AUCo_iast (ng.h/mL) 100364 104705 116222 107097 + 8195 7.65
AUCo-inf (ng.h/mL) 101 143 105283 116833 107753 + 8132 7.55
MRTo-iast (h) 4.11 4.01 4.28 4.13 + 0.133 3.23
MRTo-inf (h) 4.30 4.15 4.40 4.29 + 0.129 3.02
AUCExtra (%) 0.770 0.549 0.523 0.614 + 0.136 22.1
AUMCExtra (%) 5.23 3.81 3.41 4.15 + 0.957 23.1
Bioavailability of MCC_007840_002 in Mouse (ng/mL)
MCC_007840_002
PO
PO Time (h) M4 M5 M6 Mean PO SD CV (%)
0.250 84300 27400 69700 60467 + 29552 48.9
0.500 70300 24000 56600 50300 + 23784 47.3
1 .00 60400 20900 45700 42333 + 19964 47.2
2.00 54900 19100 53800 42600 + 20359 47.8
4.00 32900 14100 32800 26600 + 10825 40.7
8.00 14100 12800 29900 18933 + 9520 50.3
24.0 660 2370 1370 1467 + 859 58.6
PK Parameters M4 M5 M6 Mean PO SD CV (%)
Rsq_adj 0.999 0.984 0.968 -- + -- --
No. points used for T /2 3.00 6.00 4.00 ND + -- --
Cmax (ng/mL) 84300 27400 69700 60467 + 29552 48.9
Tmax (h) 0.250 0.250 0.250 0.250 + 0.000 0.0
T1/2 (h) 3.57 7.31 4.18 5.02 + 2.01 40.1
Tiast (h) 24.0 24.0 24.0 24.0 + -- --
AUCo-iast (ng-h/mL) 364947 226687 457808 349814 + 116301 33.2
AUCo.inf (ng.h/mL) 368345 251697 466063 362035 + 107323 29.6
MRTo-iast ( ) 4.80 8.07 6.41 6.43 + 1.64 25.4
M To-inf (h) 5.03 10.7 6.83 7.52 + 2.90 38.6
AUCExtra (%) 0.922 9.94 1.77 4.21 + 4.98 118
AUMCExtra (%) 5.35 32.1 7.78 15.1 + 14.8 98.1
Bioavailability (%)a 67.2 + -- --
Tables 7 and 8: PK and bioavailability data for N-((1 ,2,3,5,6,7-hexahydro-s- indacen-4-yl)carbamoyl)-1 -isopropyl-1 H-pyrazole-3-sulfonamide (MCC7840)
Figure imgf000324_0001
Figure imgf000325_0001
Table 9: Comparative IC50 data for commercial compounds.
Figure imgf000326_0001
325
Figure imgf000327_0001

