WO2017045750A1 - Compounds as asic inhibitors and uses thereof - Google Patents

Compounds as asic inhibitors and uses thereof Download PDF

Info

Publication number
WO2017045750A1
WO2017045750A1 PCT/EP2016/001530 EP2016001530W WO2017045750A1 WO 2017045750 A1 WO2017045750 A1 WO 2017045750A1 EP 2016001530 W EP2016001530 W EP 2016001530W WO 2017045750 A1 WO2017045750 A1 WO 2017045750A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
piperazin
nrc
thieno
ring
Prior art date
Application number
PCT/EP2016/001530
Other languages
French (fr)
Inventor
Amanda Sutton
Daryl Walter
Steve EAST
Yolanda PEREZ
Original Assignee
Merck Patent Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Merck Patent Gmbh filed Critical Merck Patent Gmbh
Publication of WO2017045750A1 publication Critical patent/WO2017045750A1/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D279/00Heterocyclic compounds containing six-membered rings having one nitrogen atom and one sulfur atom as the only ring hetero atoms
    • C07D279/041,3-Thiazines; Hydrogenated 1,3-thiazines
    • C07D279/081,3-Thiazines; Hydrogenated 1,3-thiazines condensed with carbocyclic rings or ring systems
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/02Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains two hetero rings
    • C07D491/04Ortho-condensed systems

Definitions

  • the present invention relates to compounds that are useful as inhibitors of acid- sensing ion channels (ASIC).
  • ASIC acid- sensing ion channels
  • the invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using said compositions in the treatment of various disorders.
  • ASIC channels are proton-gated, voltage-insensitive cation channels activated by extracellular acidosis.
  • ASICs belong to a superfamily which also contains degenerins (DEG) and epithelial sodium channels (ENaC) All members within the ENaC/DEG/ASIC superfamily share the same topology, with the N- and C-termini located inside the cell and a large cysteine-rich extracellular domain.
  • DEG degenerins
  • ENaC epithelial sodium channels
  • four genes encode at least six different ASIC subunits, ASICla, ASIC2a, ASIC2b, ASIC3 and ASIC4.
  • ASICs function as trimers and conduct mostly Na+.
  • the amino acid sequences of ASIC subunits are well conserved between species. For example, the mouse ASICla and the human ASICla share over 99% of their amino acid sequence identity.
  • ASICs are highly expressed in neurons, but are also present in non-neuronal tissues. ASICla and ASIC2a and b are widely expressed in the central nervous system, while almost all subunits are present in sensory neurons of the peripheral nervous system. In peripheral sensory neurons, ASICs have been found on cell bodies and sensory terminals, where they appear to be important for nociception and mechanosensation. In central neurons, ASICs have been found on cell bodies, dendrites and at dendritic spines and have been suggested to contribute to synaptic plasticity. ASICs are activated by extracellular acidosis and their activation induces neuronal depolarization, sometimes associated with direct and indirect Ca2+ entry through voltage-gated calcium channels.
  • ASIC activation can be subject to modulation by extracellular alkalosis, intracellular pH, neuropeptides, polyamines, cations, arachidonic acid, lactate, nitric oxide, ATP, serotonin and exogenous modulators such as toxins from venoms, PcTx, APETx2, MitTx, and Mambalgin-1.
  • ASICs have been involved in several physiological processes such as nociception, mechanosensation, blood pressure regulation, synaptic function and plasticity. Growing evidence suggests an involvement of ASICs in anxiety and depression-related disorders.
  • a common feature in neuropathological conditions is acidosis arising from ischemia, inflammation, metabolism or synaptic transmission. Acidosis kills neurons and ASICs have been involved in mediating acid-induced neurotoxicity in neurological disorders such as ischemic stroke, epilepsy, multiple sclerosis, Huntington's disease, Parkinson's disease and spinal cord injury.
  • many types of brain tumors upregulate ASIC expression, which suggests a possible role of ASICs in tumor pathophysiology.
  • Ring A, X, Y, Z, R 1 , R 2 , R 3 , m, and n, is as defined and described in embodiments herein.
  • Compounds of the present invention, and pharmaceutically acceptable compositions thereof, are useful for treating a variety of diseases, disorders or conditions, associated with ASIC. Such diseases, disorders, or conditions include those described herein.
  • the present invention provides for inhibitors of ASIC. In certain aspects, the present invention provides for inhibitors of ASIC la. In some embodiments, such compounds include those of the formulae described herein, or a pharmaceutically acceptable salt thereof, wherein each variable is as defined and described herein.
  • aliphatic or "aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as “carbocycle” "cycloaliphatic” or “cycloalkyl”), that has a single point of attachment to the rest of the molecule.
  • aliphatic groups contain 1 -6 aliphatic carbon atoms.
  • aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms.
  • cycloaliphatic (or “carbocycle” or “cycloalkyl”) refers to a monocyclic C 3 -C 6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule.
  • Exemplary aliphatic groups are linear or branched, substituted or unsubstituted Ci-C 8 alkyl, C 2 -C 8 alkenyl, C 2 -C 8 alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
  • lower alkyl refers to a Ci -4 straight or branched alkyl group.
  • exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl.
  • lower haloalkyl refers to a Ci-4 straight or branched alkyl group that is substituted with one or more halogen atoms.
  • heteroatom means one or more of oxygen, sulfur, nitrogen, or phosphorus (including, any oxidized form of nitrogen, sulfur, or phosphorus; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4- dihydro-2H-pyrrolyl), ⁇ (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl)).
  • Ci -8 (or Ci- ⁇ ) saturated or unsaturated, straight or branched, hydrocarbon chain
  • bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.
  • alkylene refers to a bivalent alkyl group.
  • An "alkylene chain” is a polymethylene group, i.e., -(CH 2 ) n - wherein n is a positive integer, preferably from 1 to 6, from
  • a substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
  • alkenylene refers to a bivalent alkenyl group.
  • a substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
  • halogen means F, CI, Br, or I.
  • aryl used alone or as part of a larger moiety as in “aralkyl”, “aralkoxy”, or “aryloxyalkyl”, refers to monocyclic and bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains three to seven ring members.
  • aryl is used interchangeably with the term “aryl ring”.
  • aryl refers to an aromatic ring system.
  • Exemplary aryl groups are phenyl, biphenyl, naphthyl, anthracyl and the like, which optionally includes one or more substituents.
  • aryl is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like.
  • heteroaryl and “heteroar-”, used alone or as part of a larger moiety refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 ⁇ electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms.
  • heteroatom refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen.
  • Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl.
  • heteroaryl and “heteroar-”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring.
  • Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3-b]-l,4-oxazin- 3(4H)-one.
  • heteroaryl group is optionally mono- or bicyclic.
  • heteroaryl is used interchangeably with the terms “heteroaryl ring”, “heteroaryl group”, or “heteroaromatic”, any of which terms include rings that are optionally substituted.
  • heteroarylkyl refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
  • heterocycle As used herein, the terms “heterocycle”, “heterocyclyl”, “heterocyclic radical”, and “heterocyclic ring” are used interchangeably and refer to a stable 5- to 7— membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above.
  • nitrogen includes a substituted nitrogen.
  • the nitrogen is N (as in 3,4-dihydro- 2H-pyrrolyl), ⁇ (as in pyrrolidinyl), or + NR (as in N-substituted pyrrolidinyl).
  • a heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted.
  • saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl.
  • heterocycle used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H-indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl, where the radical or point of attachment is on the heterocyclyl ring.
  • a heterocyclyl group is optionally mono- or bicyclic.
  • heterocyclylalkyl refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
  • partially unsaturated refers to a ring moiety that includes at least one double or triple bond.
  • partially unsaturated is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
  • an "optionally substituted” group has a suitable substituent at each substitutable position of the group, and when more than one position in any given structure is substituted with more than one substituent selected from a specified group, the substituent is either the same or different at every position.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds.
  • stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
  • Suitable monovalent substituents on R° are independently deuterium, halogen, -(CH )o- 2 R*, -(haloR*), -(CH 2 )o- 2 OH, -(CH 2 )o- OR e , -(CH 2 )o- 2 CH(OR*) ; -0(haloR e ), -CN, -N 3 , -(CH )o- 2 C(0)R e , -(CH )o- 2 C(0)OH, -(CH 2 )o- 2 C(0)OR*, -(CH 2 )o- 2 SR*, -(CH 2 )o- 2 SH, -(CH 2 )o- 2 NH 2 , -(CH 2 )o- 2 NHR', -(CH 2 )o- 2 NR* 2 , -N0 2
  • Suitable divalent substituents that are bound to vicinal substitutable carbons of an "optionally substituted” group include: -0(CR * 2 ) 2 - 3 0-, wherein each independent occurrence of R * is selected from hydrogen, d-6 aliphatic which is optionally substituted as defined below, or an unsubstituted 5-6- membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R * include halogen, -R e , -(haloR*), -OH, -OR*, -O(haloR'), -CN, -C(0)OH, -C(0)OR e , -NH 2 , -NHR*, -NR* , or -N0 2 , wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci-4 aliphatic, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on a substitutable nitrogen of an "optionally substituted" group include -R ⁇ , -NR ⁇ 2 , -C(0)R ⁇ , -C(0)OR ⁇ , -C(0)C(0)R ⁇ , -C(0)CH 2 C(0)R+,
  • each R ⁇ is independently hydrogen, d-6 aliphatic which is optionally substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0 ⁇ 1 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R ⁇ , taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R ⁇ are independently halogen, -R e , -(haloR*), -OH, -OR', -O(haloR'), -CN, -C(0)OH, -C(0)OR e , -NH 2 , -NHR*, -NR* 2 , or -N0 2 , wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently C1-4 aliphatic, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6- membered saturated, partially unsaturated, or aryl ring having 0—4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • the terms “optionally substituted”, “optionally substituted alkyl,” “optionally substituted “optionally substituted alkenyl,” “optionally substituted alkynyl”, “optionally substituted carbocyclic,” “optionally substituted aryl”, “ optionally substituted heteroaryl,” “optionally substituted heterocyclic,” and any other optionally substituted group as used herein, refer to groups that are substituted or unsubstituted by independent replacement of one, two, or three or more of the hydrogen atoms thereon with typical substituents including, but not limited to:
  • -NH 2 protected amino, -NH alkyl, -NH alkenyl, -NH alkynyl, -NH cycloalkyl, -NH - aryl, -NH -heteroaryl, -NH -heterocyclic, -dialkylamino, -diarylamino, -diheteroarylamino,
  • -OCO 2 - alkyl -OC0 2 - alkenyl, -OC0 2 - alkynyl, -OC0 2 - carbocyclyl, -OC0 2 -aryl, - OC0 2 -heteroaryl, -OC0 2 -heterocyclyl, -OCONH2, -OCONH- alkyl, -OCONH- alkenyl, - OCONH- alkynyl, -OCONH- carbocyclyl, -OCONH- aryl, -OCONH- heteroaryl, -OCONH- heterocyclyl,
  • -S(O)- alkyl - S(O)- alkenyl, - S(O)- alkynyl, - S(0)- carbocyclyl, - S(0)-aryl, - S(O)- heteroaryl, - S(0)-heterocyclyl -S0 2 NH 2 , -S0 2 H- alkyl, -S0 2 NH- alkenyl, -S0 2 NH- alkynyl, - S0 2 NH- carbocyclyl, -S0 2 H- aryl, -S0 2 NH- heteroaryl, -S0 2 H- heterocyclyl,
  • -alkyl -alkenyl, -alkynyl, -aryl, -arylalkyl, -heteroaryl, -heteroarylalkyl, heterocycloalkyl, -cycloalkyl, -carbocyclic, -heterocyclic, polyalkoxyalkyl, polyalkoxy, - methoxymethoxy, -methoxyethoxy, -SH, -S- alkyl, -S- alkenyl, -S- alkynyl, -S- carbocyclyl, -S- aryl, -S-heteroaryl, -S-heterocyclyl, or methylthiomethyl.
  • Divalent groups include each group in either directions.
  • the group "- S0 2 NH-" in between group X and group Y includes both X-S0 2 H-Y and Y-S0 2 NH-X.
  • the term "pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate,
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (C 1 ⁇ alkyl) 4 salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
  • tautomer means each of two or more isomers of a compound that exist together in equilibrium, and are readily interchanged by migration of an atom or group within the molecule.
  • Tautomers are constitutional isomers of organic compounds that readily interconvert by a chemical reaction called tautomerizafion. This reaction commonly results in the formal migration of a hydrogen atom or proton, accompanied by a switch of a single bond and adjacent double bond (e.g., and in no way limited to this example, compounds of the followin tautomers are contemplated by the invention,
  • each form can be isolated and characterized.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • the group comprises one or more deuterium atoms.
  • a compound of the formula I includes isotope- labeled forms thereof.
  • An isotope-labeled form of a compound of the formula I is identical to this compound apart from the fact that one or more atoms of the compound have been replaced by an atom or atoms having an atomic mass or mass number which differs from the atomic mass or mass number of the atom which usually occurs naturally.
  • isotopes which are readily commercially available and which can be incorporated into a compound of the formula I by well- known methods include isotopes of hydrogen, carbon, nitrogen, oxygen, phos-phorus, fluo-rine and chlorine, for example 2 H, 3 H, 13 C, 14 C, 15 N, 18 0, O, 31 P, 2 P, 35 S, 18 F and 36 CI, respectively.
  • a compound of the formula I, a prodrug, thereof or a pharmaceutically acceptable salt of either which contains one or more of the above-mentioned isotopes and/or other isotopes of other atoms is intended to be part of the present invention.
  • An isotope-labeled compound of the formula I can be used in a number of beneficial ways.
  • an isotope-labeled compound of the formula I into which, for example, a radioisotope, such as 3 H or 14 C, has been incorporated is suitable for medicament and/or substrate tissue distribution assays.
  • radioisotopes i.e. tritium ( 3 H) and carbon-14 ( 14 C)
  • 3 H tritium
  • 14 C carbon-14
  • Incorporation of heavier isotopes, for example deuterium ( 2 H) into a compound of the formula I has therapeutic advantages owing to the higher metabolic stability of this isotope-labeled compound. Higher metabolic stability translates directly into an increased in vivo half-life or lower dosages, which under most circumstances would represent a preferred embodiment of the present invention.
  • An isotope-labeled compound of the formula I can usually be prepared by carrying out the procedures disclosed in the synthesis schemes and the related description, in the example part and in the preparation part in the present text, replacing a non-isotope-labeled reactant by a readily available isotope-labeled reactant.
  • Deuterium ( 2 H) can also be incorporated into a compound of the formula I for the purpose in order to manipulate the oxidative metabolism of the compound by way of the primary kinetic isotope effect.
  • the primary kinetic isotope effect is a change of the rate for a chemical reaction that results from exchange of isotopic nuclei, which in turn is caused by the change in ground state energies necessary for covalent bond formation after this isotopic exchange.
  • Exchange of a heavier isotope usually results in a lowering of the ground state energy for a chemical bond and thus causes a reduction in the rate in rate-limiting bond breakage.
  • the product distribution ratios can be altered substantially.
  • Half-life determinations enable favorable and accurate determination of the extent of the extent to which the improvement in resistance to oxidative metabolism has improved. In this way, it is determined that the half-life of the parent compound can be extended by up to 100% as the result of deuterium-hydrogen exchange of this type.
  • Deuterium-hydrogen exchange in a compound of the formula I can also be used to achieve a favorable modification of the metabolite spectrum of the starting compound in order to diminish or eliminate undesired toxic metabolites. For example, if a toxic metabolite arises through oxidative carbon-hydrogen (C-H) bond cleavage, it can reasonably be assumed that the deuterated analogue will greatly diminish or eliminate production of the unwanted metabolite, even if the particular oxidation is not a rate-determining step. Further information on the state of the art with respect to deuterium-hydrogen exchange may be found, for example in Hanzlik et al., J. Org. Chem.
  • a modulator is defined as a compound that binds to and /or inhibits the target with measurable affinity.
  • a modulator has an IC 5 o and/or binding constant of less about 50 ⁇ , less than about 5 ⁇ , less than about 1 ⁇ , less than about 500 nM, less than about 100 nM, or less than about 10 nM.
  • measurable affinity and “measurably inhibit,” as used herein, means a measurable change in ASIC activity between a sample comprising a compound of the present invention, or composition thereof, and ASIC, and an equivalent sample comprising ASIC, in the absence of said compound, or composition thereof.
  • the present invention provides a compound of formula I,
  • X is C(R 4 ) 2 , C(R 4 ), N, N-R 4 , O, S, SO, or S0 2 ;
  • Ring A is a fused C5-6 aryl, or a fused 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each R 1 is independently -R, halogen, -OR, -SR, -CN, -N0 2 , -S0 2 R, -SOR, -C(0)R, -C0 2 R,
  • Z is C(R 4 ) or N
  • each R 2 is independently -R, halogen, -OR, -SR, -CN, -NO2, -S0 2 R, -SOR, -C(0)R, -C0 2 R, -C(0)N(R) 2 , -NRC(0)R, -NRC(0)N(R) 2 , -NRSO2R, or -N(R) 2 ; two R 2 groups on the same atom are taken together with the atom to which they are attached to form a carbonyl, thiocarbonyl, or imine; each of which is optionally substituted;
  • R 3 is -R, halogen, -OR, -SR, -CN, -NO2, -S0 2 R, -SOR, -C(0)R, -C0 2 R, -C(0)N(R) 2 , - NRC(0)R, -NRC(0)N(R) 2 , -NRS0 2 R, or -N(R ;
  • each R 4 is independently hydrogen, Ci- 6 aliphatic, C 5 _io aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted;
  • each R is independently hydrogen, Ci-e aliphatic, C 5 - 10 aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted; or
  • n 0, 1, or 2;
  • n 0, 1, 2, 3, or 4.
  • X is C(R 4 ) 2 , C(R 4 ), N, N-R 4 , O, or S.
  • X is CH 2 , CH, N, NH, N-CH 3 , or S.
  • X is CH 2 . In certain embodiments, X is CH. In certain embodiments, X is N. In certain embodiments, X is NH. In certain embodiments, X is N-CH3. In certain embodiments, X is S.
  • Ring A is fused phenyl, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl;- l,2,5oxadiazolyl,
  • Ring A is a fused phenyl, pyridinyl, thiophenyl, or furanyl.
  • Ring A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • Ring A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • each R 1 is independently -R, halogen, -OR, -SR, -CN, -N0 2 , -SO2R, -SOR, -C(0)R, -C0 2 R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R>2, -NRSO2R, or -N(R>2.
  • each R 1 is H.
  • each R 1 is independently Ci- 6 aliphatic, Cs-io aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted, or each R 1 is independently halogen, -OR, -SR, -CN, -N0 2 , -SO2R, -SOR, -C(0)R, -C0 2 R, -C(0)N(R) 2 , -NRC(0)R, -NRC(0)N(R)2, -NRS0 2 R, or -N(R>2.
  • each R 1 is independently methyl, ethyl, propyl, i-propyl, n-
  • Z is C(R 4 ).
  • Z is CH.
  • Z is N.
  • R 2 is H.
  • R 2 is C1-0 aliphatic, C5-10 aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted; or R 3 is halogen, -OR, -SR, -CN, -NO2, -S0 2 R, -SOR,
  • R 2 is methyl, ethyl, propyl, i-propyl, n-butyl, s-butyl, t-butyl, straight chain or branched pentyl, or straight chain or branched hexyl, each of which is optionally substituted.
  • R 2 is phenyl, naphthyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantyl, cyclooctyl, [3.3.0]bicyclooctanyl, [4.3.0]bicyclononanyl, [4.4.0]bicyclodecanyl, [2.2.2]bicyclooctanyl, fluorenyl, indanyl, tetrahydronaphthyl, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carb
  • R 2 is halogen, -OR, -SR, -CN, -N0 2 , -SO2R, -SOR, -C(0)R, -C0 2 R, -C(0)N(R) 2 , -NRC(0)R, -NRC(0)N(R)2, -NRSO2R, or -N(R) 2 .
  • R 3 is -R, halogen, -OR, -SR, -CN, -N0 2 , -S0 2 R, -SOR, -C(0)R, -C0 2 R, -C(0)N(R) 2 , -NRC(0)R, -NRC(0)N(R) 2 , -NRS0 2 R, or -N(R) 2 .
  • R 3 is H.
  • R 3 is Ci-6 aliphatic, Cs-io aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted; or R 3 is halogen, -OR, -SR, -CN,— NO2, -S0 2 R, -SOR, -C(0)R, -CO2R, -C(0)N(R) 2 , -NRC(0)R, -NRC(0)N(R) 2 , -NRSO2R, or -N(R) 2 .
  • R 3 is methyl, ethyl, propyl, i-propyl, n-butyl, s-butyl, t-butyl, straight chain or branched pentyl, or straight chain or branched hexyl, each of which is optionally substituted.
  • R 3 is phenyl, naphthyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantyl, cyclooctyl, [3.3.0]bicyclooctanyl, [4.3.0]bicyclononanyl, [4.4.0]bicyclodecanyl, [2.2.2]bicyclooctanyl, fluorenyl, indanyl, tetrahydronaphthyl, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carb
  • R 3 is phenyl, cyclohexyl, imidazolyl, pyrazinyl, pyridazinyl, pyridinyl, pyridyl, pyrimidinyl, tetrahydrofuranyl, or tetrahydropyranyl; each of which is optionally substituted.
  • R 3 is halogen, -OR, -SR, -CN, -N0 2 , -S0 2 R, -SOR, -C(0)R, -C0 2 R, -C(0)N(R) 2 , -NRC(0)R, -NRC(0)N(R) 2 , -NRS0 2 R, or -NCR ⁇ .
  • R 3 is -C(0)R, -C0 2 R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R) 2 , -NRS0 2 R, or -N(R>2.
  • R 3 is methyl, ethyl, propyl, phenyl, cyclohexyl, imidazolyl, pyrazinyl, pyridazinyl, pyridinyl, pyridyl, pyrimidinyl, tetrahydrofuranyl, or tetrahydropyranyl; each of which is optionally substituted; or R 3 is -C(0)R, -C0 2 R, -C(0)N(R , -NRC(0)R, - RC(0)N(R) 2 , -NRS0 2 R, or -NCR ⁇ .
  • R 4 is H.
  • R 4 is d-6 aliphatic, C5-10 aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1 -4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted, or halogen.
  • each of X, Y, Z, Ring A, R, R 1 , R 2 , R 3 , R 4 , m, and n is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula I-a,
  • the present invention provides a compound of formula I-b,
  • R 3 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula I-c,
  • each of R 3 and R 4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula I-d,
  • each of R 3 and R 4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula I-e,
  • each of R 3 and R 4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula I-f,
  • R 3 is as defined above and described embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula I-g,
  • each of R 3 and R 4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula I-h,
  • R 3 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula I-j,
  • each of R 3 and R 4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula I-k,
  • each of R 3 and R 4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula 1-1,
  • R 3 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula I-m,
  • R 3 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the present invention provides a compound of formula I-n,
  • R 3 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
  • the resent invention provides a compound of formula I-o,
  • the invention provides a compound selected from Table 1 :
  • the present invention provides a compound selected from those depicted above, or a pharmaceutically acceptable salt thereof.
  • the compounds of the invention were synthesized in accordance with the schemes provided in the Examples below.
  • the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the amount of compound in compositions of this invention is such that is effective to measurably inhibit ASIC, or a mutant thereof, in a biological sample or in a patient.
  • the amount of compound in compositions of this invention is such that is effective to measurably inhibit ASIC, or a mutant thereof, in a biological sample or in a patient.
  • a composition of this invention is formulated for administration to a patient in need of such composition.
  • patient or "subject”, as used herein, means an animal, preferably a mammal, and most preferably a human.
  • compositions of this invention refers to a nontoxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated.