Claims

A compound of formula (I), or a pharmaceutically acceptable salt, solvate prodrug thereof:
Figure imgf000328_0001
Formula (I) wherein, W is selected from 0, S and Se; J is selected from S and Se;
R-i is selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted;
R2 is selected from the group consisting of cycloalkyl, aryl, heteroaryl and heterocyclyl, all of which may be optionally substituted; and both R-i is directly bonded to J and R2 is directly bonded to the adjacent nitrogen, via a carbon atom .
2. The compound of claim 1 wherein R-i is selected from the group consisting of C5 or C6 cycloalkyl, 5-membered or 6-membered heteroaryl, bicyclic heteroaryl wherein at least one ring is heteroaryl, phenyl, biphenyl, phenylheterocyclyl, 5-membered or 6-membered heterocyclyl, and heterocyclylcycloalkyl, all of which may be optionally substituted.
3. The compound of claim 1 or claim 2 wherein Ri is selected from the group consisting of pyrazole, furan, tetrahydrofuran, tetrahydropyran, pyran, pyrrolidine, pyrrole, triazole, tetrazole, imidazole, pyridine, morpholine, piperazine, piperidine, substituted phenyl, phenylheteroaryl, phenylheterocyclyl, biphenyl, quinoline, isoquinoline, naphthyl, pyrazine and pyrimidine, all of which may be optionally substituted as appropriate.
4. The compound of any one of the preceding claims wherein Ri is 5- membered heterocyclyl or heteroaryl, each of which may be optionally substituted, comprising at least one ring heteroatom selected from N, 0 and S.
5. The compound of claim 4 wherein R-i is 5-membered nitrogen heterocyclyl or 5-membered nitrogen heteroaryl, each of which may be optionally substituted.
6. The compound of claim 4 or claim 5 wherein Ri is 5-membered heterocyclyl or 5-membered heteroaryl, each of which may be optionally substituted, comprising at least two ring nitrogen atoms.
7. The compound of any one of the preceding claims wherein R2 is selected from the group consisting of bicyclic and tricyclic hydrocarbons, 5-, 6- and 7- membered heterocycles or heteroaryls, each of which may be optionally substituted, and substituted phenyl.
8. The compound of any one of the preceding claims wherein R2 is selected from:
Figure imgf000329_0001
wherein, each incidence of Y is independently selected from C, N, S and 0, which may be optionally substituted as appropriate;
R5, R-i-i , R-I2, R-I 3, R-I4 and R 5 are independently selected from the group consisting of hydrogen, halo, cyano, amide, sulphonamide, acyl, hydroxyl, C-i- Ce alkyI, C1-C6 haloalkyi, C3-C5 cyloalkyi, d-Ce alkoxy all of which groups may be optionally substituted, as appropriate, with halo, cyano or CrC6 alkoxy; and wherein R-n and R-12 may combine to form phenyl, a 5- or 6-membered oxygen heterocycle or a 5- or 6-membered nitrogen heteroaryl, each of which may be optionally substituted;
R-12 and R-13 may combine to form a 5- or 6-membered nitrogen heteroaryl, which may be optionally substituted; and
R-14 and R 5 may combine to form a 5- or 6-membered cycloalkyl ring, phenyl, a 5- or 6-membered oxygen heterocycle or a 5- or 6-membered nitrogen heteroaryl, each of which may be optionally substituted.
9. The compound of any one of the preceding claims wherein Ri is selected from the group consisting of:
Figure imgf000330_0001
329
Figure imgf000331_0001
Figure imgf000332_0001
and in combination with each such R-i group, R2 may be independently selected from the group consisting of:
Figure imgf000332_0002
Figure imgf000333_0001
10. The compound of any one of the preceding claims wherein R2 is selected from a substituted or hydrogenated indacene, a 2,6-dialkylphenyl, a 2,6-dialkyl- 4-halophenyl, 2,6-dicycloalkylphenyl, and a 2,6-dicycloalkyl-4-halophenyl.
1 1. The compound of any one of the preceding claims wherein R2 is selected from hexahydroindacene, 2,6-diisopropylphenyl, 2,6-diisopropyl-4-chlorophenyl, 2,6-dicyclopropylphenyl and 2,6-dicyclopropyl-4-chlorophenyl.
12. The compound of any one of the preceding claims wherein J is a sulphur atom.
13. The compound of any one of the preceding claims wherein W is an oxygen atom.
14. The compound of any one of the preceding claims wherein the compound is a compound of formula (la), (lb) or (Ic), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000334_0001
formula (la) formula (lb) formula (lc) wherein, R-i is as defined in any one of the preceding claims.
15. The compound of claim 14 wherein Ri is selected from the group consisting of pyrazole, furan, tetrahydrofuran, tetrahydropyran, pyran, pyrrolidine, pyrrole, triazole, tetrazole, imidazole, pyridine, morpholine, piperazine, piperidine, substituted phenyl, phenylheteroaryl, phenylheterocydyl, biphenyl, quinoline, isoquinoline, naphthyl, pyrazine and pyrimidine, all of which may be optionally substituted as appropriate.
The compound of claim 14 or claim 15 wherein Ri is selected from the consisting of:
Figure imgf000334_0002
333
Figure imgf000335_0001
Figure imgf000336_0001
17. The compound of any one of the preceding claims wherein the compound is a compound of formula (II), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000336_0002
Formula (II) wherein, A, B, D and E are independently selected from C, N, 0, S and Se, but at least one thereof is C; each dashed line may represent a bond
R2 is as defined in any one of the preceding claims or R2 may be a fluorescent group; each incidence of R6 is independently selected from the group consisting of hydrogen, halide, cyano, C1-C6 alkyl, C1 -C6 alkylamino, C1 -C6 alkylhydroxy, C3-C6 cycloalkyl, alkylphenyl, phenyl, benzyl, C C6 ester, C2-C6 alkenyl, C C6 t fluoroalkyl and
Figure imgf000336_0003
alkoxy, each of which may be optionally substituted, as appropriate, or R6 may be a fluorescent group.
18. The compound of claim 17 wherein the compound of formula (II), is selected from the group consisting of:
Figure imgf000337_0001
Figure imgf000338_0001
Figure imgf000338_0002
wherein, R40 is selected from H, alkyl and halo; R4i is selected from H, alkyl and cycloalkyi; each incidence of P is independently selected from C, 0 or S; and wherein each incidence of R6, when present, is independently selected from those groups defined in claim 17.
19. The compound of any one of the preceding claims wherein the compound is selected from a compound of formula (lla), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000339_0001
Formula (lla) wherein R-n R 2 R13 Ru and R 5 are as defined in any one of claim 8 to claim 1 1 ;
A, B, D and E are selected from N and C and at least two of A, B, D, and E are N;
each incidence of R6 is independently selected from the group consisting of hydrogen, halide, cyano, C1-C6 alkyl, C1 -C6 alkylamino, C1 -C6 alkylhydroxy, C3-C6 cycloalkyl, alkylphenyl, phenyl, benzyl, C C6 ester, C2-C6 alkenyl, C C6 trifluoroalkyl and C1-C6 alkoxy, each of which may be optionally substituted.
20. The compound of any one of the preceding claims wherein the compound is a compound of formula (Mb), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000340_0001
Formula (lib) wherein Y and R5 are as defined in any one of claim 8 to claim 1 1 ; A, B, D and E are selected from N and C and at least two of A, B, D, and E are N;
each incidence of R6 is independently selected from the group consisting of hydrogen, halide, cyano, C C6 alkyl, Ci -C6 alkylamino, Ci -C6 alkylhydroxy, C3-C6 cycloalkyl, alkylphenyl, phenyl, benzyl, C1-C6 ester, C2-C6 alkenyl, C1-C6 trifluoroalkyi and C C6 alkoxy, each of which may be optionally substituted.
21. The compound of any one of the preceding claims wherein the compound is a compound of formula (Ilia), (lllb) or (lllc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000340_0002
Formula (lllb)
Figure imgf000340_0003
Formula (lllc) wherein, R2i is selected from H, alkyl, perhaloalkyl or hydroxylalkyl; R22 is selected from H, alkyl, perhaloalkyl, C3-C6 cycloalkyl, phenyl or benzyl;
R-I 8 is H or halogen;
R-I6 and Ri7 are H, alkyl or cycloalkyl; or R-|6 and R-|7, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
R-I 9 and R20 are H or alkyl; or R19 and R20, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
provided that R21 and R22 are not both H; and
provided that R 6, Ri7, R18, R19 and R20 are not all H.
22. The compound of claim 21 wherein:
R21 is selected from H, alkyl, perhaloalkyl or hydroxylalkyl;
R22 is selected from H, alkyl, perhaloalkyl, C3-C6 cycloalkyl, phenyl or benzyl;
R-I6 and R-|7, together with the atoms to which they are attached, form a cyclopentyl ring;
R-19 and R2o, together with the atoms to which they are attached, form a cyclopentyl ring;
R-I 8 is H or halogen; and
provided that R2i and R22 are not both H.
23. The compound of claim 21 wherein:
R21 is selected from H, alkyl, perhaloalkyl or hydroxylalkyl;
R22 is selected from H, alkyl, perhaloalkyl, C3-C6 cycloalkyl, phenyl and benzyl; R-16 and R2o are Ci-6 alkyl or C3-5 cycloalkyl;
R 7 and R 9 are H,
R-I 8 is H or halogen; and
provided that R2i and R22 are not both H.
24. The compound of any one of the preceding claims wherein the compound is a compound of formula (IVa), (IVb) or (IVc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000342_0001
Formula (IVc) wherein, R2i and R22 are selected from H, alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl or R2 and R22, together with the carbon atoms to which they are attached, may form a cyclopentyl or a cyclohexyl ring;
R-I 8 is H or halogen;
R-16 and R-|7 are H, alkyl or cycloalkyl; or R-|6 and R-|7, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S; R-I 9 and R2o are H or alkyl; or R-|9 and R2o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
provided that R2i and R22 are not both H; and
provided that R 6, R-17, R18, R19 and R20 are not all H.
25. The compound of claim 24 wherein:
R2 and R22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyi, C3-C6 cycloalkyi, phenyl and benzyl;
R-I 6 and R-17, together with the atoms to which they are attached, form a cyclopentyl ring;
R-19 and R20, together with the atoms to which they are attached, form a cyclopentyl ring;
R-I 8 is H or halogen; and
provided that R2i and R22 are not both H.
26. The compound of claim 24 wherein:
R2i and R22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyi, C3-C6 cycloalkyi, phenyl and benzyl; preferably the perhaloalkyi and hydroxylalkyi are C1 -6 perhaloalkyi and hydroxylalkyi;
R-I 6 and R20 are Ci-6 alkyl or C3-5 cycloalkyi;
Figure imgf000343_0001
R-I 8 is H or halogen;
provided that R2i and R22 are not both H.
27. The compound of any one of the preceding claims wherein the compound is a compound of formula (Va), (Vb) or (Vc), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000344_0001
Formula Vc wherein, R2i and R22 are selected from H, alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl;
R-I 8 is H or halogen;
R-I6 and R 7 are H, alkyl or cycloalkyl; or R 6 and R 7, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
R-19 and R20 are H or alkyl; or R 9 and R2o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
provided that R2i and R22 are not both H; and
provided that R-|6, R17, R18, R19 and R20 are not all H.
28. The compound of claim 27 wherein:
R2i and R22 are selected from H, alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl;
R-16 and R 7, together with the atoms to which they are attached, form a cyclopentyl ring;
R-I 9 and R2o, together with the atoms to which they are attached, form a cyclopentyl ring;
R-i s is H or halogen; and
provided that R2i and R22 are not both H.
29. The compound of claim 27 wherein:
R21 and R22 are selected from H, alkyl, perhaloalkyi, hydroxylalkyi, C3-C6 cycloalkyl, phenyl and benzyl;
R-16 and R2o are C1-6 alkyl or C3-5 cycloalkyl;
R 7 and R 9 are H;
R-i s is H or halogen; and
provided that R2i and R22 are not both H.
30. The compound of any one of the preceding claims wherein the compound is a compound of formula (Via) or (VIb), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000345_0001
Formula (Via) Formula (VIb) wherein, R22 is selected from alkyl, perhaloalkyi, hydroxylalkyi, C3-C6 cycloalkyl, phenyl and benzyl;
R-i s is H or halogen; R-16 and R-i7 are H, alkyl or cycloalkyl; or Ri6 and Ri 7, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
R-I 9 and R2o are H or alkyl; or R-|9 and R2o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S; and
provided that Ri6, R17, R18, R19 and R20 are not all H.
31. The compound of claim 30 wherein:
R22 is selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl;
R-16 and R-|7, together with the atoms to which they are attached, form a cyclopentyl ring;
R-19 and R2o, together with the atoms to which they are attached, form a cyclopentyl ring; and
R-I 8 is H or halogen.
32. The compound of claim 30 wherein:
R22 is selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, phenyl and benzyl;
R-16 and R2o are C1-6 alkyl or C3-5 cycloalkyl, preferably isopropyl or cyclopentyl;
Figure imgf000346_0001
R-I 8 is H or halogen.
33. The compound of any one of the preceding claims wherein the compound is a compound of formula (VII), or a pharmaceutically acceptable salt, solvate or prodrug thereof:
Figure imgf000347_0001
Formula (VII) wherein, Q is 0 or S; each incidence of R30 is independently selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, and alkylamino;
R-18 is H or halogen;
R-16 and R 7 are H or alkyl; or R 6 and R 7, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
R-I 9 and R20 are H or alkyl; or R 9 and R2o, together with the carbon atoms to which they are attached, form a 5 or 6 membered ring, said ring being saturated, partially unsaturated or unsaturated, said ring optionally comprising one or two heteroatoms selected from N, 0 and S;
provided that R 6, Ri7, R-is, R19 and R20 are not all H; and provided that when Q is 0 and R-|6 and R-|7, and separately R-|9 and R2o, together with the respective carbon atoms to which they are attached, form a cyclopentyl ring then R30 is not C-3 hydroxylalkyl.
34. The compound of claim 33 wherein:
Q is 0 or S; each incidence of R30 is independently selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, and alkylamino;
R 6 and R 7, together with the atoms to which they are attached, form a cyclopentyl ring;
R-19 and R2o, together with the atoms to which they are attached, form a cyclopentyl ring; and
R-I 8 is H or halogen; and
provided that when Q is 0 then R30 is not C-3 hydroxylalkyl.
35. The compound of claim 33 or claim 34 wherein: Q is 0 or S; each incidence of R30 is independently selected from alkyl, perhaloalkyl, hydroxylalkyl, C3-C6 cycloalkyl, and alkylamino;
R-I 6 and R2o are Ci-6 alkyl, preferably isopropyl;
Figure imgf000348_0001
R-I 8 is H or halogen.
36. The compound of any one of the preceding claims wherein the compound is selected from the group consisting of:
Figure imgf000348_0002
347
Figure imgf000349_0001
348
Figure imgf000350_0001
349
Figure imgf000351_0001
350
Figure imgf000352_0001
Figure imgf000353_0001
352
Figure imgf000354_0001
353
Figure imgf000355_0001
354
Figure imgf000356_0001
355
Figure imgf000357_0001
356
Figure imgf000358_0001
Figure imgf000358_0002
Figure imgf000359_0001
358
Figure imgf000360_0001
359
Figure imgf000361_0001
360
Figure imgf000362_0001
Figure imgf000363_0001
Figure imgf000363_0002
Figure imgf000364_0001
37. The compound of any one of the preceding claims wherein the compound, or pharmaceutically effective salt, solvate or prodrug thereof, is an inhibitor of the NLRP3 inflammasome.
38. A pharmaceutical composition comprising a compound of any one of claim 1 to claim 37, or a pharmaceutically acceptable salt, solvate or prodrug thereof, and a pharmaceutically acceptable carrier, diluent and/or excipient.
39. A method of treatment or prevention of a disease, disorder or condition including the step of administering an effective amount of a compound of any one of claim 1 to claim 37, or a pharmaceutically effective salt, solvate or prodrug thereof , or the pharmaceutical composition of claim 38, to thereby treat or prevent the disease disorder or condition.
40. The method of claim 39 wherein the disease, disorder or condition is one which is responsive to inhibition of activation of the NLRP3 inflammasome.
41. The method of claim 39 or claim 40 wherein the disease, disorder or condition is responsive to modulation of one or more of IL-1 β, IL-17, IL-18, IL- 1 a, IL-37, IL-33 and Th17 cells.
42. The method of any one of claim 39 to claim 41 wherein the disease, disorder or condition is a disease, disorder or condition of the immune system.
43. The method of any one of claim 39 to claim 41 wherein the disease, disorder or condition is an inflammatory disease disorder or condition or an autoimmune disease disorder or condition.
44. The method of any one of claim 39 to claim 41 wherein the disease, disorder or condition is a disease, disorder or condition of the skin.
45. The method of any one of claim 39 to claim 41 wherein the disease, disorder or condition is a disease, disorder or condition of the cardiovascular system .
46. The method of any one of claim 39 to claim 41 wherein the disease, disorder or condition is a cancer, tumour or other malignancy.
47. The method of any one of claim 39 to claim 41 wherein the disease, disorder or condition is a disease, disorder or condition is of the renal system.
48. The method of any one of claim 39 to claim 41 wherein the disease, disorder or condition is a disease, disorder or condition is of the gastro-intestinal tract.
49. The method of any one of claim 39 to claim 41 wherein the disease, disorder or condition is a disease, disorder or condition is of the respiratory system .
50. The method of any one of claim 39 to claim 41 wherein the disease, disorder or condition is a disease, disorder or condition is of the endocrine system .
51. The method of any one of claim 39 to claim 41 wherein the disease, disorder or condition is a disease, disorder or condition is of the central nervous system (CNS).
52. The method of any one of claim 39 to claim 41 wherein the disease, disorder or condition is selected from the group consisting of constitutive inflammation including the cryopyrin-associated periodic syndromes (CAPS): Muckle-Wells syndrome (MWS), familial cold autoinflammatory syndrome (FCAS) and neonatal-onset multisystem inflammatory disease (NOMID); including autoinflammatory diseases: familial Mediterranean fever (FMF), TNF receptor associated periodic syndrome (TRAPS), mevalonate kinase deficiency (MKD), hyperimmunoglobulinemia D and periodic fever syndrome (HIDS), deficiency of interleukin 1 receptor (DIRA) antagonist, Majeed syndrome, pyogenic arthritis, pyoderma gangrenosum and acne (PAPA), haploinsufficiency of A20 (HA20), pediatric granulomatous arthritis (PGA), PLCG2-associated antibody deficiency and immune dysregulation (PLAID), PLCG2-associated autoinflammation, antibody deficiency and immune dysregulation (APLAID), sideroblastic anemia with B-cell immunodeficiency, periodic fevers, and developmental delay (SIFD); Sweet's syndrome, chronic nonbacterial osteomyelitis (CNO), chronic recurrent multifocal osteomyelitis (CRMO) and synovitis, acne, pustulosis, hyperostosis, osteitis syndrome (SAPHO); autoimmune diseases including multiple sclerosis (MS), type-1 diabetes, psoriasis, rheumatoid arthritis, Behcet's disease, Sjogren's syndrome and Schnitzler syndrome; respiratory diseases including chronic obstructive pulmonary disorder (COPD), steroid-resistant asthma, asbestosis, silicosis and cystic fibrosis; central nervous system diseases including Parkinson's disease, Alzheimer's disease, motor neuron disease, Huntington's disease, cerebral malaria and brain injury from pneumococcal meningitis; metabolic diseases including Type 2 diabetes, atherosclerosis, obesity, gout, pseudo-gout; ocular diseases including those of the ocular epithelium, age-related macular degeneration (AMD), corneal infection, uveitis and dry eye; kidney disease including chronic kidney disease, oxalate nephropathy and diabetic nephropathy; liver disease including non-alcoholic steatohepatitis and alcoholic liver disease; inflammatory reactions in skin including contact hypersensitivity and sunburn; inflammatory reactions in the joints including osteoarthritis, systemic juvenile idiopathic arthritis, adult-onset Still's disease, relapsing polychondritis; viral infections including alpha virus including Chikungunya and Ross River, and flavivirus including Dengue and Zika viruses, flu, HIV; hidradenitis suppurativa (HS) and other cyst-causing skin diseases; cancers including lung cancer metastasis, pancreatic cancers, gastric cancers, myelodisplastic syndrome, leukemia; polymyositis; stroke; myocardial infarction; Graft versus Host Disease; hypertension; colitis; helminth infection; bacterial infection; abdominal aortic aneurism; wound healing; depression, psychological stress; pericarditis including Dressler's syndrome, ischaemia reperfusion injury and any disease where an individual has been determined to carry a germ line or somatic non-silent mutation in NLRP3.
53. The method of any one of claim 39 to claim 52 wherein the treatment or prevention of the disease, disorder or condition is performed on a mammal.
54. The method of claim 53 wherein the mammal is a human subject.
55. A method of diagnosing a disease, disorder or condition in a mammal including the step of administering a labelled compound of any one of claim 1 to claim 37, or a pharmaceutically effective salt, solvate or prodrug thereof, or metal ion chelate complex thereof, to the mammal or to a biological sample obtained from the mammal to facilitate diagnosis of the disease disorder or condition in the mammal.
56. A method of modulating the activity of a biological target comprising the step of exposing the biological target to a compound of any one of claim 1 to claim 37, or a pharmaceutically effective salt, solvate or prodrug thereof.
57. The method of claim 56 wherein the biological target may be selected from the group consisting of the NLRP3 inflammasome, IL-Ι β, IL-17, IL-18, IL- 1 a, IL-37, IL-33 and Th17 cells.
PCT/AU2016/050103 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same WO2016131098A1 (en)

Priority Applications (35)

Application Number Priority Date Filing Date Title
RU2017128287A RU2739356C2 (en) 2015-02-16 2016-02-16 Sulfonyl urea and related compounds and use thereof
MDE20180009T MD3259253T2 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
CN201680010446.XA CN107428696B (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and uses thereof
LTEP16751821.6T LT3259253T (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
EP21152421.0A EP3888749A1 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
US15/551,264 US10538487B2 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
EP16751821.6A EP3259253B1 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
SI201630635T SI3259253T1 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
SG11201706664QA SG11201706664QA (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
MEP-2020-49A ME03737B (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
RS20200258A RS60048B1 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
PL19187141T PL3578547T3 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
PE2021001430A PE20221627A1 (en) 2015-02-16 2016-02-16 SULFONYLUREAS AND RELATED COMPOUNDS AND USE OF THESE
KR1020177025608A KR20170109678A (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and uses thereof
MX2017010528A MX2017010528A (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same.
CA2975192A CA2975192A1 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
NZ733948A NZ733948A (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
ES16751821T ES2777626T3 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and their use
IL273065A IL273065B2 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
EP19187141.7A EP3578547B1 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
PL16751821T PL3259253T3 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
AU2016222278A AU2016222278B2 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
DK16751821.6T DK3259253T3 (en) 2015-02-16 2016-02-16 SULPHONYLURAES AND RELATED COMPOUNDS AND USE OF SAME
IL253661A IL253661B2 (en) 2015-02-16 2016-02-16 Sulfonylureas bearing a five-membered ring, and pharmaceutical compositions and uses thereof
MYPI2017001188A MY193765A (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
JP2017560843A JP6929792B2 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and their use
MA41553A MA41553B1 (en) 2015-02-16 2016-02-16 Sulfonylureas, Related Compounds, and Their Use
BR112017017610-6A BR112017017610A2 (en) 2015-02-16 2016-02-16 compound, pharmaceutical composition, treatment method, diagnostic method, activity modulation method
HK18107901.3A HK1249501A1 (en) 2015-02-16 2018-06-20 Sulfonylureas and related compounds and use of same
US16/535,002 US11130731B2 (en) 2015-02-16 2019-08-07 Sulfonylureas and related compounds and use of same
HRP20200214TT HRP20200214T1 (en) 2015-02-16 2020-02-10 Sulfonylureas and related compounds and use of same
CY20201100252T CY1122832T1 (en) 2015-02-16 2020-03-18 SULFONYLUREAS AND RELATED COMPOUNDS AND THEIR USE
AU2020203464A AU2020203464B2 (en) 2015-02-16 2020-05-26 Sulfonylureas and related compounds and use of same
US17/405,989 US20220112159A1 (en) 2015-02-16 2021-08-18 Sulfonylureas and related compounds and use of same
AU2021258033A AU2021258033A1 (en) 2015-02-16 2021-10-28 Sulfonylureas and related compounds and use of same

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
AU2015900507A AU2015900507A0 (en) 2015-02-16 Sulfonylureas and related compounds and use of same
AU2015900507 2015-02-16

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US15/551,264 A-371-Of-International US10538487B2 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same
US16/535,002 Continuation US11130731B2 (en) 2015-02-16 2019-08-07 Sulfonylureas and related compounds and use of same

Publications (2)

Publication Number Publication Date
WO2016131098A1 true WO2016131098A1 (en) 2016-08-25
WO2016131098A8 WO2016131098A8 (en) 2017-03-16

Family

ID=56691944

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2016/050103 WO2016131098A1 (en) 2015-02-16 2016-02-16 Sulfonylureas and related compounds and use of same

Country Status (31)

Country Link
US (3) US10538487B2 (en)
EP (3) EP3888749A1 (en)
JP (2) JP6929792B2 (en)
KR (1) KR20170109678A (en)
CN (3) CN107428696B (en)
AU (3) AU2016222278B2 (en)
BR (1) BR112017017610A2 (en)
CA (1) CA2975192A1 (en)
CL (2) CL2017002097A1 (en)
CY (1) CY1122832T1 (en)
DK (2) DK3578547T3 (en)
ES (2) ES2881228T3 (en)
HK (1) HK1249501A1 (en)
HR (2) HRP20200214T1 (en)
HU (1) HUE055755T2 (en)
IL (2) IL273065B2 (en)
LT (2) LT3259253T (en)
MA (3) MA56473A (en)
MD (1) MD3259253T2 (en)
ME (1) ME03737B (en)
MX (2) MX2017010528A (en)
MY (2) MY193765A (en)
NZ (2) NZ772349A (en)
PE (2) PE20221627A1 (en)
PL (2) PL3259253T3 (en)
PT (2) PT3259253T (en)
RS (2) RS62164B1 (en)
RU (1) RU2739356C2 (en)
SG (2) SG10202002599XA (en)
SI (2) SI3578547T1 (en)
WO (1) WO2016131098A1 (en)