  • Pharmaceutically acceptable carriers, adjuvants or vehicles that are used in the compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-
  • a "pharmaceutically acceptable derivative” means any non-toxic salt, ester, salt of an ester or other derivative of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.
  • compositions of the present invention are administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intraarticular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions of this invention include aqueous or oleaginous suspension. These suspensions are formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example as a solution in 1 ,3-butanediol.
  • a nontoxic parenterally acceptable diluent or solvent for example as a solution in 1 ,3-butanediol.
  • acceptable vehicles and solvents that are employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil employed includes synthetic mono- or di- glycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms are also be used for the purposes of formulation.
  • compositions of this invention are orally administered in any orally acceptable dosage form.
  • exemplary oral dosage forms are capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents are optionally also added.
  • compositions of this invention are administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
  • a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • Such materials include cocoa butter, beeswax and polyethylene glycols.
  • Pharmaceutically acceptable compositions of this invention are also administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches are also used.
  • compositions are formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • exemplary carriers for topical administration of compounds of this aremineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • provided pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • compositions of this invention are optionally administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and are prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
  • compositions of this invention are formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions of this invention are administered without food. In other embodiments, pharmaceutically acceptable compositions of this invention are administered with food.
  • compositions of the present invention that are optionally combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration.
  • provided compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the compound can be administered to a patient receiving these compositions.
  • a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
  • the amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
  • the invention provides a method for inhibiting or antagonizing ASIC in a patient or in a biological sample comprising the step of administering to said patient or contacting said biological sample with a compound according to the invention.
  • the invention is directed to the use of compounds of the invention and/or physiologically acceptable salts thereof, for modulating or inhibiting/antagonizing ASIC.
  • modulation denotes any change in ASIC-mediated signal transduction, which is based on the action of the specific inventive compounds capable to interact with the ASIC target in such a manner that makes recognition, binding and activating possible.
  • the compounds are characterized by such a high affinity to ASIC.
  • the substances are highly selective for ASIC over most other channels in order to guarantee an exclusive and directed recognition with the single ASIC target.
  • the term "recognition" - without being limited thereto - relates to any type of interaction between the specific compounds and the target, particularly covalent or non- covalent binding or association, such as a covalent bond, hydrophobic/ hydrophilic interactions, van der Waals forces, ion pairs, hydrogen bonds, ligand-receptor (enzyme-inhibitor) interactions, and the like.
  • Such association may also encompass the presence of other molecules such as peptides, proteins or nucleotide sequences.
  • the present ion channel interaction is characterized by high affinity, high selectivity and minimal or even lacking cross-reactivity to other target molecules to exclude unhealthy and harmful impacts to the treated subject.
  • the present invention relates to a method for inhibiting or antagonizing ASIC, with at least one compound of formula (I) according to the invention and/or physiologically acceptable salts thereof, under conditions such that said ASIC is inhibited/antagonozied.
  • the system is a cellular system.
  • the cellular system is defined to be any subject provided that the subject comprises cells.
  • the cellular system can be selected from the group of single cells, cell cultures, tissues, organs and animals.
  • the method for modulating ASIC is performed in-vitro.
  • the prior teaching of the present specification concerning the compounds of formula (I), including any embodiments thereof, is valid and applicable without restrictions to the compounds according to formula (I) and their salts when used in the method for inhibiting/antagonizing ASIC.
  • the prior teaching of the present specification concerning the compounds of formula (I), including any embodiments thereof, is valid and applicable without restrictions to the compounds according to formula (I) and their salts when used in the method for inhibiting/antagonizing ASIC.
  • the present invention provides a method for treating an ASIC-mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.
  • a method for the treatment or lessening the severity of acute, chronic, neuropathic, or inflammatory pain, arthritis, migrane, cluster headaches, trigeminal neuralgia, herpetic neuralgia, general neuralgias, epilepsy or epilepsy conditions, neurodegenerative disorders, psychiatric disorders such as anxiety and depression, myotonia, arrhythmia, movement disorders, neuroendocrine disorders, ataxia, multiple sclerosis, irritable bowel syndrome, incontinence, visceral pain, osteoarthritis pain, postherpetic neuralgia, diabetic neuropathy, radicular pain, sciatica, back pain, head or neck pain, severe or intractable pain, nociceptive pain, breakthrough pain, postsurgical pain, or cancer pain comprising administering an effective amount of a compound, or a pharmaceutically acceptable composition comprising a compound to a subject in need thereof.
  • a method for the treatment or lessening the severity of acute, chronic, neuropathic, or inflammatory pain comprising administering an effective amount of a compound or a pharmaceutically acceptable composition to a subject in need thereof.
  • a method for the treatment or lessening the severity of radicular pain, sciatica, back pain, head pain, or neck pain is provided comprising administering an effective amount of a compound or a pharmaceutically acceptable composition to a subject in need thereof.
  • a method for the treatment or lessening the severity of severe or intractable pain, acute pain, postsurgical pain, back pain, tinnitis or cancer pain comprising administering an effective amount of a compound or a pharmaceutically acceptable composition to a subject in need thereof.
  • the compounds of the present invention are useful in the prophylaxis and treatment of autoimmune and/or inflammatory disorders, including neurodegenerative diseases, such as multiple sclerosis (MS), polyneuritis, multiple neuritis, amyotrophic lateral sclerosis (ALS), Alzheimer's disease, optic neuritis, or Parkinson's disease.
  • neurodegenerative diseases such as multiple sclerosis (MS), polyneuritis, multiple neuritis, amyotrophic lateral sclerosis (ALS), Alzheimer's disease, optic neuritis, or Parkinson's disease.
  • the present invention furthermore relates to a method of treating a subject suffering from an immunerogulatory abnomality, comprising administering to said subject a compound of formula I in an amount that is effective for treating said immunoregulatory abnormality.
  • the present invention preferably relates to a method wherein the immunoregulatory abnormality is an autoimmune or chronic inflammatory disease selected from the group consisting of: amyotrophic lateral sclerosis (ALS), Alzheimer's disease, Parkinson's disease, systemic lupus erythematosus, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cirrhosis, uveitis, multiple sclerosis, Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, psoriasis, autoimmune myositis, Wegener's granulomatosis, ichthyosis, Graves' ophthalmopathy and asthma.
  • the present invention furthermore relates to a method wherein the immunoregulatory abnormality is bone marrow or organ transplant rejection or graft-versus-host disease.
  • the present invention furthermore relates to a method wherein the immunoregulatory abnormality is selected from the group consisting of: transplantation of organs or tissue, graft- versus-host diseases brought about by transplantation, autoimmune syndromes including rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, posterior uveitis, allergic encephalomyelitis, glomerulonephritis, post-infectious autoimmune diseases including rheumatic fever and postinfectious glomerulonephritis, inflammatory and hyperproliferative skin diseases, psoriasis, atopic dermatitis, contact dermatitis, eczematous dermatitis, sebor
  • the disorder or disease is anxiety.
  • the disorder or disease is MS.
  • the disorder or disease is optic neuritis.
  • the disorder or disease is depression-related disorders
  • the disorder or disease is acidosis. In certain embodiments, the disorder or disease is acidosis arising from ischemia, inflammation, metabolism or synaptic transmission.
  • the disorder or disease is ischemic stroke, epilepsy, multiple sclerosis, Huntington's disease, Parkinson's disease or spinal cord injury.
  • the disorder or disease is cancer.
  • the disorder or disease is brain cancer or brain tumor.
  • an "effective amount" of the compound or pharmaceutically acceptable composition is that amount effective for treating or lessening the severity of a disease or disorder provide supra.
  • compositions may be administered using any amount and any route of administration effective for treating or lessening the severity of a disease or disorder provide supra.
  • dosage unit form refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
  • patient means an animal, preferably a mammal, and most preferably a human.
  • the compounds of the invention are useful as inhibitors of voltage-gated ion channels.
  • the compounds and compositions of the invention are inhibitors of one or more of ASIC la, ASIC2a, ASIC2b, ASIC3 or ASIC4, and thus, without wishing to be bound by any particular theory, the compounds and compositions are particularly useful for treating or lessening the severity of a disease, condition, or disorder where activation or hyperactivity of one or more of ASIC la, ASIC2a, ASIC2b, ASIC3 and ASIC4 is implicated in the disease, condition, or disorder.
  • the present invention provides a method for treating or lessening the severity of a disease, condition, or disorder where activation or hyperactivity of one or more of ASIC la, ASIC2a, ASIC2b, ASIC3 and ASIC4 is implicated in the disease state.
  • the compounds and compositions of the invention are inhibitors of ASIC.
  • the compound is administered in an effective amount as defined above.
  • the treatment is an oral administration.
  • the method of the invention can be performed either in-vitro or in-vivo.
  • the susceptibility of a particular cell to treatment with the compounds according to the invention can be particularly determined by in-vitro tests, whether in the course of research or clinical application.
  • a culture of the cell is combined with a compound according to the invention at various concentrations for a period of time which is sufficient to allow the active agents to inhibit ASIC activity, usually between about one hour and one week.
  • In-vitro treatment can be carried out using cultivated cells from a biopsy sample or cell line.
  • the host or patient can belong to any mammalian species, for example a primate species, particularly humans; rodents, including mice, rats and hamsters; rabbits; horses, cows, dogs, cats, etc. Animal models are of interest for experimental investigations, providing a model for treatment of human disease.
  • a signal transduction pathway For identification of a signal transduction pathway and for detection of interactions between various signal transduction pathways, various scientists have developed suitable models or model systems, for example cell culture models and models of transgenic animals. For the determination of certain stages in the signal transduction cascade, interacting compounds can be utilized in order to modulate the signal.
  • the compounds according to the invention can also be used as reagents for testing ASIC-dependent signal transduction pathways in animals and/or cell culture models or in the clinical diseases mentioned in this application.
  • the invention also relates to the use of compounds according to formula (I) and/or physiologically acceptable salts thereof for the prophylactic or therapeutic treatment and/or monitoring of diseases that are caused, mediated and/or propagated by ASIC activity. Furthermore, the invention relates to the use of compounds according to formula (I) and/or physiologically acceptable salts thereof for the production of a medicament for the prophylactic or therapeutic treatment and/or monitoring of diseases that are caused, mediated and/or propagated by ASIC activity. In certain embodiments, the invention provides the use of a compound according to formula I or physiologically acceptable salts thereof, for the production of a medicament for the prophylactic or therapeutic treatment of an ASIC-mediated disorder.
  • Compounds of formula (I) and/or a physiologically acceptable salt thereof can furthermore be employed as intermediate for the preparation of further medicament active ingredients.
  • the medicament is preferably prepared in a non-chemical manner, e.g. by combining the active ingredient with at least one solid, fluid and/or semi-fluid carrier or excipient, and optionally in conjunction with a single or more other active substances in an appropriate dosage form.
  • the compounds of formula (I) according to the invention can be administered before or following an onset of disease once or several times acting as therapy.
  • the aforementioned compounds and medical products of the inventive use are particularly used for the therapeutic treatment.
  • a therapeutically relevant effect relieves to some extent one or more symptoms of a disorder, or returns to normality, either partially or completely, one or more physiological or biochemical parameters associated with or causative of a disease or pathological condition.
  • Monitoring is considered as a kind of treatment provided that the compounds are administered in distinct intervals, e.g. in order to boost the response and eradicate the pathogens and/or symptoms of the disease completely. Either the identical compound or different compounds can be applied.
  • the methods of the invention can also be used to reduce the likelihood of developing a disorder or even prevent the initiation of disorders associated with ASIC activity in advance or to treat the arising and continuing symptoms.
  • prophylactic treatment is advisable if the subject possesses any preconditions for the aforementioned physiological or pathological conditions, such as a familial disposition, a genetic defect, or a previously incurred disease.
  • the invention furthermore relates to a medicament comprising at least one compound according to the invention and/or pharmaceutically usable derivatives, salts, solvates and stereoisomers thereof, including mixtures thereof in all ratios.
  • the invention relates to a medicament comprising at least one compound according to the invention and/or physiologically acceptable salts thereof.
  • a “medicament” in the meaning of the invention is any agent in the field of medicine, which comprises one or more compounds of formula (I) or preparations thereof (e.g. a pharmaceutical composition or pharmaceutical formulation) and can be used in prophylaxis, therapy, follow-up or aftercare of patients who suffer from diseases, which are associated with ASIC activity, in such a way that a pathogenic modification of their overall condition or of the condition of particular regions of the organism could establish at least temporarily.
  • the active ingredient may be administered alone or in combination with other treatments.
  • a synergistic effect may be achieved by using more than one compound in the pharmaceutical composition, i.e. the compound of formula (I) is combined with at least another agent as active ingredient, which is either another compound of formula (I) or a compound of different structural scaffold.
  • the active ingredients can be used either simultaneously or sequentially.
  • Antiinflammatory agents include but are not limited to NSAIDs, non-specific and COX-2 specific cyclooxygenase enzyme inhibitors, gold compounds, corticosteroids, methotrexate, tumor necrosis factor (TNF) antagonists, immunosuppressants and methotrexate.
  • NSAIDs include, but are not limited to, ibuprofen, flurbiprofen, naproxen and naproxen sodium, diclofenac, combinations of diclofenac sodium and misoprostol, sulindac, oxaprozin, diflunisal, piroxicam, indomethacin, etodolac, fenoprofen calcium, ketoprofen, sodium nabumetone, sulfasalazine, tolmetin sodium, and hydroxychloroquine.
  • NSAIDs also include COX-2 specific inhibitors such as celecoxib, valdecoxib, lumiracoxib dnd/or etoricoxib.
  • the anti-inflammatory agent is a salicylate.
  • Salicylates include by are not limited to acetylsalicylic acid or aspirin, sodium salicylate, and choline and magnesium salicylates.
  • the anti-inflammatory agent may also be a corticosteroid.
  • the corticosteroid may be cortisone, dexamethasone, methylprednisolone, prednisolone, prednisolone sodium phosphate, or prednisone.
  • the anti-inflammatory agent is a gold compound such as gold sodium thiomalate or auranofin.
  • the invention also includes embodiments in which the anti-inflammatory agent is a metabolic inhibitor such as a dihydrofolate reductase inhibitor, such as methotrexate or a dihydroorotate dehydrogenase inhibitor, such as leflunomide.
  • a metabolic inhibitor such as a dihydrofolate reductase inhibitor, such as methotrexate or a dihydroorotate dehydrogenase inhibitor, such as leflunomide.
  • At least one anti-inflammatory compound is an anti-monoclonal antibody (such as eculizumab or pexelizumab), a TNF antagonist, such as entanercept, or infliximab, which is an anti-TNF alpha monoclonal antibody.
  • an anti-monoclonal antibody such as eculizumab or pexelizumab
  • TNF antagonist such as entanercept, or infliximab
  • Still other embodiments of the invention pertain to combinations in which at least one active agent is an immunosuppressant compound such as an immunosuppressant compound chosen from methotrexate, leflunomide, cyclosporine, tacrolimus, azathioprine, and mycophenolate mofetil.
  • an immunosuppressant compound such as an immunosuppressant compound chosen from methotrexate, leflunomide, cyclosporine, tacrolimus, azathioprine, and mycophenolate mofetil.
  • the compounds of the invention are also used in combination with chemotherapeutic drugs, in particular, drugs that induce apoptosis.
  • chemotherapeutic drugs that can be used in combination with chemosensitizing ASIC inhibitors include topoisomerase I inhibitors (camptothecin or topotecan), topoisomerase II inhibitors (e.g. daunomycin and etoposide), alkylating agents (e.g. cyclophosphamide, melphalan and BCNU), tubulin directed agents (e.g. taxol and vinblastine), and biological agents (e.g. antibodies such as anti CD20 antibody, IDEC 8, immunotoxins, and cytokines).
  • topoisomerase I inhibitors camptothecin or topotecan
  • topoisomerase II inhibitors e.g. daunomycin and etoposide
  • alkylating agents e.g. cyclophosphamide, melphalan and BCNU
  • anticancer agent relates to any agent which is administered to a patient with cancer for the purposes of treating the cancer.
  • the anti-cancer treatment defined above may be applied as a monotherapy or may involve, in addition to the herein disclosed compounds of formula I, conventional surgery or radiotherapy or medicinal therapy.
  • Such medicinal therapy e.g. a chemotherapy or a targeted therapy, may include one or more, but preferably one, of the following anti-tumor agents:
  • Alkylating agents such as altretamine, bendamustine, busulfan, carmustine, chlorambucil, chlormethine, cyclophosphamide, dacarbazine, ifosfamide, improsulfan, tosilate, lomustine, melphalan, mitobronitol, mitolactol, nimustine, ranimustine, temozolomide, thiotepa, treosulfan, mechloretamine, carboquone; apaziquone, fotemustine, glufosfamide, palifosfamide, pipobroman, trofosfamide, uramustine, TH-302 4 , VAL-083 4 ;
  • Platinum Compounds such as carboplatin, cisplatin, eptaplatin, miriplatine hydrate, oxaliplatin, lobaplatin, nedaplatin, picoplatin, satraplatin; lobaplatin, nedaplatin, picoplatin, satraplatin;
  • DNA altering agents such as amrubicin, bisantrene, decitabine, mitoxantrone, procarbazine, trabectedin, clofarabine; amsacrine, brostallicin, pixantrone, laromustine 1 ' 3 ;
  • Topoisomerase Inhibitors such as etoposide, irinotecan, razoxane, sobuzoxane, teniposide, topotecan; amonafide, belotecan, elliptinium acetate, voreloxin;
  • Microtubule modifiers such as cabazitaxel, docetaxel, eribulin, ixabepilone, paclitaxel, vinblastine, vincristine, vinorelbine, vindesine, vinflunine; fosbretabulin, tesetaxel;
  • Antimetabolites such as asparaginase 3 , azacitidine, calcium levofolinate, capecitabine,
  • cladribine cytarabine
  • enocitabine floxuridine
  • fludarabine fluorouracil
  • gemcitabine
  • mercaptopurine methotrexate, nelarabine, pemetrexed, pralatrexate, azathioprine, thioguanine, carmofur; doxifluridine, elacytarabine, raltitrexed, sapacitabine, tegafur 2 ' 3 , trimetrexate;
  • Anticancer antibiotics such as bleomycin, dactinomycin, doxorubicin, epirubicin, idarubicin, levamisole, miltefosine, mitomycin C, romidepsin, streptozocin, valrubicin, zinostatin, zorubicin, daunurobicin, plicamycin; aclarubicin, peplomycin, pirarubicin;
  • Hormones/ Antagonists such as abarelix, abiraterone, bicalutamide, buserelin, calusterone, chlorotrianisene, degarelix, dexamethasone, estradiol, fluocortolone
  • Aromatase inhibitors such as aminoglutethimide, anastrozole, exemestane, fadrozole, letrozole, testolactone; formestane;
  • Small molecule kinase inhibitors such as crizotinib, dasatinib, erlotinib, imatinib, lapatinib, nilotinib, pazopanib, regorafenib, ruxolitinib, sorafenib, sunitinib, vandetanib, vemurafenib, bosutinib, gefitinib, axitinib; afatinib, alisertib, dabrafenib, dacomitinib, dinaciclib, dovitinib, enzastaurin, nintedanib, lenvatinib, linifanib, linsitinib, masitinib, midostaurin, motesanib, neratinib, orantinib, perifosine, ponatinib, radotinib, rigo
  • Photosensitizers such as methoxsalen 3 ; porfimer sodium, talaporfin, temoporfin;
  • Antibodies such as alemtuzumab, besilesomab, brentuximab vedotin, cetuximab, denosumab, ipilimumab, ofatumumab, panitumumab, rituximab, tositumomab,
  • trastuzumab bevacizumab, pertuzumab 2 3 ; catumaxomab, elotuzumab, epratuzumab, farletuzumab, mogamulizumab, necitumumab, nimotuzumab, obinutuzumab, ocaratuzumab, oregovomab, ramucirumab, rilotumumab, siltuximab, tocilizumab, zalutumumab, zanolimumab, matuzumab, dalotuzumab 1 2 ' 3 , onartuzumab 1 ' 3 , racotumomab 1 , tabalumab 1,3 , EMD-525797 4 , nivolumab 1 - 3 ;
  • Cytokines such as aldesleukin, interferon alfa 2 , interferon alfa2a 3 , interferon alfa2b 2 - 3 ;
  • Drug Conjugates such as denileukin diftitox, ibritumomab tiuxetan, iobenguane 1123,
  • trastuzumab emtansine prednimustine, trastuzumab emtansine, estramustine, gemtuzumab, ozogamicin, aflibercept;
  • cintredekin besudotox edotreotide, inotuzumab ozogamicin, naptumomab estafenatox, oportuzumab monatox, technetium (99mTc) arcitumomab 1,3 , vintafolide 1 ' 3 ;
  • Vaccines such as sipuleucel 3 ; vitespen 3 , emepepimut-S 3 , oncoVAX 4 , rindopepimut 3 , troVax 4 , MGN-1601 4 , MGN-1703 4 ; and
  • the invention provides for a kit consisting of separate packs of an effective amount of a compound according to the invention and/or pharmaceutically acceptable salts, derivatives, solvates and stereoisomers thereof, including mixtures thereof in all ratios, and optionally, an effective amount of a further active ingredient.
  • the kit comprises suitable containers, such as boxes, individual bottles, bags or ampoules.
  • the kit may, for example, comprise separate ampoules, each containing an effective amount of a compound according to the invention and/or pharmaceutically acceptable salts, derivatives, solvates and stereoisomers thereof, including mixtures thereof in all ratios, and an effective amount of a further active ingredient in dissolved or lyophilized form.
  • treatment refers to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein.
  • treatment is administered after one or more symptoms have developed.
  • treatment is admmistered in the absence of symptoms.
  • treatment is administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment is also continued after symptoms have resolved, for example to prevent or delay their recurrence.
  • the compounds and compositions, according to the method of the present invention are administered using any amount and any route of administration effective for treating or lessening the severity of a disorder provided above.
  • the exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like.
  • Compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
  • compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated.
  • the compounds of the invention are administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 100 mg/kg and preferably from about 1 mg/kg to about 50 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms optionally contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • the oral compositions can also include
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions are formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation are also a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • a compound of the present invention In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This is accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide- polyglycolide.
  • the rate of compound release can be controlled.
  • biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar—agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol
  • Solid compositions of a similar type are also employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
  • Solid compositions of a similar type are also employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • the active compounds can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms optionally also comprise buffering agents. They optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • buffering agents optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • embedding compositions that can be used include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as required.
  • Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention.
  • the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • Such dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • the invention relates to a method of inhibiting ASIC activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
  • the invention relates to a method of inhibiting ASIC, or a mutant thereof, activity in a biological sample in a positive manner, comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
  • the compounds of the invention can be applied either themselves and/or in combination with physical measurements for diagnostics of treatment effectiveness.
  • Pharmaceutical compositions containing said compounds and the use of said compounds to treat ASIC-mediated conditions is a promising, novel approach for a broad spectrum of therapies causing a direct and immediate improvement in the state of health, whether in human or animal.
  • the orally bioavailable and active new chemical entities of the invention improve convenience for patients and compliance for physicians.
  • biological sample includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • Modulation of ASIC, or a mutant thereof, activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ transplantation, biological specimen storage, and biological assays.
  • -BSVT temperature controller [00198] -GRASP II gradient spectroscopy accessory for fast acquisition of 2D pulse sequences
  • Example compounds and their intermediates were analysed by HPLC-MS using a combination of the following instrumentation: Shimadzu, Waters or Micromass ZMD, ZQ or LCT mass spectrometers with an Agilent, Waters or Polymer Labs UV and ELS detector.
  • the HPLC conditions are tabulated below.
  • Micromass MassLynx Operating Software with OpenLynx Browser were used for data acquisition, processing and reporting.
  • Route A was followed using methyl 3-isothiocyanato-5-methylthiophene-2- Carboxylate in the place of methyl 3-isothiocyanatothiophene-2 -carboxylate.