Cited By (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017140778A1 (en) 2016-02-16 2017-08-24 The University Of Queensland Sulfonylureas and related compounds and use of same
WO2017184623A1 (en) 2016-04-18 2017-10-26 Ifm Therapeutics, Inc Compounds and compositions for treating conditions associated with nlrp activity
WO2018103583A1 (en) * 2016-12-05 2018-06-14 Zibo Anxuan Pharmaceutical Science And Technology Co., Ltd. N-hydroxy-benzene-sulfonamide derivatives and their uses thereof
WO2018136890A1 (en) * 2017-01-23 2018-07-26 Jecure Therapeutics, Inc. Chemical compounds as inhibitors of interleukin-1 activity
WO2018167468A1 (en) * 2017-03-13 2018-09-20 NodThera Limited Chemical compounds
WO2018215818A1 (en) * 2017-05-24 2018-11-29 The University Of Queensland Novel compounds and uses
WO2018225018A1 (en) 2017-06-09 2018-12-13 Cadila Healthcare Limited Novel substituted sulfoximine compounds
WO2019008029A1 (en) * 2017-07-07 2019-01-10 Inflazome Limited Sulfonylureas and sulfonylthioureas as nlrp3 inhibitors
WO2019008025A1 (en) * 2017-07-07 2019-01-10 Inflazome Limited Novel sulfonamide carboxamide compounds
WO2019023147A1 (en) * 2017-07-24 2019-01-31 IFM Tre, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2019023145A1 (en) * 2017-07-24 2019-01-31 IFM Tre, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2019025467A1 (en) * 2017-07-31 2019-02-07 NodThera Limited Selective inhibitors of nlrp3 inflammasome
WO2019034697A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Novel sulfonamide carboxamide compounds
WO2019034690A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Sulfonylureas and sulfonylthioureas as nlrp3 inhibitors
WO2019034692A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Sulfonylureas and sulfonylthioureas as nlrp3 inhibitors
WO2019034686A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Novel sulfonamide carboxamide compounds
WO2019034688A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Novel sulfonamide carboxamide compounds
WO2019034693A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Sulfonylureas and sulfonylthioureas as nlrp3 inhibitors
WO2019034696A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Novel sulfonamide carboxamide compounds
WO2019043610A1 (en) 2017-08-31 2019-03-07 Cadila Healthcare Limited Novel substituted sulfonylurea derivatives
WO2019068772A1 (en) * 2017-10-03 2019-04-11 Inflazome Limited Novel compounds
WO2019092171A1 (en) * 2017-11-09 2019-05-16 Inflazome Limited Novel sulfonamide carboxamide compounds
WO2019092172A1 (en) * 2017-11-09 2019-05-16 Inflazome Limited Novel sulfonamide carboxamide compounds
WO2019092170A1 (en) * 2017-11-09 2019-05-16 Inflazome Limited Novel sulfonamide carboxamide compounds
WO2019166619A1 (en) * 2018-03-02 2019-09-06 Inflazome Limited Novel compounds
WO2019166627A1 (en) 2018-03-02 2019-09-06 Inflazome Limited Novel compounds
WO2019166633A1 (en) 2018-03-02 2019-09-06 Inflazome Limited Sulfonamide derivates as nlrp3 inhibitors
WO2019182981A1 (en) * 2018-03-21 2019-09-26 Olatec Therapeutics Llc Methods for treating melanoma
WO2019206871A1 (en) 2018-04-23 2019-10-31 Inflazome Limited A sodium salt of n-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 -isopropyl-1 h-pyrazole-3-sulfonamide
WO2019209693A1 (en) 2018-04-23 2019-10-31 Corcept Therapeutics, Inc. Methods of preparing regioselective n-alkyl triazoles
WO2019211463A1 (en) 2018-05-04 2019-11-07 Inflazome Limited Novel compounds
WO2020010143A1 (en) 2018-07-03 2020-01-09 Novartis Inflammasome Research, Inc. Nlrp modulators
WO2020010118A1 (en) * 2018-07-03 2020-01-09 Novartis Inflammasome Research, Inc. Methods of treating or selecting a treatment for a subject resistant to tnf inhibitor using a nlrp3 antagonist
WO2020010140A1 (en) * 2018-07-03 2020-01-09 Novartis Inflammasome Research, Inc. Nlrp modulators
EP3259253B1 (en) 2015-02-16 2020-01-15 The University of Queensland Sulfonylureas and related compounds and use of same
WO2020018970A1 (en) * 2018-07-20 2020-01-23 Genentech, Inc. Sulfonylurea compounds as inhibitors of interleukin-1 activity
WO2020018975A1 (en) * 2018-07-20 2020-01-23 Genentech, Inc. Sulfonimidamide compounds as inhibitors of interleukin-1 activity
WO2020035466A1 (en) 2018-08-15 2020-02-20 Inflazome Limited Novel sulfoneurea compounds
WO2020035464A1 (en) 2018-08-15 2020-02-20 Inflazome Limited Novel sulfonamideurea compounds
WO2020079207A1 (en) * 2018-10-19 2020-04-23 Inflazome Limited Novel processes
WO2020086732A1 (en) 2018-10-24 2020-04-30 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2020102576A1 (en) * 2018-11-16 2020-05-22 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2020102096A1 (en) * 2018-11-13 2020-05-22 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2020102098A1 (en) * 2018-11-13 2020-05-22 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2020102574A1 (en) * 2018-11-16 2020-05-22 Novartis Inflammasome Research, Inc. The compounds and compositions for treating conditions associated with nlrp activity
WO2020102100A1 (en) * 2018-11-13 2020-05-22 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2020104657A1 (en) 2018-11-23 2020-05-28 Inflazome Limited Nlrp3 inhibitors
WO2020148619A1 (en) 2019-01-14 2020-07-23 Cadila Healthcare Limited Novel substituted sulfonylurea derivatives
WO2020154321A1 (en) * 2019-01-22 2020-07-30 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2020152361A1 (en) * 2019-01-25 2020-07-30 NodThera Limited Carbamate derivatives and uses thereof
JP2020527139A (en) * 2017-07-14 2020-09-03 イネイト・テューマー・イミュニティ・インコーポレイテッドInnate Tumor Immunity, Inc. NLRP3 modulator
WO2020178441A1 (en) * 2019-03-06 2020-09-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Inhibitors of ngal protein
WO2020208249A1 (en) 2019-04-12 2020-10-15 Inflazome Limited Nlrp3 inflammasome inhibition
WO2020254697A1 (en) 2019-06-21 2020-12-24 Ac Immune Sa Fused 1,2 thiazoles and 1,2 thiazines which act as nl3p3 modulators
WO2021002887A1 (en) 2019-07-02 2021-01-07 Novartis Inflammasome Research, Inc. Gut-targeted nlrp3 antagonists and their use in therapy
WO2021009567A1 (en) * 2019-07-17 2021-01-21 Zomagen Biosciences Ltd Nlrp3 modulators
WO2021009566A1 (en) * 2019-07-17 2021-01-21 Zomagen Biosciences Ltd N-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4,5,6,7-tetrahydrobenzofuran -2-sulfonamide derivatives and related compounds as nlpr3 modulators for the treatment of multiple sclerosis (ms)
JP2021506966A (en) * 2017-12-18 2021-02-22 ノッドセラ リミテッド Sulfonyl urea derivative as NLRP3 inflammasome regulator
WO2021032591A1 (en) 2019-08-16 2021-02-25 Inflazome Limited Macrocyclic sulfonylurea derivatives useful as nlrp3 inhibitors
WO2021043966A1 (en) 2019-09-06 2021-03-11 Inflazome Limited Nlrp3 inhibitors
WO2021048809A1 (en) * 2019-09-12 2021-03-18 Cadila Healthcare Limited Novel substituted sulfoximine derivatives
WO2021089781A1 (en) * 2019-11-07 2021-05-14 Inflazome Limited Treatment or prevention of psychiatric brain disorders using the nlrp3 inhibitor n-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 -isopropyl-1 h-pyrazole-3-sulfonamide
WO2021089769A1 (en) * 2019-11-07 2021-05-14 Inflazome Limited Treatment and prevention of neuroinflammation or an inflammatory brain disorder
WO2021089776A1 (en) * 2019-11-07 2021-05-14 Inflazome Limited Treatment and prevention of a traumatic brain disorder
WO2021089783A1 (en) * 2019-11-07 2021-05-14 Inflazome Limited Treatment of arthritis
WO2021089782A1 (en) * 2019-11-07 2021-05-14 Inflazome Limited Treatment of autoinflammatory disorders
WO2021089768A2 (en) 2019-11-07 2021-05-14 Inflazome Limited Novel treatment
US20210147349A1 (en) * 2018-03-02 2021-05-20 Inflazome Limited Phenylsulfonylurea derivatives useful as nlrp3 inhibitors
WO2021111351A1 (en) * 2019-12-03 2021-06-10 Cadila Healthcare Limited Novel substituted sulfonylurea and sulfoximineurea derivatives
WO2021150574A1 (en) * 2020-01-22 2021-07-29 Genentech, Inc. Sulfonimidamide compounds as nlrp3 modulators
WO2021165245A1 (en) 2020-02-18 2021-08-26 Inflazome Limited Compounds
WO2021185912A1 (en) 2020-03-19 2021-09-23 Softhale Nv Method for the treatment nlrp3-associated diseases
WO2021249337A1 (en) 2020-06-11 2021-12-16 南京明德新药研发有限公司 Dimethylsulfoximine derivative
WO2021255279A1 (en) 2020-06-19 2021-12-23 Ac Immune Sa D i h yd rooxazo le and thiourea derivatives modulating the nlrp3 inflammasome pathway
WO2022023907A1 (en) 2020-07-31 2022-02-03 Novartis Ag Methods of selecting and treating patients at elevated risk of major adverse cardiac events
WO2022115417A1 (en) * 2020-11-25 2022-06-02 VenatoRx Pharmaceuticals, Inc. Sulfonyl urea nlrp3 inflammasome inhibitors
CN114761383A (en) * 2019-06-12 2022-07-15 诺瑟拉有限公司 Sulfonamide derivatives and uses thereof
WO2022171185A1 (en) * 2021-02-10 2022-08-18 杭州英创医药科技有限公司 Compound serving as nlrp3 inhibitor
IT202100011237A1 (en) 2021-05-03 2022-11-03 Univ Degli Studi Di Torino NLRP3 INFLAMMASOME INHIBITOR COMPOUNDS AND THEIR USE
US11530200B2 (en) 2018-03-02 2022-12-20 Inflazome Limited Compounds
RU2786719C2 (en) * 2017-07-24 2022-12-26 Новартис Аг Compounds and compositions for the treatment of conditions associated with nlrp activity
WO2022269010A1 (en) 2021-06-23 2022-12-29 F. Hoffmann-La Roche Ag A crystalline potassium salt of 1-ethyl- n -((1,2,3,5,6,7-hexahydro- s -indacen-4-yl)carbamoyl)piperidine-4 -sulfonamide
CN115772173A (en) * 2022-12-20 2023-03-10 武汉国粹医药科技有限公司 Benzofuran compound, preparation method and application thereof, and antibacterial agent
US11603375B2 (en) 2020-03-16 2023-03-14 Zomagen Biosciences Ltd NLRP3 modulators
WO2023037024A1 (en) * 2021-09-08 2023-03-16 Fundacion Para La Investigacion Biomedica Del Hospital Universitario De La Princesa (90%) N-sulfonylurea derivatives and the therapeutic use thereof
US11618751B1 (en) 2022-03-25 2023-04-04 Ventus Therapeutics U.S., Inc. Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 derivatives
KR20230066899A (en) 2021-11-08 2023-05-16 제일약품주식회사 Novel compounds as nlrp3 inhibitor and pharmaceutical composition comprising the same
WO2023118521A1 (en) 2021-12-22 2023-06-29 Ac Immune Sa Dihydro-oxazol derivative compounds
WO2023230002A1 (en) * 2022-05-23 2023-11-30 VenatoRx Pharmaceuticals, Inc. Nlrp3 inflammasome inhibitors
WO2024013395A1 (en) 2022-07-14 2024-01-18 Ac Immune Sa Pyrrolotriazine and imidazotriazine derivatives as modulators of the nlrp3 inflammasome pathway
US11884645B2 (en) 2018-03-02 2024-01-30 Inflazome Limited Sulfonyl acetamides as NLRP3 inhibitors
WO2024023266A1 (en) 2022-07-28 2024-02-01 Ac Immune Sa Novel compounds
KR20240022938A (en) 2022-08-12 2024-02-20 제일약품주식회사 Novel compounds as nlrp3 inhibitor and pharmaceutical composition comprising the same

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR3046933B1 (en) * 2016-01-25 2018-03-02 Galderma Research & Development NLRP3 INHIBITORS FOR THE TREATMENT OF INFLAMMATORY SKIN CONDITIONS
WO2019166624A1 (en) * 2018-03-02 2019-09-06 Inflazome Limited Novel compounds
CN108404117B (en) * 2018-05-29 2020-06-30 广东龙帆生物科技有限公司 Application of nucleotide-binding oligomerization domain-like receptor protein in medicament for treating Zika virus infection
CN111848461A (en) * 2019-04-29 2020-10-30 苏州大学 NLRP3 inflammasome inhibitor and preparation method and application thereof
JP7392169B2 (en) * 2019-11-12 2023-12-05 成都百裕制薬股▲ふん▼有限公司 Amide derivatives and their preparation methods and applications in medicine
CN111358778A (en) * 2020-03-17 2020-07-03 中国医科大学附属第一医院 Application of MCC950 in preparation of medicine for preventing or treating Alzheimer disease
WO2022022646A1 (en) * 2020-07-29 2022-02-03 南京明德新药研发有限公司 Selenium-containing five-membered heteroaromatic ring compound
WO2022098108A1 (en) * 2020-11-04 2022-05-12 (주) 업테라 Nlrp3 protein degradation inducing compound
FR3115682B1 (en) * 2020-11-05 2023-02-24 Roquette Freres Compositions based on methyl-cyclodextrins for the treatment and/or prevention of hepatic steatosis
KR20220112329A (en) * 2021-02-03 2022-08-11 환인제약 주식회사 Small Molecule Inhibitors of NLRP3 Inflammasome for Treatment of Alzheimer's Disease
WO2022237780A1 (en) * 2021-05-10 2022-11-17 成都百裕制药股份有限公司 Amide derivative and use thereof
TWI815439B (en) * 2021-05-10 2023-09-11 大陸商成都百裕製藥股份有限公司 Amide derivatives and applications thereof
CN116635373A (en) * 2021-05-10 2023-08-22 成都百裕制药股份有限公司 Amide derivatives and use thereof
WO2023098612A1 (en) * 2021-12-03 2023-06-08 南京明德新药研发有限公司 Salt form and crystal form of dimethyl sulfoximine derivative
WO2024010772A1 (en) * 2022-07-06 2024-01-11 Kodiak Sciences Inc. Nlrp3 inhibitors

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4018929A (en) * 1974-04-17 1977-04-19 A. Christiaens Societe Anonyme 3-Loweralkylcarbamylsulfonamido-4-phenylaminopyridine-N-oxides, derivatives thereof and pharmaceutical compositions containing same
US5169860A (en) * 1992-03-13 1992-12-08 Eli Lilly And Company Antitumor compositions and methods of treatment
WO1998032733A1 (en) * 1997-01-29 1998-07-30 Pfizer Inc. Sulfonyl urea derivatives and their use in the control of interleukin-1 activity
WO2001019390A1 (en) * 1999-09-14 2001-03-22 Pfizer Products Inc. Combination treatment with il-1ra and diaryl sulphonyl urea compounds
EP1236468A1 (en) * 2001-02-12 2002-09-04 Warner-Lambert Company Sulfonylaminocarbonyl derivatives for the treatment of nuclear factor-kappa B mediated diseases and disorders

Family Cites Families (87)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB797474A (en) 1955-07-20 1958-07-02 Boehringer & Soehne Gmbh Sulphonylurea derivatives
US3242174A (en) 1962-02-06 1966-03-22 Pfizer & Co C 1-(tertiary aminosulfonyl)-3-(hydrocarbon) ureas
US3305556A (en) 1962-11-23 1967-02-21 Pfizer & Co C Novel hypoglycemic agents
NL129208C (en) 1965-07-14
CH509991A (en) 1966-08-08 1971-07-15 Hoffmann La Roche Sulphonamides and process for the manufacture thereof
NL131473C (en) 1966-10-28
BE754588A (en) 1969-08-07 1971-02-08 Hoechst Ag THIOPHENE-SULFONYL-UREES AND THE MEDICINAL PRODUCTS CONTAINING THESE SUBSTANCES
FR2063472A5 (en) 1969-10-17 1971-07-09 Mercier Pierre
AU571869B2 (en) 1983-05-09 1988-04-28 E.I. Du Pont De Nemours And Company Pyridyl- and pyrimidyl- sulphonamides
EP0126711B1 (en) 1983-05-16 1989-08-02 Ciba-Geigy Ag Herbicidally active and plant growth regulating pyrimidine derivatives, their preparation and use
JPS6045573A (en) 1983-08-22 1985-03-12 Nissan Chem Ind Ltd Pyrazolesulfonylurea derivative, its preparation and selective herbicide containing said derivative
US4723991A (en) 1984-03-23 1988-02-09 E. I. Du Pont De Nemours And Company Lower alkyl 2-[[N-(3-cyano-pyridin-2-yl)aminocarbonyl]aminosulphonyl]benzoate derivatives having herbicidal activity
US4659369A (en) 1984-08-27 1987-04-21 E. I. Du Pont De Nemours And Company Herbicidal acetals and ketals
AU578307B2 (en) 1984-09-17 1988-10-20 E.I. Du Pont De Nemours And Company Herbicidal pyrazolesulfonamides
FI855180A (en) 1985-01-18 1986-07-19 Nissan Chemical Ind Ltd PYRAZOLESULFONAMIDDERIVAT, FOERFARANDE FOER DESS FRAMSTAELLANDE OCH DET INNEHAOLLANDE OGRAESGIFT.
JPH0720957B2 (en) 1985-11-26 1995-03-08 日産化学工業株式会社 Pyrazole sulfonylurea derivative, process and herbicide
JP2570686B2 (en) 1985-12-23 1997-01-08 日産化学工業株式会社 Pyrazole derivatives
US4830660A (en) 1986-06-19 1989-05-16 Nissan Chemical Industries, Ltd. Imidazolesulfonamide derivatives and herbicides
EP0262096B1 (en) 1986-09-26 1992-01-02 Ciba-Geigy Ag Aminopyrazinones and aminotriazinones
US4802908A (en) 1987-01-22 1989-02-07 E. I. Du Pont De Nemours And Company Herbicidal 2-(1H)-pyrazinones
EP0511993A1 (en) 1990-01-22 1992-11-11 E.I. Du Pont De Nemours And Company Herbicidal sulfonylureas
NZ238911A (en) * 1990-07-17 1993-10-26 Lilly Co Eli Furyl, pyrrolyl and thienyl sulphonyl urea derivatives, pharmaceutical compositions and intermediates therefor
EP0546082B1 (en) 1990-08-29 1996-12-18 E.I. Du Pont De Nemours And Company Herbicidal pyrrolesulfonylureas
US5214206A (en) * 1990-11-07 1993-05-25 Warner-Lambert Company Aminosulfonyl urea acat inhibitors
DE4039733A1 (en) 1990-12-13 1992-06-17 Basf Ag SUBSTITUTED 5-AMINOPYRAZOLE
CA2115990A1 (en) 1991-08-19 1993-03-04 George Albert Boswell Angiotensin ii receptor blocking imidazolinone derivatives
AU2496492A (en) 1991-08-19 1993-03-16 E.I. Du Pont De Nemours And Company Angiotensin ii receptor blocking imidazolinone derivatives
US5219856A (en) 1992-04-06 1993-06-15 E. I. Du Pont De Nemours And Company Angiotensin-II receptor blocking, heterocycle substituted imidazoles
JPH0645573A (en) 1992-07-23 1994-02-18 Nikon Corp Infrared ray solid-state image pick-up device
JPH06199053A (en) 1992-12-28 1994-07-19 Sankyo Kagaku Kk Color element for heat transfer recording
JPH06199054A (en) 1992-12-28 1994-07-19 Dainippon Printing Co Ltd Heat transfer sheet
JPH06199047A (en) 1993-01-08 1994-07-19 New Oji Paper Co Ltd Heat-sensitive recording substance
US5466823A (en) * 1993-11-30 1995-11-14 G.D. Searle & Co. Substituted pyrazolyl benzenesulfonamides
WO1997011057A1 (en) 1995-09-22 1997-03-27 E.I. Du Pont De Nemours And Company Arthropodicidal 1,4-dihydropyridines and 1,4-dihydropyrimidines
EP0795548B1 (en) 1995-09-28 2002-07-03 Suntory Limited Quinazoline derivatives and use thereof
EP0885890A1 (en) * 1996-02-26 1998-12-23 Sumitomo Pharmaceuticals Company, Limited Sulfonylureidopyrazole derivatives
US6022984A (en) 1998-07-27 2000-02-08 Pfizer Inc. Efficient synthesis of furan sulfonamide compounds useful in the synthesis of new IL-1 inhibitors
JP2000053649A (en) 1998-08-11 2000-02-22 Sumitomo Pharmaceut Co Ltd Sulfonylureidopyrazole derivative
EP0987552A3 (en) 1998-08-31 2000-06-07 Pfizer Products Inc. Diarylsulfonylurea binding proteins
TR200103337T2 (en) 1999-03-15 2002-03-21 Axys Pharmaceuticals, Inc. New bracelets and bracelets as prosthesis inhibitors
DK1257550T3 (en) 2000-02-04 2006-03-27 Portola Pharm Inc The platelet ADP receptor inhibitor
US6906063B2 (en) 2000-02-04 2005-06-14 Portola Pharmaceuticals, Inc. Platelet ADP receptor inhibitors
GB0017676D0 (en) 2000-07-19 2000-09-06 Angeletti P Ist Richerche Bio Inhibitors of viral polymerase
JP2004536063A (en) 2001-05-18 2004-12-02 ゾルファイ ファーマスーティカルズ ゲゼルシャフト ミット ベシュレンクテル ハフツング Use of combined compounds having NEP / MP-inhibitory activity in the manufacture of a medicament
AR037097A1 (en) 2001-10-05 2004-10-20 Novartis Ag ACILSULFONAMID COMPOUNDS, PHARMACEUTICAL COMPOSITIONS AND THE USE OF SUCH COMPOUNDS FOR THE PREPARATION OF A MEDICINAL PRODUCT
FI110677B (en) 2001-10-12 2003-03-14 Jujo Thermal Oy Heat-sensitive recording material for use in, e.g. stickers, has coating layer comprising chelate-type color forming system and leuco dye with urea-based developer
ATE355064T1 (en) 2001-10-26 2006-03-15 Angeletti P Ist Richerche Bio DIHYDROXYPYRIMIDINE CARBONIC ACID RAMIDE INHIBITORS OF HIV INTEGRASE
JP2005510542A (en) 2001-11-30 2005-04-21 ファイザー・プロダクツ・インク Combination of IL-1 / 18 inhibitor and TNF inhibitor for the treatment of inflammation.
MX249034B (en) 2002-05-28 2007-09-14 Dimensional Pharm Inc Novel thiophene amidines, compositions thereof, and methods of treating complement-mediated diseases and conditions.
AU2004279809B2 (en) 2003-10-03 2010-07-15 Portola Pharmaceuticals, Inc. Substituted isoquinolinones
ATE448222T1 (en) 2003-10-03 2009-11-15 Portola Pharm Inc 2,4-DIOXO-3-CHINAZOLINYLARYLSULFONYL UREAS
WO2005105777A1 (en) * 2004-05-05 2005-11-10 Pharmacia & Upjohn Company Llc Substituted thiophene amide compounds for the treatment of inflammation
CA2581638A1 (en) 2004-09-29 2006-04-13 Portola Pharmaceuticals, Inc. Substituted 2h-1,3-benzoxazin-4(3h)-ones
WO2006085815A1 (en) 2005-02-11 2006-08-17 Astrazeneca Ab Thiazole derivatives, their process for their preparation and their use in therapy
GB0505539D0 (en) 2005-03-17 2005-04-27 Novartis Ag Organic compounds
DK200600313A (en) 2006-03-03 2006-03-13 Novo Nordisk As Treating type 2 diabetes or metabolic syndrome with an interleukin 1beta inhibitor or an interleukin 1beta synthesis or release inhibitor
JPWO2007105637A1 (en) * 2006-03-10 2009-07-30 小野薬品工業株式会社 Nitrogen-containing heterocyclic derivatives and drugs containing them as active ingredients
TWI391378B (en) 2006-03-16 2013-04-01 Astellas Pharma Inc Quinolone derivative or pharmaceutically acceptable salt thereof
GB0701426D0 (en) 2007-01-25 2007-03-07 Univ Sheffield Compounds and their use
WO2009065096A1 (en) 2007-11-16 2009-05-22 University Of Medicine And Dentistry Of New Jersey Mechanism-based small-molecule parasite inhibitors
US8211928B2 (en) * 2009-05-29 2012-07-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
GB2474120B (en) 2009-10-01 2011-12-21 Amira Pharmaceuticals Inc Compounds as Lysophosphatidic acid receptor antagonists
ES2585287T3 (en) 2010-03-05 2016-10-04 Kyowa Hakko Kirin Co., Ltd. Pyrazolopyrimidine derivative
WO2012067265A1 (en) * 2010-11-18 2012-05-24 Kyoto University Method for screening drugs for suppressing inflammasome activity
WO2013031931A1 (en) 2011-09-02 2013-03-07 協和発酵キリン株式会社 Chemokine receptor activity regulator
JP6036193B2 (en) * 2012-11-09 2016-11-30 国立大学法人富山大学 Inflammasome activity regulator
PL221813B1 (en) 2013-02-22 2016-05-31 Univ Medyczny W Lublinie Derivatives of 1-aryl-6-benzenesulfonylimidazo[1,2-a][1.3.5]triazine and their preparation
TW201501713A (en) 2013-03-01 2015-01-16 Kyowa Hakko Kirin Co Ltd Agent for preventing and/or treating ocular inflammatory disease
CN104513239B (en) 2014-12-10 2017-08-22 沈阳药科大学 The ketone compounds of pyrazolo [3,4 c] pyridine 7 and its application
WO2016119349A1 (en) 2015-01-29 2016-08-04 中国农业大学 Preparation method for sulfonylurea and sulfonamide formate compounds
SI3578547T1 (en) 2015-02-16 2021-09-30 The University Of Queensland Sulfonylureas and related compounds and use of same
US10688077B2 (en) 2015-02-26 2020-06-23 H. Lee Moffitt Cancer Center And Research Institute, Inc. Inflammasome activation in myelodysplastic syndromes
AU2016379454B2 (en) 2015-12-23 2021-01-28 The University Of British Columbia Lipid-linked prodrugs
FR3046933B1 (en) 2016-01-25 2018-03-02 Galderma Research & Development NLRP3 INHIBITORS FOR THE TREATMENT OF INFLAMMATORY SKIN CONDITIONS
CN109071454B (en) 2016-02-16 2023-02-17 昆士兰大学 Sulfonylureas and related compounds and uses thereof
ES2940611T3 (en) 2016-04-18 2023-05-09 Novartis Ag Compounds and compositions for the treatment of conditions associated with NLRP activity
US10080743B2 (en) 2016-04-26 2018-09-25 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
WO2017189651A1 (en) 2016-04-26 2017-11-02 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as fxr agonists and methods of use thereof
US10080741B2 (en) 2016-04-26 2018-09-25 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use thereof
US10138228B2 (en) 2016-05-18 2018-11-27 Enanta Pharmaceuticals, Inc. Isoxazole derivatives as FXR agonists and methods of use therof
WO2017201150A1 (en) 2016-05-18 2017-11-23 Enanta Pharmaceuticals, Inc. Isoxazole analogs as fxr agonists and methods of use thereof
CA3059458A1 (en) 2017-05-24 2018-11-29 The University Of Queensland Novel compounds and uses
RS62910B1 (en) 2017-07-07 2022-03-31 Inflazome Ltd Novel sulfonamide carboxamide compounds
WO2019008029A1 (en) 2017-07-07 2019-01-10 Inflazome Limited Sulfonylureas and sulfonylthioureas as nlrp3 inhibitors
TW201910314A (en) 2017-08-15 2019-03-16 愛爾蘭商英弗雷佐姆有限公司 Novel compound
EP4246859A3 (en) 2018-09-27 2024-01-03 Apple Inc. Radio link monitoring and failure for new radio-unlicensed operation
GB201819083D0 (en) * 2018-11-23 2019-01-09 Inflazome Ltd Novel compounds

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4018929A (en) * 1974-04-17 1977-04-19 A. Christiaens Societe Anonyme 3-Loweralkylcarbamylsulfonamido-4-phenylaminopyridine-N-oxides, derivatives thereof and pharmaceutical compositions containing same
US5169860A (en) * 1992-03-13 1992-12-08 Eli Lilly And Company Antitumor compositions and methods of treatment
WO1998032733A1 (en) * 1997-01-29 1998-07-30 Pfizer Inc. Sulfonyl urea derivatives and their use in the control of interleukin-1 activity
WO2001019390A1 (en) * 1999-09-14 2001-03-22 Pfizer Products Inc. Combination treatment with il-1ra and diaryl sulphonyl urea compounds
EP1236468A1 (en) * 2001-02-12 2002-09-04 Warner-Lambert Company Sulfonylaminocarbonyl derivatives for the treatment of nuclear factor-kappa B mediated diseases and disorders