  • methyl 3-isothiocyanato-5-methylthiophene-2-Carboxylate was prepared in the following manner: A solution of thiophosgene (0.12 ml, 1.61 mmol) in Chloroform (4ml) was added dropwise to aqueous NaHC03 (2M, 2ml), then methyl 3-amino-5-methylthiophene-2- carboxylate (250 mg, 1.46 mmol) in Chloroform (4ml) was added via a dropping funnel and was stirred at r.t over night. The organic layer was separated and the aqueous layer extracted with CHC13. Combined organics were dried over Na2S04.
  • Methyl 3-isothiocyanatopyridine-2-carboxylate (570 mg, 2.93 mmol) was dissolved in THF (7 ml) in a pressure tube and 7M ammonia in MeOH (0.55 ml) was added. Reaction was sealed and heated to 80°C. Reaction was heated for 6h. IPC showed mainly cyclised product. The reaction was allowed to cool to r.t and the ppt was filtered and washed with THF to afford 465 mg (86.6%) the title compound as an off white solid.
  • 6-(4- ⁇ 4-oxo-3H,4H-thieno [3 ,2-d]pyrimidin-2-yl ⁇ piperazin- 1 -y l)pyridine-3 - carbonitrile was prepared using general procedure B and then hydrolysis as described below gave the final product, 6-(4- ⁇ 4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl ⁇ piperazin-l-yl)pyridine- 3-carboxamide: A mixture of 6-(4- ⁇ 4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl ⁇ piperazin-l- yl)pyridine-3 -carbonitrile (50 mg, 0.15 mmol) in H2S04 (0.5 ml) under nitrogen was stirred at rt overnight.
  • RuPhos Precatalyst (0.59 g, 0.72 mmol) was then added and the mixture heated to 85 °C overnight.
  • the reaction mixture was partitioned between EtOAc (50 ml) and water (100 ml). The two phases were separated and the aqueous layer extracted with EtOAc (50 ml). The combined organic extracts were washed with brine (100 ml), dried over MgS04, filtered and concentrated in vacuo.
  • the compound is purified using column chromatography, eluting at a gradient of 17- 92% EtOAc in heptane to yield 1.74g, 77% of the title compound as a yellow powder.
  • METCR1673 Generic 2 minutes M/Z (ES+) 289, Retention time 1.26.
  • tert-butyl 4-(5-cyanopyrazin-2-yl)piperazine-l -carboxylate required for the synthesis was prepared in the following manner: To a solution of 5-chloropyrazine-2-carbonitrile (200 mg, 1.43 mmol) and tert-butyl piperazine-l-carboxylate (266.95 mg, 1.43 mmol) in anhydrous DMA (5 ml) under N2 was added DIPEA (748.95 ⁇ , 4.3 mmol). The mixture was stirred and heated at 150°C in a microwave for 90 mins. The mixture was allowed to cool to RT, prior to adding water until a significant amount of precipitate was observed. This was filtered under vacuum, washed with water and dried to afford 363mg, 87% of titled compound as a light brown solid. METCR1673 Generic 2 minutes M/Z (ES+) 233.9, Retention time 1.28.
  • tert-butyl 4- ⁇ 4-[(3-hydroxypropyl)carbamoyl]phenyl ⁇ piperazine-l -carboxylate required for the synthesis was prepared as follows: 4- ⁇ 4-[(tert-butoxy)carbonyl]piperazin-l- yl ⁇ benzoic acid (1 g, 3.26 mmol) was dissolved in DMF (15ml) and DIPEA (1.62 ml, 9.79 mmol) and HATU (1489.38 mg, 3.92 mmol) were added to the mixture and stirred at room temperature for 30 minutes. 3-aminopropan-l-ol (0.5 ml, 6.53 mmol) is added and the reaction is stirred for further 2 hours.
  • N-[3-(morpholin-4-yl)propyl]-4-(4- ⁇ 4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2- yl ⁇ piperazin-l-yl)benzamide was prepapred in a manner analogous to that described above for N-(3-hydroxypropyl)-4-(4- ⁇ 4-oxo-3H,4H-tWeno[3,2-d]pyrimidin-2-yl ⁇ piperazin-l-yl)benzamide 33mg (11.2%) as a beige powder.
  • MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 483.2, Retention time 1.41.
  • N-[3-(dimethylamino)propyl]-4-(4- ⁇ 4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2- yl ⁇ piperazin-l-yl)benzamide was prepapred in a manner analogous to that described above for N-(3-hydroxypropyl)-4-(4- ⁇ 4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl ⁇ piperazin-l-yl)benzamide 18.1 mg (7.5%) as brown powder.
  • N,N-dimethyl-4-(4- ⁇ 4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl ⁇ piperazin-l- yl)benzamide was prepared in a manner analogous to that described above for N-methyl-4-(4- ⁇ 4- oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl ⁇ piperazin-l-yl)benzamide: 48.7 mg (56.9%) as white solid.
  • N-methyl-6-(4- ⁇ 4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl ⁇ piperazin-l- yl)pyridazine-3-carboxamide was prepared in a manner analogous to that described above for N- methyl-4-(4- ⁇ 4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yI ⁇ piperazin-l-yl)benzamide: 0.01 g (6.2%) as off white solid.
  • tert-butyl 4-(6-cyanopyridazin-3-yl)piperazine-l-carboxylate required for the synthesis was prepared as follows: 6-chloropyridazine-3-carbonitrile (600 mg, 4.3 mmol) and tert-butyl piperazine-l-carboxylate (1803.69 ⁇ , 10.75 mmol) were dissolved in MeCN (20ml) and the reaction is heated in a pressure tube to 60C for one hour. Solid had precipitated during reaction and LCMS indicated no remaining starting material. Reaction is cooled down to room temperature and the solid is filtered off. This compound is washed with water and left to dry. 1033 mg (83%) as orange powder. METCR1673 Generic 2 minutes M/Z (ES+) 233.95, Retention time 1.21.
  • a sealed tube was charged with 5-bromo-l-benzothiophene (147.55 ⁇ , 1.03 mmol), 4-(piperazin-l -yJ)benzoic acid (255.51 mg, 1.24 mmol), RuPhos (24.09 mg, 0.05 mmol), CS2CO3 (198.26 ⁇ , 2.48 mmol) and anhydrous THF (7 ml).
  • the reaction mixture was degassed by bubbling nitrogen with sonication for 30 min, prior to adding RuPhos Precatalyst (42.17 mg, 0.05 mmol) and the reaction mixture stirred at 70°C for 40 h. The mixture was allowed to cool to rt and the precipitate filtered and washes with water.
  • Example 85 4-methoxy-2-[4-(pyridin-2-yl)piperazin-l-yl]quinazoline (79) and 3-methyl-2- [4-(pyridin-2-yl)piperazin-l-yl]-3,4-dihydroquinazolin-4-one (80)
  • injection vials A solution of 100 g of an active ingredient according to the invention and 5 g of disodium hydrogen phosphate in 3 1 of bidistilled water is adjusted to pH 6.5 using 2 N hydrochloric acid, sterile filtered, transferred into injection vials, is lyophilized under sterile conditions and is sealed under sterile conditions. Each injection vial contains 5 mg of active ingredient.
  • (B) Suppositories A mixture of 20 g of an active ingredient according to the invention is melted with 100 g of soy lecithin and 1400 g of cocoa butter, is poured into moulds and is allowed to cool. Each suppository contains 20 mg of active ingredient.
  • Coated tablets Tablets are pressed analogously to Example E and subsequently are coated in a conventional manner with a coating of sucrose, potato starch, talc, tragacanth and dye.
  • Capsules 2 kg of an active ingredient according to the invention are introduced into hard gelatin capsules in a conventional manner in such a way that each capsule contains 20 mg of the active ingredient.
  • Ampoules A solution of 1 kg of an active ingredient according to the invention in 60 1 of bidistilled water is sterile filtered, transferred into ampoules, is lyophilized under sterile conditions and is sealed under sterile conditions. Each ampoule contains 10 mg of active ingredient.
  • Inhalation spray 14 g of an active ingredient according to the invention are dissolved in 10 1 of isotonic NaCl solution, and the solution is transferred into commercially available spray containers with a pump mechanism. The solution could be sprayed into the mouth or nose. One spray shot (about 0.1 ml) corresponds to a dose of about 0.14 mg.

Abstract

The present invention relates to compounds, and pharmaceutically acceptable compositions thereof, useful as ASIC inhibitors.

Description

COMPOUNDS AS ASIC INHIBITORS
AND USES THEREOF
TECHNICAL FIELD OF THE INVENTION
[0001] The present invention relates to compounds that are useful as inhibitors of acid- sensing ion channels (ASIC). The invention also provides pharmaceutically acceptable compositions comprising compounds of the present invention and methods of using said compositions in the treatment of various disorders.
BACKGROUND OF THE INVENTION
[0002] ASIC channels are proton-gated, voltage-insensitive cation channels activated by extracellular acidosis. ASICs belong to a superfamily which also contains degenerins (DEG) and epithelial sodium channels (ENaC) All members within the ENaC/DEG/ASIC superfamily share the same topology, with the N- and C-termini located inside the cell and a large cysteine-rich extracellular domain. In rodents, four genes encode at least six different ASIC subunits, ASICla, ASIC2a, ASIC2b, ASIC3 and ASIC4. ASICs function as trimers and conduct mostly Na+. Homomeric ASICla and human ASIClb, as well as ASICla 2b heteromers, also have a low permeability to Ca2+. The amino acid sequences of ASIC subunits are well conserved between species. For example, the mouse ASICla and the human ASICla share over 99% of their amino acid sequence identity.
[0003] ASICs are highly expressed in neurons, but are also present in non-neuronal tissues. ASICla and ASIC2a and b are widely expressed in the central nervous system, while almost all subunits are present in sensory neurons of the peripheral nervous system. In peripheral sensory neurons, ASICs have been found on cell bodies and sensory terminals, where they appear to be important for nociception and mechanosensation. In central neurons, ASICs have been found on cell bodies, dendrites and at dendritic spines and have been suggested to contribute to synaptic plasticity. ASICs are activated by extracellular acidosis and their activation induces neuronal depolarization, sometimes associated with direct and indirect Ca2+ entry through voltage-gated calcium channels. ASIC activation can be subject to modulation by extracellular alkalosis, intracellular pH, neuropeptides, polyamines, cations, arachidonic acid, lactate, nitric oxide, ATP, serotonin and exogenous modulators such as toxins from venoms, PcTx, APETx2, MitTx, and Mambalgin-1.
[0004] ASICs have been involved in several physiological processes such as nociception, mechanosensation, blood pressure regulation, synaptic function and plasticity. Growing evidence suggests an involvement of ASICs in anxiety and depression-related disorders. A common feature in neuropathological conditions is acidosis arising from ischemia, inflammation, metabolism or synaptic transmission. Acidosis kills neurons and ASICs have been involved in mediating acid-induced neurotoxicity in neurological disorders such as ischemic stroke, epilepsy, multiple sclerosis, Huntington's disease, Parkinson's disease and spinal cord injury. In addition many types of brain tumors upregulate ASIC expression, which suggests a possible role of ASICs in tumor pathophysiology.
SUMMARY OF THE INVENTION
[0005] It has now been found that compounds of this invention, and pharmaceutically acceptable compositions thereof, are effective as inhibitors of ASIC. Such compounds have general formula I:
Figure imgf000003_0001
I
or a pharmaceutically acceptable salt thereof, wherein each of Ring A, X, Y, Z, R1, R2, R3, m, and n, is as defined and described in embodiments herein.
[0006] Compounds of the present invention, and pharmaceutically acceptable compositions thereof, are useful for treating a variety of diseases, disorders or conditions, associated with ASIC. Such diseases, disorders, or conditions include those described herein.
DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS
1. General Description of Compounds of the Invention
[0007] In certain aspects, the present invention provides for inhibitors of ASIC. In certain aspects, the present invention provides for inhibitors of ASIC la. In some embodiments, such compounds include those of the formulae described herein, or a pharmaceutically acceptable salt thereof, wherein each variable is as defined and described herein.
2. Compounds and Definitions
[0008] Compounds of this invention include those described generally above, and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed. Additionally, general principles of organic chemistry are described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausalito: 1999, and "March's Advanced Organic Chemistry", 5th Ed., Ed.: Smith, M.B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.
[0009] The term "aliphatic" or "aliphatic group", as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as "carbocycle" "cycloaliphatic" or "cycloalkyl"), that has a single point of attachment to the rest of the molecule. Unless otherwise specified, aliphatic groups contain 1 -6 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms. In some embodiments, "cycloaliphatic" (or "carbocycle" or "cycloalkyl") refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule. Exemplary aliphatic groups are linear or branched, substituted or unsubstituted Ci-C8 alkyl, C2-C8 alkenyl, C2-C8 alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
[0010] The term "lower alkyl" refers to a Ci-4 straight or branched alkyl group. Exemplary lower alkyl groups are methyl, ethyl, propyl, isopropyl, butyl, isobutyl, and tert-butyl. [0011] The term "lower haloalkyl" refers to a Ci-4 straight or branched alkyl group that is substituted with one or more halogen atoms.
[0012] The term "heteroatom" means one or more of oxygen, sulfur, nitrogen, or phosphorus (including, any oxidized form of nitrogen, sulfur, or phosphorus; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4- dihydro-2H-pyrrolyl), ΝΗ (as in pyrrolidinyl) or NR+ (as in N-substituted pyrrolidinyl)).
[0013] The term "unsaturated", as used herein, means that a moiety has one or more units of unsaturation.
[0014] As used herein, the term "bivalent Ci-8 (or Ci-β) saturated or unsaturated, straight or branched, hydrocarbon chain", refers to bivalent alkylene, alkenylene, and alkynylene chains that are straight or branched as defined herein.
[0015] The term "alkylene" refers to a bivalent alkyl group. An "alkylene chain" is a polymethylene group, i.e., -(CH2)n- wherein n is a positive integer, preferably from 1 to 6, from
1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3. A substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
[0016] The term "alkenylene" refers to a bivalent alkenyl group. A substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
[0017] The term "halogen" means F, CI, Br, or I.
[0018] The term "aryl" used alone or as part of a larger moiety as in "aralkyl", "aralkoxy", or "aryloxyalkyl", refers to monocyclic and bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains three to seven ring members. The term "aryl" is used interchangeably with the term "aryl ring". In certain embodiments of the present invention, "aryl" refers to an aromatic ring system. Exemplary aryl groups are phenyl, biphenyl, naphthyl, anthracyl and the like, which optionally includes one or more substituents. Also included within the scope of the term "aryl", as it is used herein, is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like. [0019] The terms "heteroaryl" and "heteroar-", used alone or as part of a larger moiety, e.g., "heteroaralkyl", or "heteroaralkoxy", refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 π electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms. The term "heteroatom" refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen. Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl. The terms "heteroaryl" and "heteroar-", as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring. Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3-b]-l,4-oxazin- 3(4H)-one. A heteroaryl group is optionally mono- or bicyclic. The term "heteroaryl" is used interchangeably with the terms "heteroaryl ring", "heteroaryl group", or "heteroaromatic", any of which terms include rings that are optionally substituted. The term "heteroaralkyl" refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
[0020] As used herein, the terms "heterocycle", "heterocyclyl", "heterocyclic radical", and "heterocyclic ring" are used interchangeably and refer to a stable 5- to 7— membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above. When used in reference to a ring atom of a heterocycle, the term "nitrogen" includes a substituted nitrogen. As an example, in a saturated or partially unsaturated ring having 0-3 heteroatoms selected from oxygen, sulfur or nitrogen, the nitrogen is N (as in 3,4-dihydro- 2H-pyrrolyl), ΝΗ (as in pyrrolidinyl), or +NR (as in N-substituted pyrrolidinyl).
[0021] A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted. Examples of such saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl. The terms "heterocycle", "heterocyclyl", "heterocyclyl ring", "heterocyclic group", "heterocyclic moiety", and "heterocyclic radical", are used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H-indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl, where the radical or point of attachment is on the heterocyclyl ring. A heterocyclyl group is optionally mono- or bicyclic. The term "heterocyclylalkyl" refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
[0022] As used herein, the term "partially unsaturated" refers to a ring moiety that includes at least one double or triple bond. The term "partially unsaturated" is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
[0023] As described herein, certain compounds of the invention contain "optionally substituted" moieties. In general, the term "substituted", whether preceded by the term "optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. "Substituted" a lies to one or more h dro ens that are either
explicit or implicit from the
Figure imgf000007_0001
refers to at least
otherwise indicated, an "optionally substituted" group has a suitable substituent at each substitutable position of the group, and when more than one position in any given structure is substituted with more than one substituent selected from a specified group, the substituent is either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. The term "stable", as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
[0024] Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted" group are independently deuterium; halogen; -(CH2)o- R°; -(CH2)o- 0R°; -0(CH2)o- 4R°, -0-(CH2)o^C(0)OR°; -(CH2)o^CH(OR°)2; -(CH2)C SR0; -(CH2)o-4Ph, which are optionally substituted with R°; -(CH2)o-40(CH2)o-iPh which is optionally substituted with R°; - CH=CHPh, which is optionally substituted with R°; -(CH2)o-tO(CH2)o-]-pyridyl which is optionally substituted with R°; -N02; -CN; -N3; -(CH2)o-^N(R°)2; -(CH2)( N(R°)C(0)R°; - N(R°)C(S)R°; -(CH2)o^N(R°)C(0)NR°2; -N(R°)C(S)NR°2; -(CH2)0- N(R°)C(O)OR°; - N(R°)N(R°)C(0)R°; -N(R0)N(R°)C(0)NR0 2; -N(R°)N(R°)C(0)OR°; -(CH2)( C(0)R°; - C(S)R°; -(CH2)o-4C(0)OR°; -(CH2)o-4C(0)SR°; -(CH2)o-4C(0)OSiR°3; -<CH2)( OC(0)R°; - OC(0)(CH2)o-tSR°, SC(S)SR°; -(CH2)o-4SC(0)R°; -(CH2)o-4C(0)NR° ; -C(S)NR°2; -C(S)SR°; -SC(S)SR°, -(CH2)o-40C(0)NR°2; -C(O)N(0R°)Ro; -C(0)C(0)R°; -C(0)CH2C(0)R°; - C(NOR°)R°; -(CH2)o-4SSR°; -(CH2)o-tS(0)2R0; -(CH^SCCTfeOR0; -(CH2)0-4OS(O)2Ro; - S(0)2NR°2; -(CH2)o-)S(0)R0; -N(R°)S(0)22; -N(R°)S(0)2R0; -N(OR°)R°; -C(NH)NR°2; - P(0)2R0; -P(0)R°2; -OP(0)R°2; -OP(0)(OR°) ; SiR°3; -(C1-4 straight or branched alkylene)0- N(R°)2; or straight or branched alkylene)C(0)O-N(R°)2, wherein each R° is optionally substituted as defined below and is independently hydrogen, C1-5 aliphatic, -CH2Ph, -0(CH2)o~ lPh, -CH2-(5-6 membered heteroaryl ring), or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which is optionally substituted as defined below.
[0025] Suitable monovalent substituents on R° (or the ring formed by taking two independent occurrences of R° together with their intervening atoms), are independently deuterium, halogen, -(CH )o-2R*, -(haloR*), -(CH2)o-2OH, -(CH2)o- ORe, -(CH2)o-2CH(OR*) ; -0(haloRe), -CN, -N3, -(CH )o-2C(0)Re, -(CH )o-2C(0)OH, -(CH2)o-2C(0)OR*, -(CH2)o-2SR*, -(CH2)o-2SH, -(CH2)o-2NH2, -(CH2)o-2NHR', -(CH2)o-2NR*2, -N02, -SiR*3, -OSiR*3, -C(0)SR* -(Ci^t straight or branched alkylene)C(0)OR', or -SSR* wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently selected from Ci-4 aliphatic, -CH2Ph, -0(CH2)o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R° include =0 and =S.
[0026] Suitable divalent substituents on a saturated carbon atom of an "optionally substituted" group include the following: =0, =S, =NNR* 2, =NNHC(0)R*, =NNHC(0)OR*, =NNHS(0)2R*, =NR\ =NOR*, -0(C(R* 2))2_30-, or -S(C(R* 2))2_3S- wherein each independent occurrence of R* is selected from hydrogen, Ci-6 aliphatic which is substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an "optionally substituted" group include: -0(CR* 2)2-30-, wherein each independent occurrence of R* is selected from hydrogen, d-6 aliphatic which is optionally substituted as defined below, or an unsubstituted 5-6- membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0027] Suitable substituents on the aliphatic group of R* include halogen, -Re, -(haloR*), -OH, -OR*, -O(haloR'), -CN, -C(0)OH, -C(0)ORe, -NH2, -NHR*, -NR* , or -N02, wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci-4 aliphatic, -CH2Ph, -0(CH2)o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0028] Suitable substituents on a substitutable nitrogen of an "optionally substituted" group include -R, -NR 2, -C(0)R, -C(0)OR, -C(0)C(0)R, -C(0)CH2C(0)R+,
Figure imgf000009_0001
-S(0)2NR 2, -C(S)NR 2, -C(NH)NR 2, or
Figure imgf000009_0002
wherein each R is independently hydrogen, d-6 aliphatic which is optionally substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0^1 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R, taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. [0029] Suitable substituents on the aliphatic group of R are independently halogen, -Re, -(haloR*), -OH, -OR', -O(haloR'), -CN, -C(0)OH, -C(0)ORe, -NH2, -NHR*, -NR*2, or -N02, wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently C1-4 aliphatic, -CH2Ph, -0(CH2)o-iPh, or a 5-6- membered saturated, partially unsaturated, or aryl ring having 0—4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
[0030] In certain embodiments, the terms "optionally substituted", "optionally substituted alkyl," "optionally substituted "optionally substituted alkenyl," "optionally substituted alkynyl", "optionally substituted carbocyclic," "optionally substituted aryl", " optionally substituted heteroaryl," "optionally substituted heterocyclic," and any other optionally substituted group as used herein, refer to groups that are substituted or unsubstituted by independent replacement of one, two, or three or more of the hydrogen atoms thereon with typical substituents including, but not limited to:
-F, -CI, -Br, -I, deuterium,
-OH, protected hydroxy, alkoxy, oxo, thiooxo,
-N02, -CN, CF3, N3,
-NH2, protected amino, -NH alkyl, -NH alkenyl, -NH alkynyl, -NH cycloalkyl, -NH - aryl, -NH -heteroaryl, -NH -heterocyclic, -dialkylamino, -diarylamino, -diheteroarylamino,
-O- alkyl, -O- alkenyl, -O- alkynyl, -O- cycloalkyl, -O-aryl, -O-heteroaryl, -O- heterocyclic,
-C(O)- alkyl, -C(O)- alkenyl, -C(O)- alkynyl, -C(O)- carbocyclyl, -C(0 aryl, -C(O)- heteroaryl, -C(0)-heterocyclyI,
-CONH2, -CONH- alkyl, -CONH- alkenyl, -CONH- alkynyl, -CONH-carbocyclyl, - CONH-aryl, -CONH-heteroaryl, -CONH-heterocyclyl,
-OCO2- alkyl, -OC02- alkenyl, -OC02- alkynyl, -OC02- carbocyclyl, -OC02-aryl, - OC02-heteroaryl, -OC02-heterocyclyl, -OCONH2, -OCONH- alkyl, -OCONH- alkenyl, - OCONH- alkynyl, -OCONH- carbocyclyl, -OCONH- aryl, -OCONH- heteroaryl, -OCONH- heterocyclyl,
-NHC(O)- alkyl, -NHC(O)- alkenyl, -NHC(O)- alkynyl, -NHC(O)- carbocyclyl, - NHC(0>aryl, -NHC(0)-heteroaryl, -NHC(0>heterocyclyl, -NHC02- alkyl, -NHC02- alkenyl, - NHC02- alkynyl, -NHC02 - carbocyclyl, -NHC02- aryl, -NHC02- heteroaryl, -NHCO2- heterocyclyl, -NHC(0)NH2, -NHC(0)NH- alkyl, -NHC(0)NH- alkenyl, -NHC(0)NH- alkenyl, - NHC(0)NH- carbocyclyl, -NHC(0)NH-aryl, -NHC(0)NH-heteroaryI, -NHC(0)NH- heterocyclyl, NHC(S) H2, -NHC(S) H- alkyl, -NHC(S)NH- alkenyl, -NHC(S)NH- alkynyl, - NHC(S)NH- carbocyclyl, -NHC(S)NH-aryl, -NHC(S)NH-heteroaryl, - HC(S) H-heterocyclyl, -NHC(NH)NH2, -NHC(NH)NH- alkyl, -NHC(NH)NH- -alkenyl, -NHC(NH)NH- alkenyl, - NHC(NH)NH- carbocyclyl, -NHC(NH)NH-aryl, -NHC(NH)NH-heteroaryl, -NHC(NH)NH- heterocyclyl, -NHC(NH)- alkyl, -NHC( H)- alkenyl, -NHC(NH)- alkenyl, -NHC(NH)- carbocyclyl, -NHC(NH)-aryl, -NHC(NH)-heteroaryl, -NHC(NH>heterocyclyl,
-C(NH)NH- alkyl, -C(NH) H- alkenyl, -C(NH)NH- alkynyl, -C(NH)NH- carbocyclyl, - C(NH) H-aryl, -C(NH)NH-heteroaryl, -C(NH)NH-heterocyclyl,
-S(O)- alkyl, - S(O)- alkenyl, - S(O)- alkynyl, - S(0)- carbocyclyl, - S(0)-aryl, - S(O)- heteroaryl, - S(0)-heterocyclyl -S02NH2, -S02 H- alkyl, -S02NH- alkenyl, -S02NH- alkynyl, - S02NH- carbocyclyl, -S02 H- aryl, -S02NH- heteroaryl, -S02 H- heterocyclyl,
-NHS02- alkyl, -NHS02- alkenyl, - NHS02- alkynyl, -NHS02- carbocyclyl, -NHS02- aryl, -NHS02-heteroaryl, -NHS02-heterocyclyl,
-CH2NH2, -CH2S02CH3,
-mono-, di-, or tri-alkyl silyl,
-alkyl, -alkenyl, -alkynyl, -aryl, -arylalkyl, -heteroaryl, -heteroarylalkyl, heterocycloalkyl, -cycloalkyl, -carbocyclic, -heterocyclic, polyalkoxyalkyl, polyalkoxy, - methoxymethoxy, -methoxyethoxy, -SH, -S- alkyl, -S- alkenyl, -S- alkynyl, -S- carbocyclyl, -S- aryl, -S-heteroaryl, -S-heterocyclyl, or methylthiomethyl.
[0031] Divalent groups include each group in either directions. For example, the group "- S02NH-" in between group X and group Y, includes both X-S02 H-Y and Y-S02NH-X.
[0032] As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like.
[0033] Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(C1^alkyl)4 salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
[0034] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
[0035] As used herein, the term "tautomer" means each of two or more isomers of a compound that exist together in equilibrium, and are readily interchanged by migration of an atom or group within the molecule. Tautomers are constitutional isomers of organic compounds that readily interconvert by a chemical reaction called tautomerizafion. This reaction commonly results in the formal migration of a hydrogen atom or proton, accompanied by a switch of a single bond and adjacent double bond (e.g., and in no way limited to this example, compounds of the followin tautomers are contemplated by the invention,
Figure imgf000013_0001
[0036] In certain instances, each form can be isolated and characterized.
[0037] Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
For example, compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention. In some embodiments, the group comprises one or more deuterium atoms.
[0038] There is furthermore intended that a compound of the formula I includes isotope- labeled forms thereof. An isotope-labeled form of a compound of the formula I is identical to this compound apart from the fact that one or more atoms of the compound have been replaced by an atom or atoms having an atomic mass or mass number which differs from the atomic mass or mass number of the atom which usually occurs naturally. Examples of isotopes which are readily commercially available and which can be incorporated into a compound of the formula I by well- known methods include isotopes of hydrogen, carbon, nitrogen, oxygen, phos-phorus, fluo-rine and chlorine, for example 2H, 3H, 13C, 14C, 15N, 180, O, 31P, 2P, 35S, 18F and 36CI, respectively. A compound of the formula I, a prodrug, thereof or a pharmaceutically acceptable salt of either which contains one or more of the above-mentioned isotopes and/or other isotopes of other atoms is intended to be part of the present invention. An isotope-labeled compound of the formula I can be used in a number of beneficial ways. For example, an isotope-labeled compound of the formula I into which, for example, a radioisotope, such as 3H or 14C, has been incorporated, is suitable for medicament and/or substrate tissue distribution assays. These radioisotopes, i.e. tritium (3H) and carbon-14 (14C), are particularly preferred owing to simple preparation and excellent detectability. Incorporation of heavier isotopes, for example deuterium (2H), into a compound of the formula I has therapeutic advantages owing to the higher metabolic stability of this isotope-labeled compound. Higher metabolic stability translates directly into an increased in vivo half-life or lower dosages, which under most circumstances would represent a preferred embodiment of the present invention. An isotope-labeled compound of the formula I can usually be prepared by carrying out the procedures disclosed in the synthesis schemes and the related description, in the example part and in the preparation part in the present text, replacing a non-isotope-labeled reactant by a readily available isotope-labeled reactant.
[0039] Deuterium (2H) can also be incorporated into a compound of the formula I for the purpose in order to manipulate the oxidative metabolism of the compound by way of the primary kinetic isotope effect. The primary kinetic isotope effect is a change of the rate for a chemical reaction that results from exchange of isotopic nuclei, which in turn is caused by the change in ground state energies necessary for covalent bond formation after this isotopic exchange. Exchange of a heavier isotope usually results in a lowering of the ground state energy for a chemical bond and thus causes a reduction in the rate in rate-limiting bond breakage. If the bond breakage occurs in or in the vicinity of a saddle-point region along the coordinate of a multi- product reaction, the product distribution ratios can be altered substantially. For explanation: if deuterium is bonded to a carbon atom at a non-exchangeable position, rate differences of kM¾> = 2-7 are typical. If this rate difference is successfully applied to a corn-pound of the formula I that is susceptible to oxidation, the profile of this compound in vivo can be drastically modified and result in improved pharmacokinetic properties.
[0040] When discovering and developing therapeutic agents, the person skilled in the art is able to optimize pharmacokinetic parameters while retaining desirable in vitro properties. It is reasonable to assume that many compounds with poor pharmacokinetic profiles are susceptible to oxidative metabolism. In vitro liver microsomal assays currently available provide valuable information on the course of oxidative metabolism of this type, which in turn permits the rational design of deuterated compounds of the formula I with improved stability through resistance to such oxidative metabolism. Significant improvements in the pharmacokinetic profiles of compounds of the formula I are thereby obtained, and can be expressed quantitatively in terms of increases in the in vivo half-life (t/2), concentration at maximum therapeutic effect (Cmax), area under the dose response curve (AUC), and F; and in terms of reduced clearance, dose and materials costs. [0041] The following is intended to illustrate the above: a compound of the formula I which has multiple potential sites of attack for oxidative metabolism, for example benzylic hydrogen atoms and hydrogen atoms bonded to a nitrogen atom, is prepared as a series of analogues in which various combinations of hydrogen atoms are replaced by deuterium atoms, so that some, most or all of these hydrogen atoms have been replaced by deuterium atoms. Half-life determinations enable favorable and accurate determination of the extent of the extent to which the improvement in resistance to oxidative metabolism has improved. In this way, it is determined that the half-life of the parent compound can be extended by up to 100% as the result of deuterium-hydrogen exchange of this type.
[0042] Deuterium-hydrogen exchange in a compound of the formula I can also be used to achieve a favorable modification of the metabolite spectrum of the starting compound in order to diminish or eliminate undesired toxic metabolites. For example, if a toxic metabolite arises through oxidative carbon-hydrogen (C-H) bond cleavage, it can reasonably be assumed that the deuterated analogue will greatly diminish or eliminate production of the unwanted metabolite, even if the particular oxidation is not a rate-determining step. Further information on the state of the art with respect to deuterium-hydrogen exchange may be found, for example in Hanzlik et al., J. Org. Chem. 55, 3992-3997, 1990, Reider et al., J. Org. Chem. 52, 3326-3334, 1987, Foster, Adv. Drug Res. 14, 1-40, 1985, Gillette et al, Biochemistry 33(10) 2927-2937, 1994, and Jarman et al. Carcinogenesis 16(4), 683-688, 1993.
[0043] As used herein, the term "modulator" is defined as a compound that binds to and /or inhibits the target with measurable affinity. In certain embodiments, a modulator has an IC5o and/or binding constant of less about 50 μΜ, less than about 5 μΜ, less than about 1 μΜ, less than about 500 nM, less than about 100 nM, or less than about 10 nM.
[0044] The terms "measurable affinity" and "measurably inhibit," as used herein, means a measurable change in ASIC activity between a sample comprising a compound of the present invention, or composition thereof, and ASIC, and an equivalent sample comprising ASIC, in the absence of said compound, or composition thereof.
[0045] Combinations of substituents and variables envisioned by this invention are only those that result in the formation of stable compounds. The term "stable", as used herein, refers to compounds which possess stability sufficient to allow manufacture and which maintains the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., therapeutic or prophylactic administration to a subject).
[0046] The recitation of a listing of chemical groups in any definition of a variable herein includes definitions of that variable as any single group or combination of listed groups. The recitation of an embodiment for a variable herein includes that embodiment as any single embodiment or in combination with any other embodiments or portions thereof.
3. Description of Exemplary Compounds
[0047] According to one aspect the present invention provides a compound of formula I,
Figure imgf000016_0001
I
or a tautomer, or a pharmaceutically acceptable salt thereof, wherein:
X is C(R4)2, C(R4), N, N-R4, O, S, SO, or S02;
Y is C(R4), C-O-R4, or C=0; each 1 is a single or double bond, as allowed by valence, wherein at least one of Ί is a double bond;
Ring A is a fused C5-6 aryl, or a fused 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R1 is independently -R, halogen, -OR, -SR, -CN, -N02, -S02R, -SOR, -C(0)R, -C02R,
-C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRSO2R, or -N(R>2;
Z is C(R4) or N;
each R2 is independently -R, halogen, -OR, -SR, -CN, -NO2, -S02R, -SOR, -C(0)R, -C02R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRSO2R, or -N(R)2; two R2 groups on the same atom are taken together with the atom to which they are attached to form a carbonyl, thiocarbonyl, or imine; each of which is optionally substituted;
R3 is -R, halogen, -OR, -SR, -CN, -NO2, -S02R, -SOR, -C(0)R, -C02R, -C(0)N(R)2, - NRC(0)R, -NRC(0)N(R)2, -NRS02R, or -N(R ;
each R4 is independently hydrogen, Ci-6 aliphatic, C5_io aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted;
each R is independently hydrogen, Ci-e aliphatic, C5-10 aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted; or
two R groups on the same atom are taken together with the atom to which they are attached to form a C5-10 aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted;
m is 0, 1, or 2; and
n is 0, 1, 2, 3, or 4.
[0048] In certain embodiments, X is C(R4)2, C(R4), N, N-R4, O, or S.
[0049] In certain embodiments, X is CH2, CH, N, NH, N-CH3, or S.
[0050] In certain embodiments, X is CH2. In certain embodiments, X is CH. In certain embodiments, X is N. In certain embodiments, X is NH. In certain embodiments, X is N-CH3. In certain embodiments, X is S.
[0051] In certain embodiments, Y is C(R4), C-O-R4, or C=0.
[0052] In certain embodiments, Y is CH, C-OH, C-0-CH3, C-0-/-Pr, or C=0.
[0053] In certain embodiments, Y is CH. In certain embodiments, Y is C-OH. In certain embodiments, Y is C-0-CH3. In certain embodiments, Y is C-O-z'-Pr. In certain embodiments, Y is C=0.
[0054] In certain embodiments, Ring A is fused phenyl, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl;- l,2,5oxadiazolyl,
1.3.4- oxadiazolyl, oxazolyl, pyrimidinyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, thiazolyl, thienyl, thiophenyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl,
1.2.5- triazolyl, or 1,3,4-triazolyl. [0055] In certain embodiments, Ring A is a fused phenyl, pyridinyl, thiophenyl, or furanyl.
[0056] In certain embodiments, Ring A is
Figure imgf000018_0001
[0057] In certain embodiments, Ring A is
Figure imgf000018_0002
[0058] In certain embodments, each R1 is independently -R, halogen, -OR, -SR, -CN, -N02, -SO2R, -SOR, -C(0)R, -C02R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R>2, -NRSO2R, or -N(R>2.
[0059] In certain embodments, each R1 is H.
[0060] In certain embodments, each R1 is independently Ci-6 aliphatic, Cs-io aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted, or each R1 is independently halogen, -OR, -SR, -CN, -N02, -SO2R, -SOR, -C(0)R, -C02R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRS02R, or -N(R>2.
[0061] In certain embodments, each R1 is independently methyl, ethyl, propyl, i-propyl, n-
Bu, s-Bu, t-Bu, straight chain or branched pentyl, or straight chain or branched hexyl; each of which is optionally substituted.
[0062] In certain embodiments, Z is C(R4).
[0063] In certain embodiments, Z is CH.
[0064] In certain embodiments, Z is N.
[0065] In certain embodiments, R2 is H.
[0066] In certain embodiments, R2 is C1-0 aliphatic, C5-10 aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted; or R3 is halogen, -OR, -SR, -CN, -NO2, -S02R, -SOR,
-C(0)R, -C02R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRSO2R, or -N(R)2.
[0067] In certain embodiments, R2 is methyl, ethyl, propyl, i-propyl, n-butyl, s-butyl, t-butyl, straight chain or branched pentyl, or straight chain or branched hexyl, each of which is optionally substituted.
[0068] In certain embodiments, R2 is phenyl, naphthyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantyl, cyclooctyl, [3.3.0]bicyclooctanyl, [4.3.0]bicyclononanyl, [4.4.0]bicyclodecanyl, [2.2.2]bicyclooctanyl, fluorenyl, indanyl, tetrahydronaphthyl, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, NH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H6H-l,5,2-dithiazinyl, dihydrofuro [2,3-6] tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, lH-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isoindolinyl, isoindolenyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, morpholinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl;- 1 ,2,5oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, 6H-l,2,5-thiadiazinyl, 1,2,3- thiadiazolyl, 1,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, l,3,4thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1 ,2,5-triazolyl, 1,3,4-triazolyl, oxetanyl, azetidinyl, or xanthenyl; each of which is optionally substituted.
[0069] In certain embodiments, R2 is halogen, -OR, -SR, -CN, -N02, -SO2R, -SOR, -C(0)R, -C02R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRSO2R, or -N(R)2.
[0070] In certain embodiments, each R2 is indepenently -Η or -Me, or two R2 groups on the same atom are C=0. [0071] In certain embodiments, R3 is -R, halogen, -OR, -SR, -CN, -N02, -S02R, -SOR, -C(0)R, -C02R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRS02R, or -N(R)2.
[0072] In certain embodiments, R3 is H.
[0073] In certain embodiments, R3 is Ci-6 aliphatic, Cs-io aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted; or R3 is halogen, -OR, -SR, -CN,— NO2, -S02R, -SOR, -C(0)R, -CO2R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRSO2R, or -N(R)2.
[0074] In certain embodiments, R3 is methyl, ethyl, propyl, i-propyl, n-butyl, s-butyl, t-butyl, straight chain or branched pentyl, or straight chain or branched hexyl, each of which is optionally substituted.
[0075] In certain embodiments, R3 is phenyl, naphthyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, adamantyl, cyclooctyl, [3.3.0]bicyclooctanyl, [4.3.0]bicyclononanyl, [4.4.0]bicyclodecanyl, [2.2.2]bicyclooctanyl, fluorenyl, indanyl, tetrahydronaphthyl, acridinyl, azocinyl, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzthiazolyl, benztriazolyl, benztetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, NH-carbazolyl, carbolinyl, chromanyl, chromenyl, cinnolinyl, decahydroquinolinyl, 2H,6H-l,5,2-dithiazinyl, dihydrofuro [2,3-6] tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, lH-indazolyl, indolenyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isoindolinyl, isoindolenyl, isobenzofuranyl, isochromanyl, isoindazolyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiazolyl, isoxazolyl, mo holinyl, naphthyridinyl, octahydroisoquinolinyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl;- 1 ,2,5oxadiazolyl, 1,3,4-oxadiazolyl, oxazolidinyl, oxazolyl, oxazolidinyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathiinyl, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridinyl, pyridyl, pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, 6H-l,2,5-thiadiazinyl, 1,2,3-thiadiazolyl, 1 ,2,4-thiadiazolyl, 1,2,5-thiadiazolyl, l,3,4thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thiophenyl, triazinyl, 1,2,3-triazolyl, 1 ,2,4-triazolyl, 1,2,5-triazolyl, 1,3,4-triazolyl, oxetanyl, azetidinyl, or xanthenyl; each of which is optionally substituted.
[0076] In certain embodiments, R3 is phenyl, cyclohexyl, imidazolyl, pyrazinyl, pyridazinyl, pyridinyl, pyridyl, pyrimidinyl, tetrahydrofuranyl, or tetrahydropyranyl; each of which is optionally substituted.
[0077] In certain embodiments, R3 is halogen, -OR, -SR, -CN, -N02, -S02R, -SOR, -C(0)R, -C02R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRS02R, or -NCR^.
[0078] In certain embodiments, R3 is -C(0)R, -C02R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRS02R, or -N(R>2.
[0079] In certain embodiments, R3 is methyl, ethyl, propyl, phenyl, cyclohexyl, imidazolyl, pyrazinyl, pyridazinyl, pyridinyl, pyridyl, pyrimidinyl, tetrahydrofuranyl, or tetrahydropyranyl; each of which is optionally substituted; or R3 is -C(0)R, -C02R, -C(0)N(R , -NRC(0)R, - RC(0)N(R)2, -NRS02R, or -NCR^.
Figure imgf000021_0001
Figure imgf000022_0001
[0081] In certain embodiments, R4 is H.
[0082] In certain embodiments, R4 is d-6 aliphatic, C5-10 aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1 -4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted, or halogen.
[0083] In certain embodiments, each of X, Y, Z, Ring A, R, R1, R2, R3, R4, m, and n, is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination. [0084] In certain embodiments, the present invention provides a compound of formula I-a,
Figure imgf000023_0001
l a;
[0085] or a pharmaceutically acceptable salt thereof, wherein each of Z, R, R1, R2, R3, m, and n, is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0086] In certain embodiments, the present invention provides a compound of formula I-b,
Figure imgf000023_0002
l b;
or a pharmaceutically acceptable salt thereof, wherein R3 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0087] In certain embodiments, the present invention provides a compound of formula I-c,
Figure imgf000023_0003
I-c;
or a pharmaceutically acceptable salt thereof, wherein each of R3 and R4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0088] In certain embodiments, the present invention provides a compound of formula I-d,
Figure imgf000024_0001
or a pharmaceutically acceptable salt thereof, wherein each of R3 and R4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0089] In certain embodiments, the present invention provides a compound of formula I-e,
Figure imgf000024_0002
I-e;
or a pharmaceutically acceptable salt thereof, wherein each of R3 and R4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0090] In certain embodiments, the present invention provides a compound of formula I-f,
Figure imgf000024_0003
or a pharmaceutically acceptable salt thereof, wherein R3 is as defined above and described embodiments, classes and subclasses above and herein, singly or in combination.
[0091] In certain embodiments, the present invention provides a compound of formula I-g,
Figure imgf000024_0004
i-g;
or a pharmaceutically acceptable salt thereof, wherein each of R3 and R4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0092] In certain embodiments, the present invention provides a compound of formula I-h,
Figure imgf000025_0001
I-h;
or a pharmaceutically acceptable salt thereof, wherein R3 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0093] In certain embodiments, the present invention provides a compound of formula I-j,
Figure imgf000025_0002
i-j;
or a pharmaceutically acceptable salt thereof, wherein each of R3 and R4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0094] In certain embodiments, the present invention provides a compound of formula I-k,
Figure imgf000025_0003
I-k;
or a pharmaceutically acceptable salt thereof, wherein each of R3 and R4 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0095] In certain embodiments, the present invention provides a compound of formula 1-1,
Figure imgf000026_0001
i-i;
or a pharmaceutically acceptable salt thereof, wherein R3 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0096] In certain embodiments, the present invention provides a compound of formula I-m,
Figure imgf000026_0002
I-m;
or a pharmaceutically acceptable salt thereof, wherein R3 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0097] In certain embodiments, the present invention provides a compound of formula I-n,
Figure imgf000026_0003
I n;
or a pharmaceutically acceptable salt thereof, wherein R3 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination.
[0098] In certain embodiments the resent invention provides a compound of formula I-o,
Figure imgf000026_0004
I-o;
or a pharmaceutically acceptable salt thereof, wherein R3 is as defined above and described in embodiments, classes and subclasses above and herein, singly or in combination. [0099] In certain embodiments, the invention provides a compound selected from Table 1 :
Table 1
Figure imgf000027_0001
10 11 12
Figure imgf000028_0001

Figure imgf000029_0001
Figure imgf000030_0001

Figure imgf000031_0001
Figure imgf000032_0001
31
Figure imgf000033_0001
32
Figure imgf000034_0001
Figure imgf000034_0002
Figure imgf000034_0003
84 85.
[00100] In some embodiments, the present invention provides a compound selected from those depicted above, or a pharmaceutically acceptable salt thereof.
[00101] Various structural depictions may show a heteroatom without an attached group, radical, charge, or counterion. Those of ordinary skill in the art are aware that such depictions are meant to indicate that the heteroatom is attached to hydrogen (e.g., ¾ is understood to be
V0H ).
[00102] In certain embodiments, the compounds of the invention were synthesized in accordance with the schemes provided in the Examples below.
4, Uses, Formulation and Administration Pharmaceutically Acceptable Compositions
[00103] According to another embodiment, the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The amount of compound in compositions of this invention is such that is effective to measurably inhibit ASIC, or a mutant thereof, in a biological sample or in a patient. In certain embodiments, the amount of compound in compositions of this invention is such that is effective to measurably inhibit ASIC, or a mutant thereof, in a biological sample or in a patient. In certain embodiments, a composition of this invention is formulated for administration to a patient in need of such composition.
[00104] The term "patient" or "subject", as used herein, means an animal, preferably a mammal, and most preferably a human.
[00105] The term "pharmaceutically acceptable carrier, adjuvant, or vehicle" refers to a nontoxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated. Pharmaceutically acceptable carriers, adjuvants or vehicles that are used in the compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene-block polymers, polyethylene glycol and wool fat.
[00106] A "pharmaceutically acceptable derivative" means any non-toxic salt, ester, salt of an ester or other derivative of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.
[00107] Compositions of the present invention are administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intraarticular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention include aqueous or oleaginous suspension. These suspensions are formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally acceptable diluent or solvent, for example as a solution in 1 ,3-butanediol. Among the acceptable vehicles and solvents that are employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
[00108] For this purpose, any bland fixed oil employed includes synthetic mono- or di- glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms are also be used for the purposes of formulation.
[001091 Pharmaceutically acceptable compositions of this invention are orally administered in any orally acceptable dosage form. Exemplary oral dosage forms are capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents are optionally also added.
[00110] Alternatively, pharmaceutically acceptable compositions of this invention are administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols. [00111] Pharmaceutically acceptable compositions of this invention are also administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
[00112] Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches are also used.