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
CROKER ET AL., IMMUNOL CELL BIOL, vol. 91, 2013, pages 625
DATABASE CAS 15 March 2004 (2004-03-15), "N-(2-chlorophenyl)-5-[[[[(2- chlorophenyl)amino] carbonyl] amino] sulfonyl]-1H-1,2,4-Triazole-1-carboxamide", XP055475046, retrieved from STN Database accession no. 663215-37-0 *
DATABASE CAS 19 December 2000 (2000-12-19), "N-[[(4- chlorophenyl)amino]carbonyl]-1-(phenylmethyl)-1H-1,2,4-Triazole-3-sulfonamide", XP055475050, retrieved from STN Database accession no. 309742-96-9 *
FOROUMADI, A ET AL.: "Synthesis of certain diarylsulfonylurea derivatives as new potential antitumor agents", CHEMISTRY: AN INDIAN JOURNAL, vol. 1, no. 12, 2005, pages 745 - 748, XP009505510 *
GODA, F ET AL.: "Development of some 1,2,4-triazole derivatives as potential hypoglycemic agents", ALEXANDRIA JOURNAL OF PHARMACEUTICAL SCIENCES, vol. 1, no. 2, 1987, pages 63 - 66, XP009505505 *
MOHAMADI, F. ET AL.: "Sulfonylureas: A New Class of Cancer Chemotherapeutic Agents", JOURNAL OF MEDICINAL CHEMISTRY, vol. 35, 1992, pages 3012 - 3016, XP000919054 *
MONTE, J.MED.CHEM., vol. 39, 1996, pages 2953 - 2961
PERREGAUX ET AL., J. PHARMACOL. EXP. THER., vol. 299, 2001, pages 187 - 197
YOUSSEF, K ET AL.: "Synthesis of certain diarylsulfonylureas as antitumor agents", MEDICINAL CHEMISTRY RESEARCH, vol. 10, no. 6, 2001, pages 404 - 418, XP008020857 *