[00113] For topical applications, provided pharmaceutically acceptable compositions are formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Exemplary carriers for topical administration of compounds of this aremineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, provided pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
[00114] Pharmaceutically acceptable compositions of this invention are optionally administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and are prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
[00115] Most preferably, pharmaceutically acceptable compositions of this invention are formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions of this invention are administered without food. In other embodiments, pharmaceutically acceptable compositions of this invention are administered with food.
[00116] The amount of compounds of the present invention that are optionally combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration. Preferably, provided compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the compound can be administered to a patient receiving these compositions.
[00117] It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
Uses of Compounds and Pharmaceutically Acceptable Compositions
[00118] In certain embodiments, the invention provides a method for inhibiting or antagonizing ASIC in a patient or in a biological sample comprising the step of administering to said patient or contacting said biological sample with a compound according to the invention.
[00119] In certain embodiments, the invention is directed to the use of compounds of the invention and/or physiologically acceptable salts thereof, for modulating or inhibiting/antagonizing ASIC. The term "modulation" denotes any change in ASIC-mediated signal transduction, which is based on the action of the specific inventive compounds capable to interact with the ASIC target in such a manner that makes recognition, binding and activating possible. The compounds are characterized by such a high affinity to ASIC. In certain embodiments, the substances are highly selective for ASIC over most other channels in order to guarantee an exclusive and directed recognition with the single ASIC target. In the context of the present invention, the term "recognition" - without being limited thereto - relates to any type of interaction between the specific compounds and the target, particularly covalent or non- covalent binding or association, such as a covalent bond, hydrophobic/ hydrophilic interactions, van der Waals forces, ion pairs, hydrogen bonds, ligand-receptor (enzyme-inhibitor) interactions, and the like. Such association may also encompass the presence of other molecules such as peptides, proteins or nucleotide sequences. The present ion channel interaction is characterized by high affinity, high selectivity and minimal or even lacking cross-reactivity to other target molecules to exclude unhealthy and harmful impacts to the treated subject.
[00120] In certain embodiments, the present invention relates to a method for inhibiting or antagonizing ASIC, with at least one compound of formula (I) according to the invention and/or physiologically acceptable salts thereof, under conditions such that said ASIC is inhibited/antagonozied. In certain embodiments, the system is a cellular system. The cellular system is defined to be any subject provided that the subject comprises cells. Hence, the cellular system can be selected from the group of single cells, cell cultures, tissues, organs and animals. In certain embodiments, the method for modulating ASIC is performed in-vitro. The prior teaching of the present specification concerning the compounds of formula (I), including any embodiments thereof, is valid and applicable without restrictions to the compounds according to formula (I) and their salts when used in the method for inhibiting/antagonizing ASIC. The prior teaching of the present specification concerning the compounds of formula (I), including any embodiments thereof, is valid and applicable without restrictions to the compounds according to formula (I) and their salts when used in the method for inhibiting/antagonizing ASIC.
[00121] Provided compounds are inhibitors/antagonists of ASIC and are therefore useful for treating one or more disorders associated with activity of ASIC. Thus, in some embodiments, the present invention provides a method for treating an ASIC-mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.
[00122] In yet another aspect, a method for the treatment or lessening the severity of acute, chronic, neuropathic, or inflammatory pain, arthritis, migrane, cluster headaches, trigeminal neuralgia, herpetic neuralgia, general neuralgias, epilepsy or epilepsy conditions, neurodegenerative disorders, psychiatric disorders such as anxiety and depression, myotonia, arrhythmia, movement disorders, neuroendocrine disorders, ataxia, multiple sclerosis, irritable bowel syndrome, incontinence, visceral pain, osteoarthritis pain, postherpetic neuralgia, diabetic neuropathy, radicular pain, sciatica, back pain, head or neck pain, severe or intractable pain, nociceptive pain, breakthrough pain, postsurgical pain, or cancer pain is provided comprising administering an effective amount of a compound, or a pharmaceutically acceptable composition comprising a compound to a subject in need thereof. In certain embodiments, a method for the treatment or lessening the severity of acute, chronic, neuropathic, or inflammatory pain is provided comprising administering an effective amount of a compound or a pharmaceutically acceptable composition to a subject in need thereof. In certain other embodiments, a method for the treatment or lessening the severity of radicular pain, sciatica, back pain, head pain, or neck pain is provided comprising administering an effective amount of a compound or a pharmaceutically acceptable composition to a subject in need thereof. In still other embodiments, a method for the treatment or lessening the severity of severe or intractable pain, acute pain, postsurgical pain, back pain, tinnitis or cancer pain is provided comprising administering an effective amount of a compound or a pharmaceutically acceptable composition to a subject in need thereof.
[00123] The compounds of the present invention are useful in the prophylaxis and treatment of autoimmune and/or inflammatory disorders, including neurodegenerative diseases, such as multiple sclerosis (MS), polyneuritis, multiple neuritis, amyotrophic lateral sclerosis (ALS), Alzheimer's disease, optic neuritis, or Parkinson's disease.
[00124] The present invention furthermore relates to a method of treating a subject suffering from an immunerogulatory abnomality, comprising administering to said subject a compound of formula I in an amount that is effective for treating said immunoregulatory abnormality. The present invention preferably relates to a method wherein the immunoregulatory abnormality is an autoimmune or chronic inflammatory disease selected from the group consisting of: amyotrophic lateral sclerosis (ALS), Alzheimer's disease, Parkinson's disease, systemic lupus erythematosus, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cirrhosis, uveitis, multiple sclerosis, Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, psoriasis, autoimmune myositis, Wegener's granulomatosis, ichthyosis, Graves' ophthalmopathy and asthma. The present invention furthermore relates to a method wherein the immunoregulatory abnormality is bone marrow or organ transplant rejection or graft-versus-host disease. The present invention furthermore relates to a method wherein the immunoregulatory abnormality is selected from the group consisting of: transplantation of organs or tissue, graft- versus-host diseases brought about by transplantation, autoimmune syndromes including rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, posterior uveitis, allergic encephalomyelitis, glomerulonephritis, post-infectious autoimmune diseases including rheumatic fever and postinfectious glomerulonephritis, inflammatory and hyperproliferative skin diseases, psoriasis, atopic dermatitis, contact dermatitis, eczematous dermatitis, seborrhoeic dermatitis, lichen planus, pemphigus, bullous pemphigoid, epidermolysis bullosa, urticaria, angioedemas, vasculitis, erythema, cutaneous eosinophilia, lupus erythematosus, acne, alopecia areata, keratoconjunctivitis, vernal conjunctivitis, uveitis associated with Behcet's disease, keratitis, herpetic keratitis, conical cornea, dystrophia epithelialis corneae, corneal leukoma, ocular pemphigus, 5 Mooren's ulcer, scleritis, Graves' opthalmopathy, Vogt-Koyanagi-Harada syndrome, sarcoidosis, pollen allergies, reversible obstructive airway disease, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, dust asthma, chronic or inveterate asthma, late asthma and airway hyper-responsiveness, bronchitis, gastric ulcers, vascular damage caused by ischemic diseases and thrombosis, ischemic bowel diseases, inflammatory bowel diseases, necrotizing enterocolitis, intestinal lesions associated with thermal burns, coeliac diseases, proctitis, eosinophilic gastroenteritis, mastocytosis, Crohn's disease, ulcerative colitis, migraine, rhinitis, eczema, interstitial nephritis, Goodpasture's syndrome, hemolytic-uremic syndrome, diabetic nephropathy, multiple myositis, Guillain-Barre syndrome, Meniere's disease, polyneuritis, multiple neuritis, mononeuritis, radiculopathy, hyperthyroidism, Basedow's disease, pure red cell aplasia, aplastic anemia, hypoplastic anemia, idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, agranulocytosis, pernicious anemia, megaloblastic anemia, anerythroplasia, osteoporosis, sarcoidosis, fibroid lung, idiopathic interstitial pneumonia, dermatomyositis, leukoderma vulgaris, ichthyosis vulgaris, photoallergy sensitivity, cutaneous T cell lymphoma, chronic lymphocytic leukemia, arteriosclerosis, atherosclerosis, aortitis syndrome, polyarteritis nodosa, myocardosis, scleroderma, Wegener's granuloma, Sjogren's syndrome, adiposis, eosinophilic fascitis, lesions of gingiva, periodontium, alveolar bone, substantia ossea dentis, glomerulonephritis, male pattern alopecia or alopecia senilis by preventing epilation or providing hair germination and/or promoting hair generation and hair growth, muscular dystrophy, pyoderma and Sezary's syndrome, Addison's disease, ischemiareperfusion injury of organs which occurs upon preservation, transplantation or ischemic disease, endotoxin-shock, pseudomembranous colitis, colitis caused by drug or radiation, ischemic acute renal insufficiency, chronic renal insufficiency, toxinosis caused by lung-oxygen or drugs, lung cancer, pulmonary emphysema, cataracta, siderosis, retinitis pigmentosa, senile macular degeneration, vitreal scarring, corneal alkali burn, dermatitis erythema multiforme, linear IgA ballous dermatitis and cement dermatitis, gingivitis, periodontitis, sepsis, pancreatitis, diseases caused by environmental pollution, aging, carcinogenesis, metastasis of carcinoma and hypobaropathy, disease caused by histamine or leukotriene-C4 release, Behcet's disease, autoimmune hepatitis, primary biliary cirrhosis, sclerosing cholangitis, partial liver resection, 35 acute liver necrosis, necrosis caused by toxin, viral hepatitis, shock, or anoxia, B-virus hepatitis, non-A/non-B hepatitis, cirrhosis, alcoholic cirrhosis, hepatic failure, fulminant hepatic failure, late-onset hepatic failure, "acute-on-chronic" liver failure, augmentation of chemotherapeutic effect, cytomegalovirus infection, HCMV infection, AIDS, cancer, senile dementia, trauma, and chronic bacterial infection.
[00125] In certain embodiments, the disorder or disease is anxiety.
[00126] In certain embodiments, the disorder or disease is MS.
[00127] In certain embodiments, the disorder or disease is optic neuritis.
[00128] In certain embodiments, the disorder or disease is depression-related disorders
[00129] In certain embodiments, the disorder or disease is acidosis. In certain embodiments, the disorder or disease is acidosis arising from ischemia, inflammation, metabolism or synaptic transmission.
[00130] In certain embodiments, the disorder or disease is ischemic stroke, epilepsy, multiple sclerosis, Huntington's disease, Parkinson's disease or spinal cord injury.
[00131] In certain embodiments, the disorder or disease is cancer.
[00132] In certain embodiments, the disorder or disease is brain cancer or brain tumor.
[00133] In certain embodiments of the present invention an "effective amount" of the compound or pharmaceutically acceptable composition is that amount effective for treating or lessening the severity of a disease or disorder provide supra.
[00134] The compounds and compositions, according to the method of the present invention, may be administered using any amount and any route of administration effective for treating or lessening the severity of a disease or disorder provide supra.
[00135] The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. The compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression "dosage unit form", as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts. The term "patient", as used herein, means an animal, preferably a mammal, and most preferably a human.
[00136] As described generally above, the compounds of the invention are useful as inhibitors of voltage-gated ion channels. In one embodiment, the compounds and compositions of the invention are inhibitors of one or more of ASIC la, ASIC2a, ASIC2b, ASIC3 or ASIC4, and thus, without wishing to be bound by any particular theory, the compounds and compositions are particularly useful for treating or lessening the severity of a disease, condition, or disorder where activation or hyperactivity of one or more of ASIC la, ASIC2a, ASIC2b, ASIC3 and ASIC4 is implicated in the disease, condition, or disorder. When activation or hyperactivity of ASIC la, ASIC2a, ASIC2b, ASIC3 or ASIC4, is implicated in a particular disease, condition, or disorder, the disease, condition, or disorder may also be referred to as an "ASIC la, ASIC2a, ASIC2b, ASIC3 or ASIC4-mediated disease, condition or disorder". Accordingly, in another aspect, the present invention provides a method for treating or lessening the severity of a disease, condition, or disorder where activation or hyperactivity of one or more of ASIC la, ASIC2a, ASIC2b, ASIC3 and ASIC4 is implicated in the disease state.
[00137] In certain embodiments, the compounds and compositions of the invention are inhibitors of ASIC.
[00138] The activity of a compound utilized in this invention as an inhibitor of ASIC la, ASIC2a, ASIC2b, ASIC3 or ASIC4, may be assayed according to methods described generally in the examples herein, or according to methods available to one of ordinary skill in the art.
[00139] It is another object of the invention to provide a method for treating diseases that are caused, mediated and/or propagated by ASIC activity, wherein at least one compound of formula (I) according to the invention and/or physiologically acceptable salts thereof is administered to a mammal in need of such treatment. In certain embodiments, the compound is administered in an effective amount as defined above. In certain embodiments, the treatment is an oral administration.
[00140] The method of the invention can be performed either in-vitro or in-vivo. The susceptibility of a particular cell to treatment with the compounds according to the invention can be particularly determined by in-vitro tests, whether in the course of research or clinical application. Typically, a culture of the cell is combined with a compound according to the invention at various concentrations for a period of time which is sufficient to allow the active agents to inhibit ASIC activity, usually between about one hour and one week. In-vitro treatment can be carried out using cultivated cells from a biopsy sample or cell line.
[00141] The host or patient can belong to any mammalian species, for example a primate species, particularly humans; rodents, including mice, rats and hamsters; rabbits; horses, cows, dogs, cats, etc. Animal models are of interest for experimental investigations, providing a model for treatment of human disease.
[00142] For identification of a signal transduction pathway and for detection of interactions between various signal transduction pathways, various scientists have developed suitable models or model systems, for example cell culture models and models of transgenic animals. For the determination of certain stages in the signal transduction cascade, interacting compounds can be utilized in order to modulate the signal. The compounds according to the invention can also be used as reagents for testing ASIC-dependent signal transduction pathways in animals and/or cell culture models or in the clinical diseases mentioned in this application.
[00143] Moreover, the subsequent teaching of the present specification concerning the use of the compounds according to formula (I) and its derivatives for the production of a medicament for the prophylactic or therapeutic treatment and/or monitoring is considered as valid and applicable without restrictions to the use of the compound for the inhibition of ASIC activity if expedient.
[00144] The invention also relates to the use of compounds according to formula (I) and/or physiologically acceptable salts thereof for the prophylactic or therapeutic treatment and/or monitoring of diseases that are caused, mediated and/or propagated by ASIC activity. Furthermore, the invention relates to the use of compounds according to formula (I) and/or physiologically acceptable salts thereof for the production of a medicament for the prophylactic or therapeutic treatment and/or monitoring of diseases that are caused, mediated and/or propagated by ASIC activity. In certain embodiments, the invention provides the use of a compound according to formula I or physiologically acceptable salts thereof, for the production of a medicament for the prophylactic or therapeutic treatment of an ASIC-mediated disorder.
[00145] Compounds of formula (I) and/or a physiologically acceptable salt thereof can furthermore be employed as intermediate for the preparation of further medicament active ingredients. The medicament is preferably prepared in a non-chemical manner, e.g. by combining the active ingredient with at least one solid, fluid and/or semi-fluid carrier or excipient, and optionally in conjunction with a single or more other active substances in an appropriate dosage form.
[00146] The compounds of formula (I) according to the invention can be administered before or following an onset of disease once or several times acting as therapy. The aforementioned compounds and medical products of the inventive use are particularly used for the therapeutic treatment. A therapeutically relevant effect relieves to some extent one or more symptoms of a disorder, or returns to normality, either partially or completely, one or more physiological or biochemical parameters associated with or causative of a disease or pathological condition. Monitoring is considered as a kind of treatment provided that the compounds are administered in distinct intervals, e.g. in order to boost the response and eradicate the pathogens and/or symptoms of the disease completely. Either the identical compound or different compounds can be applied. The methods of the invention can also be used to reduce the likelihood of developing a disorder or even prevent the initiation of disorders associated with ASIC activity in advance or to treat the arising and continuing symptoms.
[00147] In the meaning of the invention, prophylactic treatment is advisable if the subject possesses any preconditions for the aforementioned physiological or pathological conditions, such as a familial disposition, a genetic defect, or a previously incurred disease.
[00148] The invention furthermore relates to a medicament comprising at least one compound according to the invention and/or pharmaceutically usable derivatives, salts, solvates and stereoisomers thereof, including mixtures thereof in all ratios. In certain embodiments, the invention relates to a medicament comprising at least one compound according to the invention and/or physiologically acceptable salts thereof.
[00149] A "medicament" in the meaning of the invention is any agent in the field of medicine, which comprises one or more compounds of formula (I) or preparations thereof (e.g. a pharmaceutical composition or pharmaceutical formulation) and can be used in prophylaxis, therapy, follow-up or aftercare of patients who suffer from diseases, which are associated with ASIC activity, in such a way that a pathogenic modification of their overall condition or of the condition of particular regions of the organism could establish at least temporarily. [00150] In various embodiments, the active ingredient may be administered alone or in combination with other treatments. A synergistic effect may be achieved by using more than one compound in the pharmaceutical composition, i.e. the compound of formula (I) is combined with at least another agent as active ingredient, which is either another compound of formula (I) or a compound of different structural scaffold. The active ingredients can be used either simultaneously or sequentially.
[00151] Included herein are methods of treatment in which at least one chemical entity provided herein is administered in combination with an anti-inflammatory agent. Antiinflammatory agents include but are not limited to NSAIDs, non-specific and COX-2 specific cyclooxygenase enzyme inhibitors, gold compounds, corticosteroids, methotrexate, tumor necrosis factor (TNF) antagonists, immunosuppressants and methotrexate.
[00152] Examples of NSAIDs include, but are not limited to, ibuprofen, flurbiprofen, naproxen and naproxen sodium, diclofenac, combinations of diclofenac sodium and misoprostol, sulindac, oxaprozin, diflunisal, piroxicam, indomethacin, etodolac, fenoprofen calcium, ketoprofen, sodium nabumetone, sulfasalazine, tolmetin sodium, and hydroxychloroquine. Examples of NSAIDs also include COX-2 specific inhibitors such as celecoxib, valdecoxib, lumiracoxib dnd/or etoricoxib.
[00153] In some embodiments, the anti-inflammatory agent is a salicylate. Salicylates include by are not limited to acetylsalicylic acid or aspirin, sodium salicylate, and choline and magnesium salicylates.
[00154] The anti-inflammatory agent may also be a corticosteroid. For example, the corticosteroid may be cortisone, dexamethasone, methylprednisolone, prednisolone, prednisolone sodium phosphate, or prednisone.
[00155] In additional embodiments the anti-inflammatory agent is a gold compound such as gold sodium thiomalate or auranofin.
[00156] The invention also includes embodiments in which the anti-inflammatory agent is a metabolic inhibitor such as a dihydrofolate reductase inhibitor, such as methotrexate or a dihydroorotate dehydrogenase inhibitor, such as leflunomide.
[00157] Other embodiments of the invention pertain to combinations in which at least one anti-inflammatory compound is an anti-monoclonal antibody (such as eculizumab or pexelizumab), a TNF antagonist, such as entanercept, or infliximab, which is an anti-TNF alpha monoclonal antibody.
[00158] Still other embodiments of the invention pertain to combinations in which at least one active agent is an immunosuppressant compound such as an immunosuppressant compound chosen from methotrexate, leflunomide, cyclosporine, tacrolimus, azathioprine, and mycophenolate mofetil.
[00159] The compounds of the invention are also used in combination with chemotherapeutic drugs, in particular, drugs that induce apoptosis. Examples of other chemotherapeutic drugs that can be used in combination with chemosensitizing ASIC inhibitors include topoisomerase I inhibitors (camptothecin or topotecan), topoisomerase II inhibitors (e.g. daunomycin and etoposide), alkylating agents (e.g. cyclophosphamide, melphalan and BCNU), tubulin directed agents (e.g. taxol and vinblastine), and biological agents (e.g. antibodies such as anti CD20 antibody, IDEC 8, immunotoxins, and cytokines).
[00160] The disclosed compounds of the formula I can be administered in combination with other known therapeutic agents, including anticancer agents. As used here, the term "anticancer agent" relates to any agent which is administered to a patient with cancer for the purposes of treating the cancer.
[00161] The anti-cancer treatment defined above may be applied as a monotherapy or may involve, in addition to the herein disclosed compounds of formula I, conventional surgery or radiotherapy or medicinal therapy. Such medicinal therapy, e.g. a chemotherapy or a targeted therapy, may include one or more, but preferably one, of the following anti-tumor agents:
Alkylating agents: such as altretamine, bendamustine, busulfan, carmustine, chlorambucil, chlormethine, cyclophosphamide, dacarbazine, ifosfamide, improsulfan, tosilate, lomustine, melphalan, mitobronitol, mitolactol, nimustine, ranimustine, temozolomide, thiotepa, treosulfan, mechloretamine, carboquone; apaziquone, fotemustine, glufosfamide, palifosfamide, pipobroman, trofosfamide, uramustine, TH-3024, VAL-0834;
Platinum Compounds: such as carboplatin, cisplatin, eptaplatin, miriplatine hydrate, oxaliplatin, lobaplatin, nedaplatin, picoplatin, satraplatin; lobaplatin, nedaplatin, picoplatin, satraplatin;
DNA altering agents: such as amrubicin, bisantrene, decitabine, mitoxantrone, procarbazine, trabectedin, clofarabine; amsacrine, brostallicin, pixantrone, laromustine1'3; Topoisomerase Inhibitors: such as etoposide, irinotecan, razoxane, sobuzoxane, teniposide, topotecan; amonafide, belotecan, elliptinium acetate, voreloxin;
Microtubule modifiers: such as cabazitaxel, docetaxel, eribulin, ixabepilone, paclitaxel, vinblastine, vincristine, vinorelbine, vindesine, vinflunine; fosbretabulin, tesetaxel;
Antimetabolites: such as asparaginase3, azacitidine, calcium levofolinate, capecitabine,
cladribine, cytarabine, enocitabine, floxuridine, fludarabine, fluorouracil, gemcitabine,
mercaptopurine, methotrexate, nelarabine, pemetrexed, pralatrexate, azathioprine, thioguanine, carmofur; doxifluridine, elacytarabine, raltitrexed, sapacitabine, tegafur2'3, trimetrexate;
Anticancer antibiotics: such as bleomycin, dactinomycin, doxorubicin, epirubicin, idarubicin, levamisole, miltefosine, mitomycin C, romidepsin, streptozocin, valrubicin, zinostatin, zorubicin, daunurobicin, plicamycin; aclarubicin, peplomycin, pirarubicin;
Hormones/ Antagonists: such as abarelix, abiraterone, bicalutamide, buserelin, calusterone, chlorotrianisene, degarelix, dexamethasone, estradiol, fluocortolone
fluoxymesterone, flutamide, fulvestrant, goserelin, histrelin, leuprorelin, megestrol, mitotane, nafarelin, nandrolone, nilutamide, octreotide, prednisolone, raloxifene, tamoxifen, thyrotropin alfa, toremifene, trilostane, triptorelin, diethylstilbestrol; acolbifene, danazol, deslorelin, epitiostanol, orteronel, enzalutamide1,3;
Aromatase inhibitors: such as aminoglutethimide, anastrozole, exemestane, fadrozole, letrozole, testolactone; formestane;
Small molecule kinase inhibitors: such as crizotinib, dasatinib, erlotinib, imatinib, lapatinib, nilotinib, pazopanib, regorafenib, ruxolitinib, sorafenib, sunitinib, vandetanib, vemurafenib, bosutinib, gefitinib, axitinib; afatinib, alisertib, dabrafenib, dacomitinib, dinaciclib, dovitinib, enzastaurin, nintedanib, lenvatinib, linifanib, linsitinib, masitinib, midostaurin, motesanib, neratinib, orantinib, perifosine, ponatinib, radotinib, rigosertib, tipifarnib, tivantinib, tivozanib, trametinib, pimasertib, brivanib alaninate, cediranib, apatinib4, cabozantinib S-malate1 3, ibrutinib1'3, icotinib4, buparlisib2, cipatinib4, cobimetinib1'3, idelalisib1,3, fedratinib1, XL-6474;
Photosensitizers: such as methoxsalen3; porfimer sodium, talaporfin, temoporfin;
Antibodies: such as alemtuzumab, besilesomab, brentuximab vedotin, cetuximab, denosumab, ipilimumab, ofatumumab, panitumumab, rituximab, tositumomab,
trastuzumab, bevacizumab, pertuzumab2 3; catumaxomab, elotuzumab, epratuzumab, farletuzumab, mogamulizumab, necitumumab, nimotuzumab, obinutuzumab, ocaratuzumab, oregovomab, ramucirumab, rilotumumab, siltuximab, tocilizumab, zalutumumab, zanolimumab, matuzumab, dalotuzumab1 2'3, onartuzumab1'3, racotumomab1, tabalumab1,3, EMD-5257974, nivolumab1-3;
Cytokines: such as aldesleukin, interferon alfa2, interferon alfa2a3, interferon alfa2b2-3;
celmoleukin, tasonermin, teceleukin, oprelvekin1-3, recombinant interferon beta- la4;
Drug Conjugates: such as denileukin diftitox, ibritumomab tiuxetan, iobenguane 1123,
prednimustine, trastuzumab emtansine, estramustine, gemtuzumab, ozogamicin, aflibercept;
cintredekin besudotox, edotreotide, inotuzumab ozogamicin, naptumomab estafenatox, oportuzumab monatox, technetium (99mTc) arcitumomab1,3, vintafolide1'3;
Vaccines: such as sipuleucel3; vitespen3, emepepimut-S3, oncoVAX4, rindopepimut3, troVax4, MGN-16014, MGN-17034; and
Miscellaneous: alitretinoin. bexarotene, bortezomib, everolimus, ibandronic acid, imiquimod, lenalidomide, lentinan, metirosine, mifamurtide, pamidronic acid, pegaspargase, pentostatin, sipuleucel3, sizofiran, tamibarotene, temsirolimus, thalidomide, tretinoin, vismodegib, zoledronic acid, vorinostat; celecoxib, cilengitide, entinostat, etanidazole, ganetespib, idronoxil, iniparib, ixazomib, lonidamine, nimorazole, panobinostat, peretinoin, plitidepsin, pomalidomide, procodazol, ridaforolimus, tasquinimod, telotristat, thymalfasin, tirapazamine, tosedostat, trabedersen, ubenimex, valspodar, gendicine4, picibanil4, reolysin4, retaspimycin hydrochloride1-3, trebananib2-3, virulizin4, carfilzomib1'3, endostatin4, immucothel4, belinostat3, MGN-17034.