Cited By (199)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3259253B1 (en) 2015-02-16 2020-01-15 The University of Queensland Sulfonylureas and related compounds and use of same
US11130731B2 (en) 2015-02-16 2021-09-28 The Provost, Fellows, Foundation Scholars, And The Other Members Of Board, Of The College Of The Holy And Undivided Trinity Of Queen Elizabeth Near Dublin Sulfonylureas and related compounds and use of same
US11858922B2 (en) 2016-02-16 2024-01-02 The University Of Queensland Sulfonylureas and related compounds and use of same
WO2017140778A1 (en) 2016-02-16 2017-08-24 The University Of Queensland Sulfonylureas and related compounds and use of same
EP3872070A1 (en) 2016-04-18 2021-09-01 Novartis AG Compounds and compositions for treating conditions associated with nlrp activity
AU2017254522B2 (en) * 2016-04-18 2021-09-23 Novartis Ag Compounds and compositions for treating conditions associated with NLRP activity
AU2017254523B2 (en) * 2016-04-18 2021-09-02 Novartis Ag Compounds and compositions for treating conditions associated with NLRP activity
WO2017184624A1 (en) 2016-04-18 2017-10-26 Ifm Therapeutics, Inc Compounds and compositions for treating conditions associated with nlrp activity
WO2017184623A1 (en) 2016-04-18 2017-10-26 Ifm Therapeutics, Inc Compounds and compositions for treating conditions associated with nlrp activity
US11339136B2 (en) 2016-04-18 2022-05-24 Novartis Ag Compounds and compositions for treating conditions associated with NLRP activity
US11597706B2 (en) 2016-04-18 2023-03-07 Novartis Ag Compounds and compositions for treating conditions associated with NLRP activity
US11760735B2 (en) 2016-04-18 2023-09-19 Novartis Ag Compounds and compositions for treating conditions associated with NLRP activity
WO2018103583A1 (en) * 2016-12-05 2018-06-14 Zibo Anxuan Pharmaceutical Science And Technology Co., Ltd. N-hydroxy-benzene-sulfonamide derivatives and their uses thereof
US11040985B2 (en) 2017-01-23 2021-06-22 Genentech, Inc. Chemical compounds as inhibitors of interleukin-1 activity
AU2018210525B2 (en) * 2017-01-23 2022-06-02 Genentech, Inc. Chemical compounds as inhibitors of interleukin-1 activity
IL267961B2 (en) * 2017-01-23 2023-07-01 Genentech Inc Chemical compounds as inhibitors of interleukin-1 activity
US11702428B2 (en) 2017-01-23 2023-07-18 Genentech, Inc. Chemical compounds as inhibitors of interleukin-1 activity
IL267961B1 (en) * 2017-01-23 2023-03-01 Genentech Inc Chemical compounds as inhibitors of interleukin-1 activity
RU2792143C2 (en) * 2017-01-23 2023-03-17 Дженентек, Инк. Chemical compounds as interleukin-1 inhibitors
JP7163293B2 (en) 2017-01-23 2022-10-31 ジェネンテック, インコーポレイテッド Chemical compounds as inhibitors of interleukin-1 activity
WO2018136890A1 (en) * 2017-01-23 2018-07-26 Jecure Therapeutics, Inc. Chemical compounds as inhibitors of interleukin-1 activity
JP2020505381A (en) * 2017-01-23 2020-02-20 ジェネンテック, インコーポレイテッド Chemical compounds as inhibitors of interleukin-1 activity
CN110366549A (en) * 2017-01-23 2019-10-22 基因泰克公司 Compound as interleukin-1 activity inhibitor
EP3851434A1 (en) * 2017-01-23 2021-07-21 Genentech, Inc. Chemical compounds as inhibitors of interleukin-1 activity
KR102581620B1 (en) 2017-03-13 2023-09-22 노드테라 리미티드 compound
CN110869352A (en) * 2017-03-13 2020-03-06 诺瑟拉有限公司 Chemical compound
US11345669B2 (en) 2017-03-13 2022-05-31 NodThera Limited Urea derivatives and methods of use thereof
WO2018167468A1 (en) * 2017-03-13 2018-09-20 NodThera Limited Chemical compounds
CN110869352B (en) * 2017-03-13 2023-11-28 诺瑟拉有限公司 Chemical compound
IL269204A (en) * 2017-03-13 2019-11-28 Nodthera Ltd Chemical compounds
KR20200010185A (en) * 2017-03-13 2020-01-30 노드테라 리미티드 compound
WO2018215818A1 (en) * 2017-05-24 2018-11-29 The University Of Queensland Novel compounds and uses
US11840543B2 (en) 2017-05-24 2023-12-12 The University Of Queensland Compounds and uses
JP2020520984A (en) * 2017-05-24 2020-07-16 ザ ユニバーシティ オブ クィーンズランド Novel compounds and uses
JP7072586B2 (en) 2017-05-24 2022-05-20 ザ ユニバーシティ オブ クィーンズランド New compounds and uses
US11236045B2 (en) 2017-06-09 2022-02-01 Cadila Healthcare Limited Substituted sulfoximine compounds
WO2018225018A1 (en) 2017-06-09 2018-12-13 Cadila Healthcare Limited Novel substituted sulfoximine compounds
JP2020526589A (en) * 2017-07-07 2020-08-31 インフレイゾーム リミテッド New sulfonamide carboxamide compounds
WO2019008029A1 (en) * 2017-07-07 2019-01-10 Inflazome Limited Sulfonylureas and sulfonylthioureas as nlrp3 inhibitors
WO2019008025A1 (en) * 2017-07-07 2019-01-10 Inflazome Limited Novel sulfonamide carboxamide compounds
US11465992B2 (en) 2017-07-07 2022-10-11 Inflazome Limited Sulfonamide carboxamide compounds
EP3978489A1 (en) 2017-07-07 2022-04-06 Inflazome Limited Novel sulfonamide carboxamide compounds
JP7235742B2 (en) 2017-07-07 2023-03-08 インフレイゾーム リミテッド Novel sulfonamide carboxamide compound
AU2018297606B2 (en) * 2017-07-07 2022-08-11 Inflazome Limited Novel sulfonamide carboxamide compounds
US11370776B2 (en) 2017-07-07 2022-06-28 Inflazome Limited Sulfonylureas and sulfonylthioureas as NLRP3 inhibitors
CN110914256A (en) * 2017-07-07 2020-03-24 英夫拉索姆有限公司 Novel sulfonamide carboxamide compounds
RU2808572C2 (en) * 2017-07-07 2023-11-29 Инфлазоум Лимитед New sulphonamide carboxamide compounds
TWI789401B (en) * 2017-07-07 2023-01-11 愛爾蘭商英弗雷佐姆有限公司 Novel compounds
JP7159282B2 (en) 2017-07-14 2022-10-24 イネイト・テューマー・イミュニティ・インコーポレイテッド NLRP3 modulator
JP2020527139A (en) * 2017-07-14 2020-09-03 イネイト・テューマー・イミュニティ・インコーポレイテッドInnate Tumor Immunity, Inc. NLRP3 modulator
CN111094242A (en) * 2017-07-24 2020-05-01 诺华炎症研究公司 Compounds and compositions for treating conditions associated with NLRP activity
KR102565167B1 (en) 2017-07-24 2023-08-11 노파르티스 아게 Compounds and compositions for treating conditions associated with NLRP activity
WO2019023147A1 (en) * 2017-07-24 2019-01-31 IFM Tre, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2019023145A1 (en) * 2017-07-24 2019-01-31 IFM Tre, Inc. Compounds and compositions for treating conditions associated with nlrp activity
JP2020528428A (en) * 2017-07-24 2020-09-24 ノバルティス インフラマソーム リサーチ,インコーポレイテッド Compounds and compositions for treating conditions associated with NLRP activity
US10654816B2 (en) 2017-07-24 2020-05-19 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with NLRP activity
CN111094242B (en) * 2017-07-24 2024-02-09 诺华股份有限公司 Compounds and compositions for treating disorders associated with NLRP activity
AU2018307743C1 (en) * 2017-07-24 2021-09-09 Novartis Ag Compounds and compositions for treating conditions associated with NLRP activity
JP7412328B2 (en) 2017-07-24 2024-01-12 ノバルティス アーゲー Compounds and compositions for treating conditions associated with NLRP activity
RU2786719C2 (en) * 2017-07-24 2022-12-26 Новартис Аг Compounds and compositions for the treatment of conditions associated with nlrp activity
KR20200032154A (en) * 2017-07-24 2020-03-25 노바티스 인플라마솜 리서치, 인크. Compounds and compositions for treating conditions associated with NLRP activity
JP2020528889A (en) * 2017-07-24 2020-10-01 ノバルティス インフラマソーム リサーチ,インコーポレイテッド Compounds and compositions for treating conditions associated with NLRP activity
AU2018307743B2 (en) * 2017-07-24 2021-03-25 Novartis Ag Compounds and compositions for treating conditions associated with NLRP activity
CN111094243A (en) * 2017-07-24 2020-05-01 诺华炎症研究公司 Compounds and compositions for treating conditions associated with NLRP activity
US11203579B2 (en) 2017-07-24 2021-12-21 Novartis Ag Compounds and compositions for treating conditions associated with NLRP activity
US11370763B2 (en) 2017-07-24 2022-06-28 Novartis Ag Compounds and compositions for treating conditions associated with NLRP activity
US11724992B2 (en) 2017-07-24 2023-08-15 Novartis Ag Compounds and compositions for treating conditions associated with NLRP activity
CN111094243B (en) * 2017-07-24 2023-09-05 诺华股份有限公司 Compounds and compositions for treating disorders associated with NLRP activity
JP7349981B2 (en) 2017-07-24 2023-09-25 ノバルティス アーゲー Compounds and compositions for treating conditions associated with NLRP activity
TWI810198B (en) * 2017-07-31 2023-08-01 英商諾瑟拉有限公司 Selective inhibitors of nlrp3 inflammasome
WO2019025467A1 (en) * 2017-07-31 2019-02-07 NodThera Limited Selective inhibitors of nlrp3 inflammasome
WO2020035465A1 (en) 2017-08-15 2020-02-20 Inflazome Limited Novel sulfoneurea compounds
WO2019034686A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Novel sulfonamide carboxamide compounds
CN111132974B (en) * 2017-08-15 2023-11-21 英夫拉索姆有限公司 Sulfonamide carboxamide compounds
JP2020531453A (en) * 2017-08-15 2020-11-05 インフレイゾーム リミテッド Sulfonylurea and Sulfonylurea as NLRP3 Inhibitors
JP2020531435A (en) * 2017-08-15 2020-11-05 インフレイゾーム リミテッド New sulfonamide carboxamide compounds
JP2020531448A (en) * 2017-08-15 2020-11-05 インフレイゾーム リミテッド New sulfonamide carboxamide compounds
US11542255B2 (en) 2017-08-15 2023-01-03 Inflazome Limited Sulfonylureas and sulfonylthioureas as NLRP3 inhibitors
CN111093773A (en) * 2017-08-15 2020-05-01 英夫拉索姆有限公司 Sulfonylureas and sulfonylthioureas as NLRP3 inhibitors
US11773058B2 (en) 2017-08-15 2023-10-03 Inflazome Limited Sulfonamide carboxamide compounds
CN111107903A (en) * 2017-08-15 2020-05-05 英夫拉索姆有限公司 Novel sulfonamide carboxamide compounds
WO2019034696A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Novel sulfonamide carboxamide compounds
WO2019034693A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Sulfonylureas and sulfonylthioureas as nlrp3 inhibitors
CN111132974A (en) * 2017-08-15 2020-05-08 英夫拉索姆有限公司 Novel sulfonamide carboxamide compounds
WO2019034697A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Novel sulfonamide carboxamide compounds
WO2019034690A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Sulfonylureas and sulfonylthioureas as nlrp3 inhibitors
US11613542B2 (en) 2017-08-15 2023-03-28 Inflazome Limited Sulfonylureas and sulfonylthioureas as NLRP3 inhibitors
WO2019034692A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Sulfonylureas and sulfonylthioureas as nlrp3 inhibitors
US11926600B2 (en) 2017-08-15 2024-03-12 Inflazome Limited Sulfonylureas and sulfonylthioureas as NLRP3 inhibitors
WO2019034688A1 (en) * 2017-08-15 2019-02-21 Inflazome Limited Novel sulfonamide carboxamide compounds
CN112789264A (en) * 2017-08-15 2021-05-11 英夫拉索姆有限公司 Novel sulfonylurea compound
US11518739B2 (en) 2017-08-15 2022-12-06 Inflazome Limited Sulfonamide carboxamide compounds
WO2019043610A1 (en) 2017-08-31 2019-03-07 Cadila Healthcare Limited Novel substituted sulfonylurea derivatives
WO2019068772A1 (en) * 2017-10-03 2019-04-11 Inflazome Limited Novel compounds
US11623922B2 (en) 2017-10-03 2023-04-11 Inflazome Limited Compounds
CN111315733A (en) * 2017-11-09 2020-06-19 英夫拉索姆有限公司 Novel sulfonamide carboxamide compounds
WO2019092170A1 (en) * 2017-11-09 2019-05-16 Inflazome Limited Novel sulfonamide carboxamide compounds
WO2019092172A1 (en) * 2017-11-09 2019-05-16 Inflazome Limited Novel sulfonamide carboxamide compounds
JP2021502364A (en) * 2017-11-09 2021-01-28 インフレイゾーム リミテッド New sulfonamide carboxamide compounds
WO2019092171A1 (en) * 2017-11-09 2019-05-16 Inflazome Limited Novel sulfonamide carboxamide compounds
JP7339959B2 (en) 2017-12-18 2023-09-06 ノッドセラ リミテッド Sulfonylurea Derivatives as NLRP3 Inflammasome Modulators
JP2021506966A (en) * 2017-12-18 2021-02-22 ノッドセラ リミテッド Sulfonyl urea derivative as NLRP3 inflammasome regulator
US11834433B2 (en) 2018-03-02 2023-12-05 Inflazome Limited Compounds
WO2019166619A1 (en) * 2018-03-02 2019-09-06 Inflazome Limited Novel compounds
WO2019166633A1 (en) 2018-03-02 2019-09-06 Inflazome Limited Sulfonamide derivates as nlrp3 inhibitors
US11905252B2 (en) 2018-03-02 2024-02-20 Inflazome Limited Compounds
US11884645B2 (en) 2018-03-02 2024-01-30 Inflazome Limited Sulfonyl acetamides as NLRP3 inhibitors
US11530200B2 (en) 2018-03-02 2022-12-20 Inflazome Limited Compounds
US20210147349A1 (en) * 2018-03-02 2021-05-20 Inflazome Limited Phenylsulfonylurea derivatives useful as nlrp3 inhibitors
WO2019166627A1 (en) 2018-03-02 2019-09-06 Inflazome Limited Novel compounds
US11857529B2 (en) 2018-03-21 2024-01-02 Olatec Therapeutics, Inc. Methods for treating melanoma
WO2019182981A1 (en) * 2018-03-21 2019-09-26 Olatec Therapeutics Llc Methods for treating melanoma
EP3722281A1 (en) 2018-04-23 2020-10-14 Inflazome Limited A sodium salt of n-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1-isopropyl-1h-pyrazole-3-sulfonamide
CN112020495A (en) * 2018-04-23 2020-12-01 英夫拉索姆有限公司 N- ((1,2,3,5,6, 7-hexahydro-sym-indacen-4-yl) carbamoyl) -1-isopropyl-1H-pyrazole-3-sulfonamide sodium salt
KR20200135562A (en) * 2018-04-23 2020-12-02 코어셉트 쎄라퓨틱스 인코포레이티드 Method for preparing regioselective N-alkyl triazole
US10973803B2 (en) 2018-04-23 2021-04-13 Inflazome Limited Sodium salt of N-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1-isopropyl-1H-pyrazole-3-sulfonamide
WO2019206871A1 (en) 2018-04-23 2019-10-31 Inflazome Limited A sodium salt of n-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 -isopropyl-1 h-pyrazole-3-sulfonamide
WO2019209693A1 (en) 2018-04-23 2019-10-31 Corcept Therapeutics, Inc. Methods of preparing regioselective n-alkyl triazoles
EP3784658A4 (en) * 2018-04-23 2021-12-22 Corcept Therapeutics, Inc. Methods of preparing regioselective n-alkyl triazoles
IL278125B (en) * 2018-04-23 2022-09-01 Corcept Therapeutics Inc Methods of preparing regioselective n-alkyl triazoles
WO2019211463A1 (en) 2018-05-04 2019-11-07 Inflazome Limited Novel compounds
WO2020010140A1 (en) * 2018-07-03 2020-01-09 Novartis Inflammasome Research, Inc. Nlrp modulators
WO2020010118A1 (en) * 2018-07-03 2020-01-09 Novartis Inflammasome Research, Inc. Methods of treating or selecting a treatment for a subject resistant to tnf inhibitor using a nlrp3 antagonist
WO2020010143A1 (en) 2018-07-03 2020-01-09 Novartis Inflammasome Research, Inc. Nlrp modulators
US20210261568A1 (en) * 2018-07-20 2021-08-26 Genentech, Inc. Sulfonylurea compounds as inhibitors of interleukin-1 activity
WO2020018970A1 (en) * 2018-07-20 2020-01-23 Genentech, Inc. Sulfonylurea compounds as inhibitors of interleukin-1 activity
WO2020018975A1 (en) * 2018-07-20 2020-01-23 Genentech, Inc. Sulfonimidamide compounds as inhibitors of interleukin-1 activity
TWI825134B (en) * 2018-07-20 2023-12-11 美商建南德克公司 Sulfonimidamide compounds as inhibitors of interleukin-1 activity
US11560391B2 (en) 2018-07-20 2023-01-24 Genentech, Inc. Sulfonylurea compounds as inhibitors of interleukin-1 activity
WO2020035466A1 (en) 2018-08-15 2020-02-20 Inflazome Limited Novel sulfoneurea compounds
WO2020035464A1 (en) 2018-08-15 2020-02-20 Inflazome Limited Novel sulfonamideurea compounds
CN112839927A (en) * 2018-10-19 2021-05-25 英夫拉索姆有限公司 Novel process
WO2020079207A1 (en) * 2018-10-19 2020-04-23 Inflazome Limited Novel processes
US11724988B2 (en) 2018-10-19 2023-08-15 Inflazome Limited Processes
WO2020086732A1 (en) 2018-10-24 2020-04-30 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
CN113056451A (en) * 2018-10-24 2021-06-29 诺华股份有限公司 Compounds and compositions for treating conditions associated with NLRP activity
CN113166065A (en) * 2018-11-13 2021-07-23 诺华股份有限公司 Compounds and compositions for treating conditions associated with NLRP activity
WO2020102096A1 (en) * 2018-11-13 2020-05-22 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2020102098A1 (en) * 2018-11-13 2020-05-22 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2020102100A1 (en) * 2018-11-13 2020-05-22 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
RU2795108C2 (en) * 2018-11-13 2023-04-28 Новартис Аг Compounds and compositions for the treatment of conditions associated with nlrp activity
AU2019379109B2 (en) * 2018-11-13 2022-07-07 Novartis Ag Compounds and compositions for treating conditions associated with NLRP activity
CN113166081A (en) * 2018-11-13 2021-07-23 诺华股份有限公司 Compounds and compositions for treating conditions associated with NLRP activity
WO2020102574A1 (en) * 2018-11-16 2020-05-22 Novartis Inflammasome Research, Inc. The compounds and compositions for treating conditions associated with nlrp activity
CN113227055A (en) * 2018-11-16 2021-08-06 诺华股份有限公司 Compounds and compositions for treating conditions associated with NLRP activity
WO2020102576A1 (en) * 2018-11-16 2020-05-22 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
CN113164763A (en) * 2018-11-23 2021-07-23 英夫拉索姆有限公司 NLRP3 inhibitor
WO2020104657A1 (en) 2018-11-23 2020-05-28 Inflazome Limited Nlrp3 inhibitors
EP3911631A4 (en) * 2019-01-14 2022-09-28 Cadila Healthcare Limited Novel substituted sulfonylurea derivatives
WO2020148619A1 (en) 2019-01-14 2020-07-23 Cadila Healthcare Limited Novel substituted sulfonylurea derivatives
WO2020154321A1 (en) * 2019-01-22 2020-07-30 Novartis Inflammasome Research, Inc. Compounds and compositions for treating conditions associated with nlrp activity
WO2020152361A1 (en) * 2019-01-25 2020-07-30 NodThera Limited Carbamate derivatives and uses thereof
CN113660983A (en) * 2019-01-25 2021-11-16 诺瑟拉有限公司 Carbamate derivatives and uses thereof
WO2020178441A1 (en) * 2019-03-06 2020-09-10 INSERM (Institut National de la Santé et de la Recherche Médicale) Inhibitors of ngal protein
WO2020208249A1 (en) 2019-04-12 2020-10-15 Inflazome Limited Nlrp3 inflammasome inhibition
CN114761383A (en) * 2019-06-12 2022-07-15 诺瑟拉有限公司 Sulfonamide derivatives and uses thereof
WO2020254697A1 (en) 2019-06-21 2020-12-24 Ac Immune Sa Fused 1,2 thiazoles and 1,2 thiazines which act as nl3p3 modulators
WO2021002887A1 (en) 2019-07-02 2021-01-07 Novartis Inflammasome Research, Inc. Gut-targeted nlrp3 antagonists and their use in therapy
WO2021009567A1 (en) * 2019-07-17 2021-01-21 Zomagen Biosciences Ltd Nlrp3 modulators
CN114555571A (en) * 2019-07-17 2022-05-27 祖玛珍生物科学有限公司 N- ((1,2,3,5,6, 7-hexahydro-s-indacen-4-yl) carbamoyl) -4,5,6, 7-tetrahydrobenzofuran-2-sulfonamide derivatives and related compounds as NLPR3 modulators for the treatment of Multiple Sclerosis (MS)
WO2021009566A1 (en) * 2019-07-17 2021-01-21 Zomagen Biosciences Ltd N-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-4,5,6,7-tetrahydrobenzofuran -2-sulfonamide derivatives and related compounds as nlpr3 modulators for the treatment of multiple sclerosis (ms)
WO2021032591A1 (en) 2019-08-16 2021-02-25 Inflazome Limited Macrocyclic sulfonylurea derivatives useful as nlrp3 inhibitors
WO2021043966A1 (en) 2019-09-06 2021-03-11 Inflazome Limited Nlrp3 inhibitors
WO2021048809A1 (en) * 2019-09-12 2021-03-18 Cadila Healthcare Limited Novel substituted sulfoximine derivatives
WO2021089769A1 (en) * 2019-11-07 2021-05-14 Inflazome Limited Treatment and prevention of neuroinflammation or an inflammatory brain disorder
WO2021089781A1 (en) * 2019-11-07 2021-05-14 Inflazome Limited Treatment or prevention of psychiatric brain disorders using the nlrp3 inhibitor n-((1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl)-1 -isopropyl-1 h-pyrazole-3-sulfonamide
WO2021089768A2 (en) 2019-11-07 2021-05-14 Inflazome Limited Novel treatment
CN114599357A (en) * 2019-11-07 2022-06-07 英夫拉索姆有限公司 Treatment of arthritis
WO2021089782A1 (en) * 2019-11-07 2021-05-14 Inflazome Limited Treatment of autoinflammatory disorders
CN114599356A (en) * 2019-11-07 2022-06-07 英夫拉索姆有限公司 Treatment of autoinflammatory disorders
WO2021089776A1 (en) * 2019-11-07 2021-05-14 Inflazome Limited Treatment and prevention of a traumatic brain disorder
CN114641287A (en) * 2019-11-07 2022-06-17 英夫拉索姆有限公司 Treatment and prevention of neurodegenerative disorders
WO2021089768A3 (en) * 2019-11-07 2021-06-24 Inflazome Limited Treatment and prevention of a neurodegenerative disorder
WO2021089783A1 (en) * 2019-11-07 2021-05-14 Inflazome Limited Treatment of arthritis
CN114630664A (en) * 2019-11-07 2022-06-14 英夫拉索姆有限公司 Treatment and prevention of neuroinflammation or inflammatory brain disorders
WO2021111351A1 (en) * 2019-12-03 2021-06-10 Cadila Healthcare Limited Novel substituted sulfonylurea and sulfoximineurea derivatives
WO2021150574A1 (en) * 2020-01-22 2021-07-29 Genentech, Inc. Sulfonimidamide compounds as nlrp3 modulators
WO2021165245A1 (en) 2020-02-18 2021-08-26 Inflazome Limited Compounds
US11603375B2 (en) 2020-03-16 2023-03-14 Zomagen Biosciences Ltd NLRP3 modulators
WO2021185912A1 (en) 2020-03-19 2021-09-23 Softhale Nv Method for the treatment nlrp3-associated diseases
WO2021249337A1 (en) 2020-06-11 2021-12-16 南京明德新药研发有限公司 Dimethylsulfoximine derivative
WO2021255279A1 (en) 2020-06-19 2021-12-23 Ac Immune Sa D i h yd rooxazo le and thiourea derivatives modulating the nlrp3 inflammasome pathway
WO2022023907A1 (en) 2020-07-31 2022-02-03 Novartis Ag Methods of selecting and treating patients at elevated risk of major adverse cardiac events
WO2022115417A1 (en) * 2020-11-25 2022-06-02 VenatoRx Pharmaceuticals, Inc. Sulfonyl urea nlrp3 inflammasome inhibitors
WO2022171185A1 (en) * 2021-02-10 2022-08-18 杭州英创医药科技有限公司 Compound serving as nlrp3 inhibitor
WO2022234447A1 (en) * 2021-05-03 2022-11-10 Universita' Degli Studi Di Torino Nlrp3 inflammasome-inhibiting compounds and the use thereof
IT202100011237A1 (en) 2021-05-03 2022-11-03 Univ Degli Studi Di Torino NLRP3 INFLAMMASOME INHIBITOR COMPOUNDS AND THEIR USE
WO2022269010A1 (en) 2021-06-23 2022-12-29 F. Hoffmann-La Roche Ag A crystalline potassium salt of 1-ethyl- n -((1,2,3,5,6,7-hexahydro- s -indacen-4-yl)carbamoyl)piperidine-4 -sulfonamide
WO2023037024A1 (en) * 2021-09-08 2023-03-16 Fundacion Para La Investigacion Biomedica Del Hospital Universitario De La Princesa (90%) N-sulfonylurea derivatives and the therapeutic use thereof
ES2948511A1 (en) * 2021-09-08 2023-09-13 Fundacion Para La Investigacion Biomedica Del Hospital Univ De La Princesa N-SULFONYLUREA DERIVATIVES AND THEIR THERAPEUTIC USE (Machine-translation by Google Translate, not legally binding)
KR20230066899A (en) 2021-11-08 2023-05-16 제일약품주식회사 Novel compounds as nlrp3 inhibitor and pharmaceutical composition comprising the same
WO2023118521A1 (en) 2021-12-22 2023-06-29 Ac Immune Sa Dihydro-oxazol derivative compounds
US11618751B1 (en) 2022-03-25 2023-04-04 Ventus Therapeutics U.S., Inc. Pyrido-[3,4-d]pyridazine amine derivatives useful as NLRP3 derivatives
WO2023230002A1 (en) * 2022-05-23 2023-11-30 VenatoRx Pharmaceuticals, Inc. Nlrp3 inflammasome inhibitors
WO2024013395A1 (en) 2022-07-14 2024-01-18 Ac Immune Sa Pyrrolotriazine and imidazotriazine derivatives as modulators of the nlrp3 inflammasome pathway
WO2024023266A1 (en) 2022-07-28 2024-02-01 Ac Immune Sa Novel compounds
KR20240022938A (en) 2022-08-12 2024-02-20 제일약품주식회사 Novel compounds as nlrp3 inhibitor and pharmaceutical composition comprising the same
CN115772173A (en) * 2022-12-20 2023-03-10 武汉国粹医药科技有限公司 Benzofuran compound, preparation method and application thereof, and antibacterial agent
CN115772173B (en) * 2022-12-20 2024-04-16 武汉国粹医药科技有限公司 Benzofuran compound, preparation method and application thereof, and antibacterial agent