(' Prop. INN (Proposed International Nonproprietary Name); 2 Rec. INN (Recommended
International Nonproprietary Names); 3 USAN (United States Adopted Name); 4 no INN).
[00162] In another aspect, the invention provides for a kit consisting of separate packs of an effective amount of a compound according to the invention and/or pharmaceutically acceptable salts, derivatives, solvates and stereoisomers thereof, including mixtures thereof in all ratios, and optionally, an effective amount of a further active ingredient. The kit comprises suitable containers, such as boxes, individual bottles, bags or ampoules. The kit may, for example, comprise separate ampoules, each containing an effective amount of a compound according to the invention and/or pharmaceutically acceptable salts, derivatives, solvates and stereoisomers thereof, including mixtures thereof in all ratios, and an effective amount of a further active ingredient in dissolved or lyophilized form.
[00163] As used herein, the terms "treatment," "treat," and "treating" refer to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein. In some embodiments, treatment is administered after one or more symptoms have developed. In other embodiments, treatment is admmistered in the absence of symptoms. For example, treatment is administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment is also continued after symptoms have resolved, for example to prevent or delay their recurrence.
[00164] The compounds and compositions, according to the method of the present invention, are administered using any amount and any route of administration effective for treating or lessening the severity of a disorder provided above. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. Compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression "dosage unit form" as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
[00165] Pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated. In certain embodiments, the compounds of the invention are administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 100 mg/kg and preferably from about 1 mg/kg to about 50 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect. [00166] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms optionally contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
[00167] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions are formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation are also a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
[00168] Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[00169] In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This is accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide- polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
[00170] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
[00171] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar—agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form also optionally comprises buffering agents.
[00172] Solid compositions of a similar type are also employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type are also employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
[00173] The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms optionally also comprise buffering agents. They optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
[00174] Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as required. Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
[00175] According to one embodiment, the invention relates to a method of inhibiting ASIC activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
[00176] According to another embodiment, the invention relates to a method of inhibiting ASIC, or a mutant thereof, activity in a biological sample in a positive manner, comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.
[00177] The compounds of the invention can be applied either themselves and/or in combination with physical measurements for diagnostics of treatment effectiveness. Pharmaceutical compositions containing said compounds and the use of said compounds to treat ASIC-mediated conditions is a promising, novel approach for a broad spectrum of therapies causing a direct and immediate improvement in the state of health, whether in human or animal. The orally bioavailable and active new chemical entities of the invention improve convenience for patients and compliance for physicians.
[00178] The compounds of formula (I), their salts, isomers, tautomers, enantiomeric forms, diastereomers, racemates, derivatives, prodrugs and/or metabolites are characterized by a high specificity and stability, low manufacturing costs and convenient handling. These features form the basis for a reproducible action, wherein the lack of cross-reactivity is included, and for a reliable and safe interaction with the target structure.
[00179] The term "biological sample", as used herein, includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
[00180] Modulation of ASIC, or a mutant thereof, activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ transplantation, biological specimen storage, and biological assays.
EXEMPLIFICATION
[00181] As depicted in the Examples below, in certain exemplary embodiments, compounds are prepared according to the following general procedures. It will be appreciated that, although the general methods depict the synthesis of certain compounds of the present invention, the following general methods, and other methods known to one of ordinary skill in the art, can be applied to all compounds and subclasses and species of each of these compounds, as described herein.
[00182] The symbols and conventions used in the following descriptions of processes, schemes, and examples are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry.
[00183] Compound numbers utilized in the Examples below correspond to compound numbers set forth supra.
General Conditions and Analytical Methods
[00184] All solvents used were commercially available and were used without further purification. Reactions were typically run using anhydrous solvents under an inert atmosphere of nitrogen unless otherwise noted.
Analytical Methods
NMR Spectrometers
[00185] Bruker Avance III HD 500 MHz NMR
[00186] Bruker Avance III HD 250 MHz NMR
Configuration of the Bruker Avance III HD 500 MHz NMR
[00187] High performance digital NMR spectrometer, 2-channel console and Windows 7 host workstation running Topspin version 3.2
[00188] Equipped with:
[00189] -Oxford instruments magnet 11.74 Tesla (500 MHz proton resonance frequency)
[00190] -BSVT temperature controller
[00191] -GRASP II gradient spectroscopy accessory for fast acquisition of 2D pulse sequences
[00192] -Deuterium lock switch for gradient shimming/Topshim
[00193] -5mm Broad Band Inverse geometry double resonance probe with automated tuning and matching (BBI ATMA). Allows ¾ observation with pulsing/decoupling of nuclei in the frequency range 15N and 31 P with 2H lock and shielded z-gradient coils.
Configuration of the Bruker Avance III HD 250 MHz NMR
[00194] High performance digital NMR spectrometer, 2-channel nanobay console and Windows 7 host workstation running Topspin version 3.2
[00195] Equipped with:
[00196] -Oxford instruments magnet 5.87 Tesla (250 MHz proton resonance frequency)
[00197] -BSVT temperature controller [00198] -GRASP II gradient spectroscopy accessory for fast acquisition of 2D pulse sequences
[00199] -Deuterium lock switch for gradient shimming/Topshim
[00200] -5mm Broad Band Observe geometry double resonance probe with automated tuning and matching (BBFO ATMA). Allows 1H observation with pulsing/decoupling of nuclei in the frequency range 15N and 31P as well as 19F with ]H decoupling/observation and 2H lock with shielded z-gradient coils.
LCMS Methods
[00201] Example compounds and their intermediates were analysed by HPLC-MS using a combination of the following instrumentation: Shimadzu, Waters or Micromass ZMD, ZQ or LCT mass spectrometers with an Agilent, Waters or Polymer Labs UV and ELS detector. The HPLC conditions are tabulated below. Micromass MassLynx Operating Software with OpenLynx Browser were used for data acquisition, processing and reporting.
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000058_0002
Figure imgf000059_0001
Figure imgf000059_0002
Figure imgf000060_0001
Purification Methods
Figure imgf000060_0002
20 5
Flow rate 40mL/min
Injection Vol 1500μΙ
Detection
Signal UV 215
Generic neutral pH prep method
Waters Sunfire CI 8
Column Part no.186003971
30 x 100mm, lOum
Available on WatersOl
Column Temp Room temperature
A, Water
Mobile Phase
B, Acetonitrile
Gradient Time (mins) % organic
0 10
2 10
2.5 15
14.5 100
15.5 100
16 10
17 10
Flow rate 40mL/min
Injection Vol 1500μΙ.
Detection
Signal UV 215
Generic low pH prep method Waters Sunfire C18
Column Part no.186003971
30 x 100mm, lOum
Available on Waters02
Column Temp Room temperature
A, Water + 0.1% Formic acid
Mobile Phase
B, Acetonitrile + 0.1% Formic acid
Gradient Time (mins) % organic
0 5
2 5
2.5 10
14.5 100
15.5 100
16 5
17 5
Flow rate 40mL/min
Injection Vol 1500μΙ.
Detection
Signal UV 215
Example 1. 2-(4-cyclohexylpiperazin-l-yl)-4H-thieno[3,2-d] [l,3]thiazin-4-
Route A
Figure imgf000063_0001
Methyl 3-[(4-cyclohexylpiperazine-l-carbothioyl)amino]t iophene-2-carboxylate
[00202] To a stirred solution of methyl 3-isothiocyanatothiophene-2-carboxylate (0.2 g, 1 mmol) in anhydrous DCM (3 mL) at room temperature under nitrogen was added dropwise a solution of 1-cyclohexylpiperazine (0.22 g, 1.3 mmol) in anhydrous DCM (1 mL). The mixture was stirred at rt for 25 h. 0.5 M HCI ( 1 mL) was then added and the pH of the reaction mixture subsequently adjusted to 1 1 using 0.5 M aqueous NaOH. The mixture was further diluted with DCM (5 mL) and the two phases separated. The organic phase washed with water (5 mL) and brine (5 mL), dried over Na2S04; filtered and concentrated in vacuo to give 0.25 g (68%) of the title compound as a light yellow oil turning into a light yellow foam. METCR1673 Generic 2 min (low pH) M/Z (ES+) 368, Retention time 1.03 min
2-(4-cyclohexylpiperazin-l-yl)-4H-thieno[3,2-d] [l,3]thiazin-4-one (1)
[00203] A mixture of methyl 3-[(4-cyclohexylpiperazine-l-carbothioyl)amino]thiophene-2- carboxylate (0.1 g, 0.28 mmol) and sulfuric acid (0.8 mL) was stirred at rt for 27 h. The mixture was then poured into ice-water (10 mL). The pH of the mixture was brought to pH 12 using 10M NaOH and the mixture extracted with DCM (3 x 10 mL). The combined organic extracts were washed with water (10 mL), brine (10 mL), dried over MgS04, filtered and concentrated in vacuo. The material was then passed through an Isolute 2g flash N¾ cartridge, the cartridge was washed with DCM (10 mL) and the combined washings were evaporated in vacuo to give 0.03 g (36%) of the title compound as a light yellow solid. MET-uPLC-AB- 101 (7 min, low pH) M/Z (ES+) 336, Retention time 1.81.
l H NMR (500 MHz, DMSO-d6) δ 8.17 (d, J = 5.3 Hz, 1 H), 7.14 (d, J = 5.3 Hz, IK), 3.69 - 3.60 (m, 4H), 2.61 - 2.53 (m, 4H), 2.31 - 2.20 (m, lH), 1.80 - 1.64 (m, 4H), 1.60 - 1 .49 (m, 1 H), 1.25 - 1 .09 (m, 4H), 1.09 -0.97 (m, 1H). [00204] The following examples were prepared in an analogous manner to that described in Route A of Example 1 above, but substituting the 1-cyclohexylpiperazine with the appropriate amine and/or the 3-isothiocyanatothiophene-2-carboxylate with the appropriate aromatic or heteroaromatic isothiocyanate (all reagents whose synthesis is not described in the text are of commercial origin or can be prepared by methods known in the chemical literature):
Example 2.2-[4-(furan-2-carbonyl)piperazin-l-yl]-4H-thieno[3,2-d][l,3]thiazin-4-one (2)
Figure imgf000064_0001
[00205] 0.17 g (71%) as a yellow solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 348, Retention time 2.94.
1H NMR (500 MHz, DMSO-d6) δ 8.26 (d, J = 5.3 Hz, 1H), 7.89 (dd, J = 1.7, 0.7 Hz, 1H), 7.20 (d, J = 5.3 Hz, 1H), 7.07 (dd, J = 3.5, 0.6 Hz, 1H), 6.66 (dd, J = 3.5, 1.8 Hz, 1H), 3.87 - 3.78 (m, 8H).
Example 3. 2-[4-(4-fluorophenyl)piperazin-l-yl]-4H-thieno[3,2-d][l^]thiazin-4-one (3)
Figure imgf000064_0002
[00206] 0.12 g (53%) as a yellow solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 348, Retention time 4.10.
1H NMR (500 MHz, DMSO-d6) δ 8.24 (d, J = 5.3 Hz, 1H), 7.20 (d, J = 5.3 Hz, 1H), 7.12 - 7.05 (m, 2H), 7.03 - 6.97 (m, 2H), 3.92 - 3.79 (m, 4H), 3.25 - 3.17 (m, 4H)
Example 4. 2-(4-phenylpiperazin-l-yl)-4H-thieno[3,2-d][l,3]thiazin-4-one (4)
Figure imgf000065_0001
[00207] 0.12 g (66%) as a yellow solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 330, Retention time 4.10.
1H NMR (500 MHz, DMSO-d6) δ 8.24 (d, J = 5.3 Hz, 1H), 7.25 (dd, J - 8.6, 7.3 Hz, 2H), 7.20 (d, J = 5.3 Hz, 1H), 6.98 (d, J = 7.9 Hz, 2H), 6.83 (t, J = 7.3 Hz, 1H), 3.89 - 3.86 (m, 4H), 3.31 - 3.27 (m, 4H).
Example 5. 2-[4-(4-chIorophenyI)piperazin-l-yI]-4H-thieno[3,2-d][l,3Jthiazin-4-one (5)
Figure imgf000065_0002
[00208] 0.1 g (43%) as a yellow solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 364, 366, Retention time 4.43.
1H NMR (500 MHz, DMSO-d6) δ 8.24 (d, J = 5.3 Hz, lH), 7.30 - 7.24 (m, 2H), 7.20 (d, J = 5.3 Hz, lH), 7.02 - 6.96 (m, 2H), 3.90 - 3.83 (m, 4H), 3.34 - 3.27 (m, 4H).
Example 6. 2-(4-methylpiperazin-l-yl)-4H-thieno[3,2-d] [l,3]thiazin-4-one (6)
Figure imgf000065_0003
[00209] 0.13 g (23%) as an off white solid. METCR1416 Hi Res (7 min, low pH) M/Z (ES+) 268, Retention time 1.13.
1H NMR (500 MHz, DMSO-d6) 6 8.22 (d, J = 5.3 Hz, 1H), 7.16 (d, J = 5.3 Hz, 1H), 3.73 - 3.68 (m, 4H), 2.43 - 2.39 (m, 4H), 2.22 (s, 3H).
Example 7.2-[4-(propan-2-yl)piperazin-l-yl]-4H-thieno[3,2-d][l,3]thiazin-4-one (7)
Figure imgf000066_0001
[00210] 0.01 g (5.5%) as a white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 296, Retention time 1.34.
1H NMR (500 MHz, DMSO-d6) 6 8.23 (d, J = 5.3 Hz, 1H), 7.17 (d, J= 5.3 Hz, 1H), 3.73 - 3.67 (m, 4H), 2.71 (h, J = 6.5 Hz, 1H), 2.55 - 2.53 (m, 4H), 0.99 (d, J = 6.6 Hz, 6H).
Example 8. 2-[4-(pyrimidin-2-yl)piperazin-l-yl]-4H-thieno[3,2-d][l,3]thiazin-4-one (8):
Figure imgf000066_0002
[00211] 0.06 g (30%) as a white solid. MET-uPLC-AB-101 (7 min, low pH) M Z (ES+) 332, Retention time 3.34.
1H NMR (500 MHz, DMSO-d6) δ 8.41 (d, J = 4.7 Hz, 2H), 8.24 (d, J = 5.3 Hz, 1H), 7.20 (d, J = 5.3 Hz, 1H), 6.69 (t, J = 4.7 Hz, 1H), 3.94 - 3.87 (m, 4H), 3.87 - 3.81 (m, 4H).
Example 9.2-[4-(4-methoxyphenyl)piperazin-l-yI]-4H-thieno[3,2-d][l 3]thiazin-4-one (9)
Figure imgf000067_0001
[00212] 0.06 g (19%) as an off white solid. METCR1416 Hi Res (7 min, low pH) M/Z (ES+) 360, Retention time 4.77.
1H NMR (500 MHz, DMSO-d6) δ 8.24 (d, J = 5.3 Hz, 1H), 7.19 (d, J = 5.3 Hz, 1H), 6.98 - 6.93 (m, 2H), 6.87 - 6.81 (m, 2H), 3.89 - 3.82 (m, 4H), 3.69 (s, 3H), 3.17 - 3.11 (m, 4H).
Example 10. 2-[4-(pyridin-2-yl)piperazin-l-yl]-4H-thieno[3,2-d][l,3]thiazin-4-one (10)
Figure imgf000067_0002
[00213] 0.06 g (35.5%) as an off white solid. MET-uPLC-AB-101 M/Z (ES+) 331, Retention time 1.85.
1H NMR (500 MHz, DMSO-d6) δ 8.25 (d, J = 5.3 Hz, 1H), 8.15 (dd, J = 4.8, 1.5 Hz, 1H), 7.59 (ddd, J = 8.9, 7.2, 2.0 Hz, 1H), 7.21 (d, J = 5.3 Hz, IH), 6.87 (d, J = 8.6 Hz, 1H), 6.70 (dd, J = 7.0, 5.0 Hz, IH), 3.89 - 3.82 (m, 4H), 3.71 - 3.65 (m, 4H).
Example 11.2-[4-(oxolan-3-yl)piperazin-l-yI]-4H-thieno[3,2-d][l,3]thiazin-4-one (11)
Figure imgf000067_0003
[00214] 0.03 g (32%) as a beige solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 324, Retention time 1.35.
1H NMR (500 MHz, DMSO-d6) δ 8.22 (d, J = 5.3 Hz, 1H), 7.17 (d, J = 5.3 Hz, 1H), 3.83 - 3.77 (m, 1H), 3.75 (dd, J = 8.6, 6.9 Hz, 1H), 3.70 (t, J = 5.1 Hz, 4H), 3.64 (dd, J = 7.9 Hz, 1H), 3.53 (dd, J = 8.6, 6.3 Hz, lH), 2.97 (p, J = 6.7 Hz, 1H), 2.59 - 2.40 (m, 4H), 2.12 - 1.87 (m, 1H), 1.87 - 1.65 (m, 1H).
Example 12.2-(4-cyclohexylpiperazin-l-yl)-4H-thieno[2r3-d][l,3]thiazin-4-one (12)
Figure imgf000068_0001
[00215] 8.7 mg (5%) as a green solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 336.1, Retention time 1.78.
1H NMR (500 MHz, DMSO-d6) δ 7.18 (s, 2H), 3.71 - 3.65 (m, 4H), 2.62 - 2.57 (m, 4H), 2.34 - 2.26 (m, 1H), 1.79 - 1.71 (m, 4H), 1.61 - 1.55 (m, 1H), 1.20 (s, 4H), 1.13 - 1.03 (m, 1H)
Example 13.2-[4-(l-methyl-lH-imidazol-2-yl)piperazin-l-yl]-4H-thieno[3,2-d] [l,3]thiazin-
4-one (13)
Figure imgf000068_0002
[00216] 0.08 g (34%) as a yellow solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 334, Retention time 1.68.
1H NMR (500 MHz, DMSO-d6) δ 8.24 (d, J = 5.3 Hz, 1H), 7.19 (d, J = 5.3 Hz, 1H), 6.90 (d, J = 1.1 Hz, 1H), 6.61 (d, J = 1.0 Hz, 1H), 3.90 - 3.83 (m, 4H), 3.49 (s, 3H), 3.11 - 3.02 (m, 4H).
Example 14. 2-[(3R)-4-cyclohexyl-3-methyIpiperazin-l-yl]-4H-thieno[3,2-d] [l,3]thiazin-4- one (14)
Figure imgf000069_0001
[00217] 0.03 g (13%) as a yellow solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 350, Retention time 1.95.
1H NMR (500 MHz, DMSO-d6) δ 8.21 (d, J = 5.3 Hz, 1H), 7.16 (d, J = 5.3 Hz, 1H), 3.99 (t, J = 11.6 Hz, 2H), 3.38 - 3.33 (m, 1H), 3.06 (dd, J = 12.8, 8.7 Hz, 1H), 2.85 (dt, J = 11.8, 3.7 Hz, 1H), 2.82 - 2.73 (m, 1H),2.71 - 2.62 (m, 1H), 2.45 - 2.34 (m, 1H), 1.80 - 1.65 (m, 3H), 1.59 (t, J = 12.1 Hz, 2H), 1.38 (qd, J = 11.9, 2.9 Hz, 1H), 1.33 - 1.20 (m, lH), 1.21 - 1.11 (m, 1H), 1.12 - 0.98 (m, 5H).
Example 15. 2-[(3S)-4-cycIohexyI-3-methylpiperazin-l-yl]-4 T-thieno[3^-d] [l,3]thiazin-4- one (15)
Figure imgf000069_0002
[00218] 0.02 g (32%) as a yellow solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 350.1, Retention time 1.88.
1H NMR (500 MHz, DMSO-d6) 6 8.21 (d, J = 5.3 Hz, 1H), 7.16 (d, J = 5.3 Hz, 1H), 3.98 (d, J = 11.0 Hz, 2H), 3.38 (q, J = 7.1 Hz, 1H), 3.06 (dd, J = 12.8, 8.7 Hz, 1H), 2.85 (dt, J = 11.8, 3.6 Hz, 1H), 2.82 - 2.73 (m, 1H), 2.71 - 2.62 (m, 1H), 2.45 - 2.34 (m, 1H), 1.79 - 1.66 (m, 3H), 1.59 (t, J = 11.7 Hz, 2H), 1.43 - 1.33 (m, 1H), 1.33 - 1.22 (m, 1H), 1.22 - 1.12 (m, 1H), 1.1 1- 1.06 (m, 2H), 1.03 (d, J = 6.3 Hz, 3H)
Example 16. l-cyclohexyl-4-{4-oxo-4H-thieno[3,2-d][l,3]thiazin-2-yl}piperazin-2-one (16)
Figure imgf000070_0001
[00219] 0.19 g (76%) as an off white solid. MET-uPLC- AB- 101 (7 min, low pH) M/Z (ES+) 350.1, Retention time 3.40.
1H NMR (500 MHz, DMSO-d6) δ 8.25 (d, J = 5.3 Hz, 1H), 7.22 (d, J = 5.3 Hz, 1H), 4.26 (s, 2H), 4.23 - 4.16 (m, 1H), 3.85 - 3.77 (m, 2H), 3.48 - 3.40 (m, 3H), 1.80- 1.72 (m, 2H), 1.64 - 1.53 (m, 3H), 1.49 - 1.38 (m, 2H), 1.36 - 1.24 (m, 2H), 1.14 - 1.03 (m, 1H).
Example 17.2-[4-(oxan-4-yl)piperazin-l-yl]-4H-thieno[3,2-d][l,3]thiazin-4-one (17)
Figure imgf000070_0002
[00220] 0.01 g ( 11.3%) as a light pink solid. MET-uPLC-AB- 101 (7 min, low pH) M/Z (ES+) 338, Retention time 1.36.
1H NMR (500 MHz, Chloroform-d) 6 7.77 (d, J = 5.3 Hz, lH), 7.10 (d, J = 5.3 Hz, 1H), 4.07 (dd, J = 11.2, 3.8 Hz, 2H), 3.80 (s, 4H), 3.41 (td, J = 11.8, 1.7 Hz, 2H), 2.68 (s, 4H), 2.53 (s, 1H), 1.79 (d, J = 11.6 Hz, 2H), 1.62 (d, J= 12.6 Hz, 2H)
Example 18. 2-(piperazin-l-yl)-4H-thieno[3,2-d][l,3]thiazin-4-one (18)
Figure imgf000070_0003
[00221] 6.4 mg (5.6%) as a white powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 254, Retention time 1.15.
1H NMR (500 MHz, DMSO-d6) δ 8.25 (d, J - 5.3 Hz, 1H), 7.18 (d, J = 5.3 Hz, lH), 3.88 - 3.66 (m, 4H), 3.12 - 2.83 (m, 4H).
Example 19.3-(4-{4-oxo-4H-thieno[3,2-d][l,3]thiazin-2-yl}piperazin-l-yI)benzoic acid (19)
Figure imgf000071_0001
[00222] 0.01 g (11.1%) as an off white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 374, Retention time 3.29.
1H NMR (500 MHz, DMSO-d6) δ 8.25 (d, J = 5.3 Hz, 1H), 7.52 - 7.47 (m, 1H), 7.44 - 7.34 (m, 2H), 7.27- 7.22 (m, 1H), 7.21 (d, J = 5.3 Hz, 1H), 3.93 - 3.86 (m, 4H), 3.40 - 3.32 (m, 4H).
Example 20. 2-[4-(pyridin-2-yl)piperazin-l-yl]-3H,4H-thieno[3,2-d]pyrimidin-4-one (20)
Figure imgf000071_0002
[00223] 4 mg (10.5%) as an off white powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 314, Retention time 1.07.
1H NMR (500 MHz, DMSO-d6) δ 1 1.51 (br s, 1H), 8.16 - 8.1 1 (m, 1H), 8.00 - 7.97 (m, 1H), 7.58 - 7.53 (m, 1H), 7.09 (d, J = 5.2 Hz, 1H), 6.88 (d, J = 8.6 Hz, 1H), 6.67 (dd, J = 7.0, 4.8 Hz, 1H), 3.76 - 3.65 (m, 4H), 3.60 - 3.55 (m, 4H).
Example 21. 4-(4-{4-oxo-4H-thieno[3,2-d] [l,3]thiazin-2-yl}piperazin-l-yl)benzoic acid (21)
Figure imgf000072_0001
[00224] 98 mg (81.9%) as a yellow powder. MET-uPLC-AB- 101 (7 min, low pH) M/Z (ES+) 374.3, Retention time 3.19.
1H NMR (250 MHz, DMSO-d6) δ 8.25 (d, J = 5.3 Hz, 1H), 7.82 - 7.78 (m, 2H), 7.21 (d, J - 5.3 Hz, 1H), 7.00 - 6.96 (m, 2H), 3.91 - 3.85 (m, 4H), 3.54 - 3.48 (m, 4H).
Example 22. 3-(4-{4-oxo-4H-thieno[3,2-d][l,3]thiazin-2-yI}piperazin-l-yl)benzamide (22)
Figure imgf000072_0002
[00225] 34 mg (81.5%) as a pale pink solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 373.1, Retention time 2.79.
1H NMR (500 MHz, DMSO-d6) δ 8.24 (d, J = 5.3 Hz, 1H), 7.81 - 7.74 (m, 2H), 7.71 (s, 1H), 7.21 (d, J = 5.3 Hz, lH), 7.03 (s, 1H), 6.99 - 6.91 (m, 2H), 3.94 - 3.80 (m, 4H), 3.50 - 3.39 (m, 4H).
Example 23. 2-[4-(pyridin-2-yl)piperazin-l-yl]-4H-thieno[3,2-d][l,3]thiazin-4-one (23)
Figure imgf000072_0003
[00226] 0.01 g (12.5%) as a light brown solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 332, Retention time 1.87.
1H NMR (500 MHz, Chlorofrm-d) δ 8.70-8.68 (m, 1H), 7.81-7.79 (m, 1H), 7.38-7.34 (m, 1H), 7.15-7.13 (m, 1H), 7.03-7.01 (m, 1H), 3.99-3.96 (m, 4H), 3.89-3.86 (m, 4H).
Example 24.7-methyl-2-(piperazin-l-yl)-4H-thieno[3,2-d][l,3]thiazin-4-one (24)
Figure imgf000073_0001
[00227] 0.05 g (34.6%) of 7-methyl-2-(piperazin-l-yl)-4H-thieno[3,2-d][l,3]thiazin-4-one. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 268, Retention time 1.45.
1H NMR (500 MHz, DMSO-d6) δ 7.90 (s, 1H), 3.73 - 3.65 (m, 4H), 2.85 - 2.77 (m, 4H), 2.22 (s, 3H).
Example 25. l-{4-oxo-4H-thieno[3^-d][l,3]thiazin-2-yl}piperidine-4-carboxamide (25)
Figure imgf000073_0002
[00228] 0.27 g (77.7%) as an off white powder. METCR1600 (high pH 7 min) M/Z (ES+) 296, Retention time 3.30.
1H NMR (500 MHz, DMSO-d6) δ 8.22 (d, J = 5.3 Hz, 1H), 7.33 (s, 1H), 7.17 (d, J = 5.3 Hz, 1H), 6.84 (s, 1H), 4.40 - 4.26 (m, 2H), 3.24 - 3.13 (m, 2H), 2.49 - 2.43 (m, 1H), 1.88 - 1.80 (m, 2H), 1.63 - 1.51 (m, 2H).
Example 26. 2-(piperazin-l-yl)-4H-3,l-benzothiazin-4-one (26)
Figure imgf000074_0001
[00229] 13.3 mg (17.8%) as an off white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 248.1 , Retention time 1.43.
1H NMPv (250 MHz, DMSO-d6) 6 9.32 (brs, 1H), 7.97 - 7.92 (m, 1H), 7.85 - 7.73 (m, 1H), 7.46 - 7.36 (m, 1H), 7.34 - 7.27 (m, 1H), 3.98 - 3.91 (m, 4H), 3.27 - 3.19 (m, 4H).
Example 27. 2-(4-aminopiperidin-l-yI)-4H-thieno[3,2-d][l,3 thiazin-4-one (27)
Figure imgf000074_0002
[00230] 212 mg (99.6%) as a pale yellow solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 268.1 , Retention time 1.30.
1H NMR (500 MHz, DMSO-d6) δ 8.27 (s, 2H), 8.24 (d, J = 5.3 Hz, 1H), 7.18 (d, J = 5.3 Hz, 1H), 4.44 - 4.30 (m, 2H), 3.45 - 3.31 (m, 1H), 3.26 - 3.16 (m, 2H), 2.12 - 1.99 (m, 2H), 1.57 (qd, J = 12.5, 4.3 Hz, 2H).
Example 28. N-(l-{4-oxo-4H-thieno[3,2-d][l,3]thiazin-2-yl}piperidin-4-yl)acetamide (28)
Figure imgf000074_0003
[00231] 9.8 mg (13.5%) as an off white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 310.1, Retention time 2.32.
1H NMR (500 MHz, DMSO-d6) δ 8.22 (d, J = 5.3 Hz, 1H), 7.88 (d, J = 7.6 Hz, 1H), 7.18 (d, J = 5.3 Hz, 1H), 4.24 (d,J = 13.4 Hz, 2H), 3.95 - 3.83 (m, 1H), 3.31 - 3.27 (m, 2H), 1.91 -1.83 (m, 2H), 1.81 (s, 3H), 1.46 - 1.34 (m, 2H).
Example 29. 4-(4-{7-methyl-4-oxo-4H-thieno[3,2-d] [l,3]thiazin-2-yl}piperazin-l- yl)benzamide (29)
Figure imgf000075_0001
[00232] 8.25 mg (13.3%) as a beige powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 387.1, Retention time 3.15.
1H NMR (500 MHz, DMSO-d6) δ 7.96 - 7.90 (m, 1H), 7.82 - 7.74 (m, 2H), 7.71 (s, 1H), 7.02 (s, 1H), 7.00 - 6.93 (m, 2H), 3.95 - 3.85 (m, 4H), 3.50 - 3.43 (m, 4H), 2.25 (d, J = 0.9 Hz, 3H).
Example 30. 4-{4-oxo-4H-thieno[3,2-d] [l,3]thiazin-2-yl}piperazine-l-carboxamide (30)
Figure imgf000075_0002
[00233] 0.1 1 g (78%) as an off white powder. MET-uPLC-AB- 101 (7 min, low pH) M/Z (ES+) 297, Retention time 2.03.
1H NMR (500 MHz, DMSO-d6) δ 8.22 (d, J = 5.2 Hz, 1H), 7.18 (d, J = 5.2 Hz, 1H), 6.10 (s, 2H), 3.75 - 3.65 (m, 4H), 3.48 - 3.41 (m, 4H).
Example 31. N-methyl-4-(4-{4-oxo-4H-thieno[3,2-d][l,3]thiazin-2-yl}piperazin-l- yl)cyclohexane-l-carboxamide (31)
Figure imgf000076_0001
[00234] 13.5 mg (30.1%) as a yellow powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 387.2, Retention time 2.96.
1H NMR (500 MHz, DMSO-d6) 5 8.25 (d, J = 5.3 Hz, 1H), 8.15 (q, J = 4.1 Hz, 1H), 7.74 (d, J = 8.9 Hz, 2H), 7.21 (d, J = 5.3 Hz, IH), 6.97 (d, J = 8.9 Hz, 2H), 3.93 - 3.78 (m, 4H), 3.50 - 3.40 (m, 4H), 2.75 (d, J = 4.5 Hz, 3H).
Example 32. 2-[4-(dimethylamino)piperidin-l-yl]-4H-thieno[3,2-d] [l,3]thiazin-4-one (32)
Figure imgf000076_0002
[00235] 13.4 mg (19.3%) of 2-[4-(dimethylamino)piperidin-l -yl]-4H-thieno[3,2- d][l,3]thiazin-4-one. MET-uPLC-AB- 101 (7 min, low pH) M/Z (ES+) 296.2, Retention time 1.36.
lH NMR (500 MHz, DMSO-d6) δ 8.21 (d, J = 5.3 Hz, IH), 7.16 (d, J = 5.3 Hz, 1H), 4.36 - 4.29 (m, 2H), 3.20 - 3.1 1 (m, 2H), 2.47 - 2.38 (m, 1H), 2.18 (d, J = 2.5 Hz, 6H), 1.91 - 1.80 (m,2H), 1.46 - 1.35 (m, 2H).
Example 33. 6-methyl-2-(piperazin-l-yl)-4H-thieno[3,2-d][l,3]thiazin-4-one (33)
Figure imgf000076_0003
[00236] Route A was followed using methyl 3-isothiocyanato-5-methylthiophene-2- Carboxylate in the place of methyl 3-isothiocyanatothiophene-2 -carboxylate.
41.6 mg (51.8%) as an off white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 268, Retention time 1.4.
1H NMR (500 MHz, DMSO-c 6) δ 6.91 (d, J= 1.1 Hz, 1H), 3.66 - 3.57 (m, 4H), 2.76 (dd, J = 6.0, 4.3 Hz, 4H), 2.52 (d, J= 1.0 Hz, 3H).
Example 34.
Figure imgf000077_0001
methyl 3-isothiocyanato-5-methylthiophene-2-Carboxylate
[00237] The methyl 3-isothiocyanato-5-methylthiophene-2-Carboxylate was prepared in the following manner: A solution of thiophosgene (0.12 ml, 1.61 mmol) in Chloroform (4ml) was added dropwise to aqueous NaHC03 (2M, 2ml), then methyl 3-amino-5-methylthiophene-2- carboxylate (250 mg, 1.46 mmol) in Chloroform (4ml) was added via a dropping funnel and was stirred at r.t over night. The organic layer was separated and the aqueous layer extracted with CHC13. Combined organics were dried over Na2S04. The residue obtained was purified by column chromatography, in a gradient of 0% Heptane to 40% EtOAc / Heptane. 134 mg (43%) of the title compound was obtained as a white solid. METCR1673 Generic 2 min M/Z (ES+) N/A , Retention time 1.42.
1H NMR (250 MHz, Chloroform-d) δ 6.61 (d, J = 1.0 Hz, 1H), 3.90 (s, 3H), 2.46 (d, J = 0.9 Hz, 3H).
Example 35.2-(4-acetylpiperazin-l-yl)-4H-thieno[3,2-d][l,3]thiazin-4-one (34)
Figure imgf000077_0002
[00238] 12 mg (9.3%) as an off white powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 296, Retention time 2.51.
1H NMR (500 MHz, DMSO-d6) δ 8.24 (d, J = 5.3 Hz, 1H), 7.19 (d, J = 5.3 Hz, 1H), 3.80 - 3.76 (m, 2H), 3.74 - 3.69 (m, 2H), 3.57-3.61 (m, 4H), 2.04 (s, 3H).
Example 36. 2-[4-(3-methylpyridin-2-yl)piperazin-l-yl]-4H-thieno[3,2-dl [l,3]thiazin-4-one
(35)
Figure imgf000078_0001
[00239] 49 mg (68.7%) as a light brown solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 345.1 , Retention time 2.68.
1H NMR (500 MHz, DMSO-d6) δ 8.29 - 8.21 (m, 1H), 8.17 - 8.10 (m, 1H), 7.61 - 7.49 (m, 1H), 7.27 - 7.14 (m,lH), 7.04 - 6.91 (m, 1H), 3.93 - 3.82 (m, 4H), 3.25 - 3.14 (m, 4H), 2.29 (s,3H).
Example 37.
Figure imgf000078_0002
l-(3-methylpyridin-2-yl)piperazine
[00240] The l-(3-methylpyridin-2-yl)piperazine required for the synthesis as per procedure A was prepared as follows: Piperazine (300 mg, 3.48 mmol) was dissolved in DMSO (3ml) in a pressure tube and 2-fluoro-3-methylpyridine (464.41 mg, 4.18 mmol) was added. Reaction was heated to 130°C overnight. IPC desired product in solvent front peak, M+l = 178.00. Reaction was diluted with 10ml water and extracted 3 x with lOml EtOAc. Combined organics were washed with water, brine and dried over Na2S04 and purified by column chromatography, eluting the desired product in 30% MeOH / EtOAc to give 222 mg (33.4%) of l-(3- methylpyridin-2-yl)piperazine.
1H NMR (250 MHz, Methanol-d4) δ 8.06 (dd, J = 4.9, 1.3 Hz, 1H), 7.53 (dd, J = 7.4, 1.0 Hz, 1H), 6.94 (dd, J = 7.4, 5.0 Hz, 1H), 3.13 - 3.04 (m, 4H), 3.03 - 2.94 (m, 4H).
Example 38. 6-(4-{4-oxo-4H-thieno[3,2-d] [l,3]thiazin-2-yl}piperazin-l-yl)pyridine-3- carboxamide (36)
Figure imgf000079_0001
[00241] 1.1 mg (0.9%) as a light grey powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 374, Retention time 2.33.
1H NMR (500 MHz, DMSO-d6) 6 8.65 (d, J = 2.3 Hz, 1H), 8.24 (d, J = 5.3 Hz, 1H), 8.01 (dd, J = 9.0, 2.4 Hz, 1H), 7.78 (s, 1H), 7.21 (d, J = 5.3 Hz, 1H), 7.15 (s, 1H), 6.87 (d, J = 9.0 Hz, 1H), 3.91 - 3.83 (m, 4H), 3.83 - 3.77 (m, 4H).
Example 39. 2-[4-(pyridin-2-yl)piperazin-l-yI]-4H-thieno[3,2-d] [l^]thiazin-4-one (37)
Figure imgf000079_0002
[00242] 0.02g (26.2%) as a white solid: MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 212.95, Retention time 1.06.
lH NMR (500 MHz, DMSO-d6) 5 11.40 (br s, 1H), 8.15 - 8.12 (m, 1H), 7.99 (d, J = 5.2 Hz, 1H), 7.59 - 7.52 (m, 1H), 7.10 (d, J = 5.2 Hz, 1H), 6.88 (d, J = 8.6 Hz, 1H), 6.67 (dd, J = 6.8, 5.1 Hz, 1H), 3.76 - 3.64 (m,4H), 3.63 - 3.52 (m, 4H). Example 40. 3-(4-{4-oxo-4H-thieno[3,2-d][l,3]thiazin-2-yI}piperazin-l-yl)benzamide (38)
Figure imgf000080_0001
[00243] To a solution of 3-(4-{4-oxo-4H-thieno[3,2-d][l,3]thiazin-2-yl}piperazin-l- yl)benzoic (see synthesis described above using procedure A (0.04 g, 0.1 mmol) in dry DMF (1.3 mL) was added HATU (0.05 g, 0.13 mmol) and N-ethyldiisopropylamine (53.1 1 μΐ, 0.32 mmol) and stirring continued for 30 min . 7 N ammonia in MeOH (30.6 μΐ, 0.21 mmol) was then added and stirring continued at room temperature overnight. The solvent was evaporated and the crude material was purified by preparative HPLC. Yield: 36 mg (57.8%) as an off white powder. METCR1600 (high pH 7 min) M/Z (ES+) 373, Retention time 3.94.
1H NMR (500 MHz, DMSO-d6) δ 8.25 (d, J = 5.3 Hz, 1H), 7.91 (s, l H), 7.45 (s, 1H), 7.36 - 7.27 (m, 3H), 7.21 (d, J = 5.3 Hz, 1H), 7.15 - 7.09 (m, lH), 3.92 - 3.85 (m, 4H), 3.39 - 3.33 (m, 4H).
Example 41. 2-[4-(pyridin-2-yI)piperazin-l-yI]-3H,4H-pyrido[3,2-d]pyrimidin-4-one (39) Route B
Figure imgf000080_0002
Methyl 3-isothiocyanatopyridine-2-carboxylate
[00244] Thiophosgene (0.28 ml, 3.61 mmol) in Chloroform (6ml) was added dropwise, using a dropping funnel, to a flask containing 2M aqueous sodium bicarbonate (3.5 ml). To this methyl 3-aminopyridine-2-carboxylate (500 mg, 3.29 mmol) in Chloroform (6ml) was added drop wise. Reaction was stirred at r.t over night.
[00245] On completion the organic layer was separated and the aqueous layer extracted with CHC13 (6 mL). The combined organic extracts were washed with brine (10 mL), dried over Na2S04, filtered and concentrated in vacuo to give 570 mg (89.3%) of the title compound as a yellow solid.
1H NMR (500 MHz, DMSO-d6) 6 8.62 (dd, J = 4.6, 1.4 Hz, 1H), 8.04 (dd, J = 8.2, 1.4 Hz, 1H), 7.72 (dd, J = 8.2, 4.6 Hz, 1H), 3.92 (s, 3H).
2-sulfanyl-3H,4H-pyrido[3,2-d]pyrimidin-4-one
[00246] Methyl 3-isothiocyanatopyridine-2-carboxylate (570 mg, 2.93 mmol) was dissolved in THF (7 ml) in a pressure tube and 7M ammonia in MeOH (0.55 ml) was added. Reaction was sealed and heated to 80°C. Reaction was heated for 6h. IPC showed mainly cyclised product. The reaction was allowed to cool to r.t and the ppt was filtered and washed with THF to afford 465 mg (86.6%) the title compound as an off white solid.
1H NMR (500 MHz, DMSO-d6) δ 12.49 (s, 1H), 8.55 (dd, J = 4.1, 1.6 Hz, 1H), 7.74 - 7.67 (m, 2H).
2-(methylsulfanyl)-3H,4H-pyrido[3,2-d]pyrimidin-4-one
[00247] 2-sulfanyl-3H,4H-pyrido[3,2-d]pyrimidin-4-one (98%, 465 mg, 2.54 mmol) was dissolved in'Tetrahydrofuran (12 ml) in a pressure tube and Sodium hydride 60% in mineral oil, (111.88 mg, 2.8 mmol) was added. Reaction was sealed and heated to 85°C for Ih. After lh the reaction was cooled to r.t and iodomethane (174.14 μΐ, 2.8 mmol) was added. Reaction was heated to 85°C again for a further 2h. IPC showed 55% SMe, M+l = 193.85 and 39% N-Me, M+l = 207.90.
[00248] Reaction was cooled to r.t and ppt was filtered, washed with water and dried under vacuo. Purified by column chromatography to give 206 mg (41.9%) of 2-(methylsulfanyl)- 3H,4H-pyrido[3,2-d]pyrimidin-4-one as a pale yellow crystalline solid. METCR Generic 2 min M/Z (ES+) 193.9, Retention time 0.79
2-[4-(pyridin-2-yl)piperazin-l-yl]-3H,4H-pyrido[3,2-d]pyrimidin-4-one
[00249] 2-(methylsulfanyl)-3H,4H-pyrido[3,2-d]pyrimidin-4-one (100 mg, 0.52 mmol) and 1-
(pyridin-2-yl)piperazine (0.39 ml, 2.59 mmol) were added to a sealable pressure tube and melted without solvent at 140°C over night. On completion reaction was allowed to cool and the ppt was filtered. Solid was slurried in hot methanol to yield 54 mg (33.8%) of the title compound. METCR1600 (high pH 7 min) M/Z (ES+) 309.2, Retention time 2.35.
lH NMR (500 MHz, DMSO-d6) 6 11.49 (s, 1H), 8.44 (dd, J = 4.2, 1.4 Hz, 1H), 8.16 - 8.13 (m, 1H), 7.70 (dd, J = 8.4, 1.3 Hz, 1H), 7.60 - 7.53 (m, 2H), 6.89 (d, J = 8.6 Hz, 1H), 6.68 (dd, J = 6.8, 5.0 Hz, 1H),3.81 - 3.74 (m, 4H), 3.63 - 3.57 (m, 4H).
[00250] The following examples were prepared in an analogous manner to that described in Route B in Example 41 above, but substituting the l-(pyridin-2-yl)piperazine with the appropriate amine and/or the Methyl 3-isothiocyanatopyridine-2-carboxylate with the appropriate aromatic or heteroaromatic isothiocyanate (all reagents whose synthesis is not described in the text are of commercial origin or can be prepared by methods known in the chemical literature):
Example 42. 4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)benzamide (40)
Figure imgf000082_0001
[00251] 2.1 mg (1%) as a white powder. METCR1416 (Hi res 7 min) M/Z (ES+) 355.9, Retention time 2.87.
1H NMR (500 MHz, DMSO-d6) δ 8.45 (s, 1H), 7.89 (d, J = 5.2 Hz, 1H), 7.77 (d, J = 8.9 Hz, 2H), 7.71 (s, 1H), 7.06 (d, J = 5.2 Hz, 1H), 7.03 - 6.94 (m, 3H), 3.78 - 3.69 (m, 4H), 3.38 - 3.28 (m, 4H).
Example 43. 3-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)benzamide (41)
Figure imgf000083_0001
[00252] 5.6 mg (37.4%) as an off white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 356.2, Retention time 1.83.