Also Published As

Publication number Publication date
HRP20200214T1 (en) 2020-08-07
RS62164B1 (en) 2021-08-31
PE20221627A1 (en) 2022-10-19
CY1122832T1 (en) 2021-05-05
JP2018510207A (en) 2018-04-12
EP3259253B1 (en) 2020-01-15
CL2019000060A1 (en) 2019-05-03
SG11201706664QA (en) 2017-09-28
AU2016222278A1 (en) 2017-08-10
HK1249501A1 (en) 2018-11-02
IL253661A0 (en) 2017-09-28
RU2017128287A (en) 2019-03-18
LT3578547T (en) 2021-08-25
RU2739356C2 (en) 2020-12-23
MX2017010528A (en) 2018-11-12
JP6929792B2 (en) 2021-09-01
JP2021185159A (en) 2021-12-09
US11130731B2 (en) 2021-09-28
PL3578547T3 (en) 2021-12-20
RS60048B1 (en) 2020-04-30
HUE055755T2 (en) 2021-12-28
EP3578547B1 (en) 2021-05-26
NZ733948A (en) 2022-02-25
LT3259253T (en) 2020-04-27
NZ772349A (en) 2024-02-23
ME03737B (en) 2021-01-20
DK3259253T3 (en) 2020-04-14
EP3259253A4 (en) 2018-07-25
CN107428696B (en) 2021-08-20
MA41553A (en) 2017-12-27
PT3259253T (en) 2020-03-11
PE20180160A1 (en) 2018-01-18
AU2020203464A1 (en) 2020-06-18
EP3259253A1 (en) 2017-12-27
PL3259253T3 (en) 2020-09-07
AU2021258033A1 (en) 2021-11-25
HRP20211225T1 (en) 2021-11-12
CL2017002097A1 (en) 2018-04-27
ES2881228T3 (en) 2021-11-29
CN113563264A (en) 2021-10-29
WO2016131098A8 (en) 2017-03-16
CN113582889A (en) 2021-11-02
RU2017128287A3 (en) 2019-08-01
ES2777626T3 (en) 2020-08-05
IL253661B1 (en) 2023-09-01
IL273065B1 (en) 2023-09-01
US20190359564A1 (en) 2019-11-28
SG10202002599XA (en) 2020-04-29
MA47440B1 (en) 2021-08-31
AU2020203464B2 (en) 2021-08-05
MX2023001647A (en) 2023-03-16
CN107428696A (en) 2017-12-01
KR20170109678A (en) 2017-09-29
MD3259253T2 (en) 2020-06-30
AU2016222278B2 (en) 2020-07-09
US20220112159A1 (en) 2022-04-14
EP3888749A1 (en) 2021-10-06
MA41553B1 (en) 2020-04-30
EP3578547A1 (en) 2019-12-11
MY193765A (en) 2022-10-27
DK3578547T3 (en) 2021-08-23
IL253661B2 (en) 2024-01-01
US20180044287A1 (en) 2018-02-15
PT3578547T (en) 2021-06-22
IL273065B2 (en) 2024-01-01
SI3259253T1 (en) 2020-07-31
MY197094A (en) 2023-05-24
IL273065A (en) 2020-04-30
BR112017017610A2 (en) 2018-05-08
SI3578547T1 (en) 2021-09-30
US10538487B2 (en) 2020-01-21
CA2975192A1 (en) 2016-08-25
MA47440A (en) 2019-12-11
MA56473A (en) 2022-05-11

Similar Documents

Publication Publication Date Title
AU2016222278B2 (en) Sulfonylureas and related compounds and use of same
CN109071454B (en) Sulfonylureas and related compounds and uses thereof
JP7291226B2 (en) imidazo[1,2-B]pyridazine IL-17A inhibitors
TWI637951B (en) Heterocyclic amides as kinase inhibitors
JP6289485B2 (en) Heteroaryl inhibitors of PDE4
JP6661749B2 (en) New compounds as ROR gamma modulators
JP5220858B2 (en) Novel piperazine amide derivatives
TW201315727A (en) Uracil derivatives and their medical use
US20180092866A1 (en) Substituted n-([1,1&#39;-biphenyl]-3-yl)-[1,1&#39;-biphenyl]-3-carboxamide analogs as inhibitors for beta-catenin/b-cell lymphoma 9 interactions
EP3700891A1 (en) Aromatic sulfonamide derivatives for the treatment of ischemic stroke
RU2795512C2 (en) Sulphonylureas and related compounds and their use
WO2024033845A1 (en) Heterocyclic compound

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: 16751821

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 253661

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2975192

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2016222278

Country of ref document: AU

Date of ref document: 20160216

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 001395-2017

Country of ref document: PE

ENP Entry into the national phase

Ref document number: 2017560843

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 11201706664Q

Country of ref document: SG

Ref document number: MX/A/2017/010528

Country of ref document: MX

Ref document number: 15551264

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 122021001390

Country of ref document: BR

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112017017610

Country of ref document: BR

REEP Request for entry into the european phase

Ref document number: 2016751821

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20177025608

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2017128287

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112017017610

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20170816