1H NMR (500 MHz, DMSO-d6) δ 11.49 (s, 1H), 8.02 (d, J = 5.2 Hz, 1H), 7.92 (s, 1H), 7.46 (s, 1H), 7.34- 7.26 (m, 3H), 7.16 - 7.12 (m, 1H), 7.11 (d, J = 5.2 Hz, 1H), 3.79 - 3.71 (m, 4H), 3.30 - 3.24 (m, 4H).
Example 44. 2-(4-cyclohexylpiperazin-l-yl)-3H,4H-thieno[3,2-d]pyrimidin-4-one (42)
Figure imgf000083_0002
[00253] 16.6 mg (20.7%) as a beige powder. MET-uPLC-AB- 101 (7 min, low pH) M/Z (ES+) 319.2, Retention time 1.20.
1H NMR (500 MHz, DMSO-d6) 6 11.31 (brs, 1H), 7.99 (d, J = 5.2 Hz, 1H), 7.06 (d, J = 5.2 Hz, 1H), 3.60 - 3.43 (m, 4H), 2.55 - 2.52 (m, 4H), 2.29 - 2.21 (m, 1H), 1.80 - 1.70 (m, 4H), 1.62 - 1.52 (m, lH), 1.26 - 1.15 (m, 4H), 1.13 - 1.03 (m, 1H).
Example 45. 5-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)pyridine-2- carboxamide (43)
Figure imgf000084_0001
[00254] 7.1 mg (2.2%) as a yellow powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 357.2, Retention time 1.65.
1H NMR (500 MHz, DMSO-d6) δ 11.36 (br s, 1H), 8.32 (d, J = 2.8 Hz, 1H), 8.00 (d, J = 5.1 Hz, 1H), 7.86 (d, J= 8.8 Hz, 1H), 7.78 (s, 1H), 7.45 (dd, J = 8.9, 2.9 Hz, 1H), 7.31 (s, 1H), 7.10 (d, J = 5.2 Hz, 1H), 3.81- 3.73 (m, 4H), 3.48 - 3.41 (m, 4H).
Example 46. N-(2-hydroxyethyl)-4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin- l-yl)benzamide (44)
Figure imgf000084_0002
[00255] 0.04 g (12.7%) as an off white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 400.2, Retention time 1.72.
1H NMR (500 MHz, DMSO-d6) δ 1 1.44 (br s, 1H), 8.15 (t, J = 5.6 Hz, 1H), 8.01 (d, J = 5.2 Hz, 1H), 7.75 (d, } = 8.9 Hz, 2H), 7.10 (d, J = 5.2 Hz, lH), 7.00 (d, J = 8.9 Hz, 2H), 4.68 (t, J = 5.6 Hz, 1H), 3.81 - 3.69 (m,4H), 3.48 (q, J = 6.2 Hz, 2H), 3.40 - 3.33 (m, 4H), 3.30 - 3.24 (m, 2H).
Example 47. 4-(4-{4-oxo-3H,4H-pyrido[3,2-d]pyrimidin-2-yl}piperazin-l-yl)benzamide (45)
Figure imgf000085_0001
[00256] 1.3 mg (0.7%) as a yellow solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 351.1, Retention time 1.40.
1H NMR (500 MHz, DMSO-d6) δ 1 1.64 (s, 1H), 8.41 (d, J = 3.6 Hz, 1H), 7.80 - 7.63 (m, 4H), 7.58 - 7.50 (m, 1H), 7.06 - 6.95 (m, 3H), 3.85 - 3.76 (m, 4H).
Example 48. N,2-dimethyl-4-(4-{4-oxo-3H,4H-py rido [3,2-d] pyrimidin-2-yl} piperazin-l-yl) benzamide (46)
Figure imgf000085_0002
[00257] 7.1 mg (2.8%) as an off-white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 379.1 , Retention time 1.56.
1H NMR (500 MHz, DMSO-d6) δ 1 1.66 (brs, 1H), 8.42 (dd, J= 4.2, 1.5 Hz, 1H), 7.90 (q, 1H), 7.67 (dd, 1H), 7.59 - 7.51 (m, 1H), 7.26 (d, J = 8.5 Hz, 1H), 6.84 - 6.82 (m, 1H), 6.82 - 6.78 (m, 1H), 3.83 - 3.74 (m, 4H), 3.29 - 3.24 (m, 4H), 2.71 (d, J = 4.6 Hz, 3H), 2.34 (s, 3H).
Example 49. N-(3-hydroxy propy l)-5-(4-{3-methy l-4-oxo-3H,4H-py rido [3,2-d] py rimidin-2- yl}piperazin-l-yl)pyridine-2-carboxamide (47)
Figure imgf000086_0001
[00258] 2.0 mg (1.1%) as a yellow powder. METCR1600 (high pH 7 min) M/Z (ES+) 424.45, Retention time 2.93.
1H NMR (500 MHz, DMSO-d6) δ 8.63 (d, J = 4.2 Hz, 1H), 8.49 (t, J = 6.0 Hz, 1H), 8.35 (d, J = 2.6 Hz, 1H), 7.92 - 7.84 (m, 2H), 7.70 (dd, J = 8.3, 4.2 Hz, 1H), 7.48 (dd, J = 8.8, 2.5 Hz, 1H), 4.52 (t, J = 5.2 Hz, 1H), 3.58 - 3.50 (m, 7H), 3.50 - 3.43 (m, 2H), 3.43 - 3.38 (m, 4H), 3.38 - 3.32 (m, 2H), 1.70 - 1.62 (m, 2H).
Example 50.
Figure imgf000086_0002
3-methyl-2-(methylsulfanyl)-3H,4H-pyrido[3,2-d]pyrimidin-4-one
[00259] The 3-methyl-2-(methylsulfanyl)-3H,4H-pyrido[3,2-d]pyrimidin-4-one required for the synthesis was prepared as follows: To a stirred suspension of Sodium hydride 60% in mineral oil (60%, 45.54 mg, 1.14 mmol) in anhydrous THF (4.3 mL) in a sealable pressure tube under nitrogen was added dropwise a solution of 2-(methylsulfanyl)-3H,4H-pyrido[3,2-d]pyrimidin-4- one (200 mg, 1.04 mmol) in anhydrous THF (4.3 mL) (slowly, temperature did not go above 25
°C). The mixture was stirred at rt for 1 hour after which iodomethane (70.88 μΐ, 1.14 mmol) was added. The tube was then sealed and the mixture heated to reflux (85 °C) overnight. More iodomethane (35 μΐ, 0.56 mmol) was added and stirring at rt continued for ~3 hours. The mixture was allowed to cool to rt and was left standing for two days. A precipitate was then collected by filtration and washed with heptane (2 mL) and water (2 2 mL). The solid obtained was dried in vacuo to give 0.07 g (34%) of 3-methyl-2-(methylsulfanyl)-3H,4H-pyrido[3,2-d]pyrimidin-4-one as a yellow solid. METCR Generic 2 min M/Z (ES+) 207.9, Retention time 0.86 Example 51. N-methyI-5-(4-{4-oxo-3H,4H-pyrido[3,2-d]pyrimidin-2-yl}piperazin-l- yl)pyridine-2-carboxamide (48)
Figure imgf000087_0001
[00260] 0.01 g (6.3%) as yellow powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 366, Retention time 1.45.
H NMR (500 MHz, DMSO-d6) δ 11.71 (s, 1H), 8.45 (d, J = 3.2 Hz, 1H), 8.42 (q, J = 4.7 Hz, 1H), 8.33 (d, J = 2.8 Hz, 1H), 7.86 (d, J = 8.8 Hz, 1H), 7.70 (d, J = 8.1 Hz, 1H), 7.59 (dd, J = 8.4, 4.2 Hz, 1H), 7.46 (dd, J = 8.8, 2.9 Hz, 1H), 3.91 - 3.77 (m, 4H), 3.51 - 3.42 (m, 4H), 2.79 (d, J = 4.8 Hz, 3H).
Example 52. 2-[4-(dimethylamino)piperidin-l-yl]-3H,4H-thieno[3,2-d]pyrimidin-4-one (49)
Figure imgf000087_0002
[00261] 3 mg (1.1%) as a yellow powder. METCR1600 (high pH 7 min) M/Z (ES+) 279.2, Retention time 2.15.
1H NMR (500 MHz, DMSO-d6) δ 7.99 (d, J = 5.2 Hz, 1H), 7.07 (d, J = 5.2 Hz, 1H), 4.43 - 4.24 (m, 2H), 3.02 - 2.83 (m, 3H), 2.27 (s, 6H), 1.87 - 1.77 (m, 2H), 1.49 - 1.34 (m, 2H).
Example 53. 2-[4-(pyridin-3-yl)piperazin-l-yl]-3H,4H-pyrido[3,2-d]pyrimidin-4-one (50)
Figure imgf000088_0001
[00262] 12 mg (7.1%) as an off white solid. METCR1600 (high pH 7 min) M/Z (ES+) 309.2, Retention time 2.16.
1H NMR (500 MHz, DMSO-d6) δ 8.44 (dd, J = 4.2, 1.4 Hz, 1H), 8.36 (d, J = 2.9 Hz, 1H), 8.03 (dd, J = 4.5, 1.1 Hz, 1H), 7.70 (dd, J = 8.4, 1.4 Hz, 1H), 7.58 (dd, J = 8.4, 4.2 Hz, 1H), 7.41 - 7.36 (m, 1H), 7.24 (dd, J = 8.4, 4.5 Hz, 1H), 3.86 - 3.80 (m, 4H).
Example 54. 4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yI)piperazin-l-yl)benzonitriIe
(51)
Figure imgf000088_0002
[00263] 0.01 g (1.9%) as beige powder. METCR1600 (high pH 7 min) M/Z (ES+) 337.95, Retention time 1.04.
1H NMR (500 MHz, DMSO-d6) δ 1 1.49 (s, 1H), 8.02 (d, J = 5.2 Hz, 1H), 7.62 (dd, J = 9.3, 2.4 Hz, 2H), 7.11 (d, J = 5.2 Hz, 1H), 7.07 (d, J = 9.1 Hz, 2H), 3.77 - 3.73 (m, 4H), 3.49 - 3.45 (m, 4H).
Example 55. 4-(4-{4-oxo-3H,4H-furo[3,2-d]pyrimidin-2-yl}piperazin-l-yl)benzamide (52)
Figure imgf000089_0001
[00264] 5.1 mg (20.1%) as a white solid. METCR1600 (high pH 7 min) M/Z (ES+) 341.2, Retention time 2.51.
1H NMR (500 MHz, DMSO-d6) δ 11.44 (s, 1H), 8.04 (d, J = 2.0 Hz, 1H), 7.76 (d, J = 8.9 Hz, 2H), 7.71 (brs, 1H), 7.01 (brs, 1H), 6.98 (d, J = 9.0 Hz, 2H), 6.71 (d, J = 2.0 Hz, lH), 3.72 - 3.66 (m, 4H), 3.37 - 3.34 (m, 4H).
Example 56. 6-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)pyridazine-3- carboxamide (53)
Figure imgf000089_0002
[00265] 5 mg (2.5%) as white powder; MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 358, Retention time 1.5.
lH NMR (500 MHz, DMSO-d6) 5 1 1.52 (s, 1H), 8.14 (s, 1H), 8.00 (d, J = 5.2 Hz, 1H), 7.88 (d, J = 9.5 Hz, 1H), 7.54 (s, 1H), 7.40 (d, J = 9.6 Hz, 1H), 7.11 (d, J = 5.2 Hz, 1H), 3.86 - 3.81 (m, 4H), 3.79 - 3.75 (m, 4H).
Example 57. 2-[4-(4-methanesulfonylphenyl)piperazin-l-yl]-3H,4H-pyrido[3,2- d]pyrimidin-4-one (54)
Figure imgf000090_0001
[00266] 1 mg (1%) as white powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 384.2, Retention time 1.72.
IH NMR (500 MHz, DMSO-d6) δ 1 1.63 (s, IH), 8.45 (d, J = 3.6 Hz, IH), 7.71 (d, J = 8.9 Hz, 3H), 7.59 (dd, J = 8.3, 4.2 Hz, IH), 7.13 (d, J = 8.9 Hz, 2H), 3.87 - 3.77 (m, 4H), 3.53 - 3.45 (m, 4H), 3.10 (s, 3H).
Example 58. 2-[4-(pyridin-3-yl)piperazin-l-yl]-3H,4H-thieno[3,2-d]pyrimidin-4-one (55)
Figure imgf000090_0002
[00267] 72.1 mg (43.4%) as off-white powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 314.1, Retention time 1.05.
IH NMR (500 MHz, DMSO-d6) δ 1 1.51 (s, IH), 8.37 - 8.33 (m, IH), 8.04 - 8.00 (m, 2H), 7.40 - 7.35 (m, IH), 7.26 - 7.20 (m, IH), 7.10 (d, J = 5.2 Hz, IH), 3.78 - 3.73 (m, 4H), 3.31 - 3.27 (m, 4H).
Example 59. 6-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)pyridine-3- carboxamide (56)
Figure imgf000091_0001
[002681 6-(4- {4-oxo-3H,4H-thieno [3 ,2-d]pyrimidin-2-yl }piperazin- 1 -y l)pyridine-3 - carbonitrile was prepared using general procedure B and then hydrolysis as described below gave the final product, 6-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)pyridine- 3-carboxamide: A mixture of 6-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l- yl)pyridine-3 -carbonitrile (50 mg, 0.15 mmol) in H2S04 (0.5 ml) under nitrogen was stirred at rt overnight. Following completion, the mixture was passed through an isolute 2g NH2 cartridge using DMSO (15 mL) to release the product. DMSO was removed and the compound was dried to give 11.7 mg (22.2%) of 6-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l- yl)pyridine-3 -carboxamide as a light yellow solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 356.9, Retention time 1.33.
1H NMR (500 MHz, DMSO-d6) δ 11.38 (br s, 1H), 8.64 (d, J = 2.3 Hz, 1H), 7.99 (dd, J = 9.0, 2.4 Hz, 1H), 7.96 (d, J = 5.2 Hz, 1H), 7.77 (s, 1H), 7.13 (s, 1H), 7.08 (d, J = 5.2 Hz, 1H), 6.89 (d, J = 9.0 Hz, 1H), 3.78 -3.65 (m, 8H).
Example 60. 4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)cycIohexane-l- carboxamide (57)
Figure imgf000091_0002
[00269] 4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)cyclohexane-l- carboxamide was prepapred in a manner analogous to that described above for 6-(4-{4-oxo- 3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)pyridine-3-carboxamide: 22.6 mg (42.5%) as white powder. METCR1600 (high pH 7 min) M/Z (ES+) 362.2, Retention time 2.19.
1H NMR (500 MHz, DMSO-d6) δ 11.31 (s, 1H), 8.00 (d, J = 5.2 Hz, 1H), 7.19 - 7.12 (m, 1H), 7.09 - 7.06 (m, 1H), 6.68 - 6.62 (m, lH), 3.61 - 3.52 (m, 4H), 2.56 - 2.53 (m, 1H), 2.53 - 2.52 (m, 4H), 2.29 - 2.22 (m, 1H), 2.23 - 2.17 (m, 1H), 1.93 - 1.77 (m, 3H), 1.76 - 1.68 (m, 1H), 1.52 - 1.37 (m, 3H), 1.27 - 1.15 (m, 1H).
Example 61. 3-methyl-2-[4-(pyridin-2-yl)piperazin-l-yl]-3H,4H-thieno[3,2-d]pyrimidin-4- one (58) and l-{4-methoxythieno[3,2-d]pyrimidin-2-yl}-4-(pyridin-2-yl)piperazine (59)
Figure imgf000092_0001
[00270] 2-[4-(pyridin-2-yl)piperazin-l-yl]-3H,4H-thieno[3,2-d]pyrimidin-4-one was prepared using general procedure B and then alkylation as described below gave the final products, 3- methyl-2-[4-(pyridin-2-yl)piperazin-l-yl]-3H,4H-thieno[3,2-d]pyrimidin-4-one and l-{4- methoxythieno[3,2-d]pyrimidin-2-yl}-4-(pyridin-2-yl)piperazine: To a stirred suspension of Sodium hydride 60% in mineral oil (60%, 5.33 mg, 0.13 mmol) in anhydrous THF (0.5 mL) in a sealable pressure tube under nitrogen was added drop wise a suspension of 2-[4-(pyridin-2- yl)piperazin-l-yl]-3H,4H-thieno[3,2-d]pyrimidin-4-one (38 mg, 0.12 mmol) in anhydrous THF (0.5 mL). The mixture was stirred at rt for 55 min after which iodomethane (8.3 μΐ, 0.13 mmol) was added. The tube was then sealed and the mixture heated at reflux (85 °C) overnight. The solvent was removed and the crude material obtained was purified in the open access preparative HPLC under neutral conditions to give 5.3 mg (13.3%) of 3-methyl-2-[4-(pyridin-2-yl)piperazin- l-yl]-3H,4H-thieno[3,2-d]pyrimidin-4-one as white powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 328.1, Retention time 1.34.
1H NMR (500 MHz, DMSO-d6) δ 8.17 - 8.13 (m, 1H), 8.11 - 8.06 (m, 1H), 7.61 - 7.53 (m, 1H), 7.28 - 7.23 (m, 1H), 6.89 (d, J = 8.5 Hz, 1H), 6.72 - 6.65 (m, 1H), 3.70 - 3.61 (m, 4H), 3.55 (s, 3H), 3.29 - 3.22 (m, 4H).
[00271] 2.5 mg (6.3%) of l-{4-methoxythieno[3,2-d]pyrimidin-2-yl}-4-(pyridin-2- yl)piperazine as a white powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 328.1, Retention time 1.64.
1H NMR (500 MHz, DMSO-d6) δ 8.16 - 8.13 (m, 1H), 8.11 (d, J = 5.3 Hz, 1H), 7.59 - 7.54 (m, lH), 7.24 (d, J= 5.3 Hz, 1H), 6.88 (d, J = 8.6 Hz, 1H), 6.67 (dd, J = 7.1, 5.0 Hz, 1H), 4.06 (s, 3H), 3.91 - 3.86 (m, 4H), 3.63 - 3.57 (m, 4H).
Example 62. l-[4-(propan-2-yIoxy)thieno[3,2-d]pyrimidin-2-yl]-4-(pyridin-2-yl)piperazine
(60)
Figure imgf000093_0001
[00272] 2-[4-(pyridin-2-yl)piperazin-l-yl]-3H,4H-thieno[3,2-d]pyrimidin-4-one was prepared using general procedure B and then alkylation as described below gave the final product, l-[4- (propan-2-yloxy)thieno[3,2-d]pyrimidin-2-yl]-4-(pyridin-2-yl)piperazine: To a stirred suspension of Sodium hydride 60% in mineral oil (60%, 10 mg, 0.25 mmol) in anhydrous THF (0.5 mL) in a sealable pressure tube under nitrogen was added dropwise a suspension of 2-[4- (pyridin-2-yl)piperazin-l-yl]-3H,4H-thieno[3,2-d]pyrimidin-4-one in anhydrous THF (0.5 mL. The mixture was stirred at rt for 1.2 h after which 2-iodopropane (11.21 μΐ, 0.11 mmol) was added (at 14:40). The tube, was then sealed and the mixture heated at reflux (105 °C) and stirred overnight. Added more 2-iodopropane (11.21 μΐ, 0.11 mmol) and heating to 120 °C overnight. The volatiles were evaporated to give 0.065 g of crude material as a brown gum. The crude material obtained was purified using a Biotage Isolera 4 flash purification system and a gradient of 1-10% MeOH in DCM. 7.4 mg (19.5%) as white powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 356.2, Retention time 2.18.
1H NMR (500 MHz, DMSO-d6) 5 8.47 (s, 1H), 8.15 - 8.13 (m, 1H), 8.09 (d, J = 5.3 Hz, 1H), 7.56 (ddd, J = 8.9, 7.1, 2.0 Hz, 1H), 7.22 (d, J= 5.3 Hz, 1H), 6.88 (d, J = 8.6 Hz, lH), 6.66 (dd, J = 6.8, 5.1 Hz, 1H), 5.50(hept, J = 6.2 Hz, 1H), 3.89 - 3.83 (m, 4H), 3.63 - 3.57 (m, 4H), 1.40 (d, J = 6.2 Hz, 6H).
Example 63.4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yI}piperazin-l-yl)cyclohexane-l- carbonitrile (61)
Figure imgf000094_0001
[00273] 2-(piperazin-l-yl)-3H,4H-tIiieno[3,2-d]pyrimidin-4-one was prepared using general procedure B and then reductive amination as described below gave the final product, 4-(4-{4- oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)cyclohexane-l-carbonitrile: To a solution of 2-(piperazin-l-yl)-3H,4H-thieno[3,2-d]pyrimidin-4-one (69.65 mg, 0.29 mmol) in THF (1.5 ml) under a N2 atmosphere was added 4-oxocyclohexane-l-carbonitrile (31.43 μΐ, 0.27 mmol) and acetic acid (15.33 μΐ, 0.27 mmol). The cloudy mixture was stirred at RT for 35 mins prior to adding STAB (113.58 mg, 0.54 mmol) and continuing stirring for 6 hrs. After which reaction is stopped and purified by column chromatography, eluting with 0% - 12.5% 7N NH3.MeOH in DCM to give 50 mg, 51.6% of 4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2- yl}piperazin-l-yl)cyclohexane-l-carbonitrile as a white foamy solid. METCR1600 (high pH 7 min) M/Z (ES+) 344, Retention time 2.27.
1H NMR (500 MHz, DMSO-d6) δ 1 1.34 (s, 1H), 8.07 - 7.90 (m, 1H), 7.14 - 7.02 (m, 1H), 3.62 - 3.49 (m, 4H), 3.10 - 3.02 (m, 1H), 2.57 - 2.54 (m, 2H), 2.34 - 2.25 (m, 1H), 2.08 - 2.02 (m, 1H), 1.95 - 1.86 (m, 2H), 1.83 - 1.72 (m, 2H), 1.63 - 1.53 (m, 2H), 1.52 - 1.42 (m, 2H), 1.34 - 1.23 (m, lH).
Example 64. (ls,4s)-4-(4-{4-oxo-3H,4H-pyrido[3,2-d]pyrimidin-2-yl}piperazin-l- yl)cycIohexane-l-carbonitrile (62)
Figure imgf000095_0001
[00274] (ls,4s)-4-(4-{4-oxo-3H,4H-pyrido[3,2-d]pyrimidin-2-y]}piperazin- l -yl)cyclohexane- 1-carbonitrile was prepared in a manner analogous to that described above for 4-(4-{4-oxo- 3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin- l-yl)cyclohexane- 1-carbonitrile 53.7 mg (35.3%) as white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 339.2, Retention time 2.22. 1H NMR (500 MHz, DMSO-d6) δ 1 1.46 (s, 1H), 8.41 (dd, J = 4.2, 1.5 Hz, 1 H), 7.65 (dd, J = 8.4, 1.3 Hz, 1H), 7.58 - 7.51 (m, 1H), 3.66 - 3.59 (m, 4H), 3.10 - 3.03 (m, 1H), 2.59 - 2.53 (m, 4H), 2.34 - 2.27 (m, 1H), 1.95 - 1.86 (m, 2H), 1.80 - 1.71 (m, 2H), 1.62 - 1.52 (m, 2H), 1.52 - 1.42 (m, 2H).
Example 65. (ls,4s)-4-(4-{4-oxo-3H,4H-pyrido[3,2-d]pyrimidin-2-yl}piperazin-l- yl)cycIohexane-l-carboxamide (63)
Figure imgf000095_0002
[00275] ( 1 s,4s)-4-(4- {4-oxo-3 H,4H-pyrido[3 ,2-d]pyrimidin-2-yl } piperazin- 1 -y l)cyclohexane- 1-carboxamide was prepapred in a manner analogous to that described above for 6-(4-{4-oxo- 3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)pyridine-3-carboxamide: 27.7 mg (51.6%) as cream powder. METCR1600 (high pH 7 min) M/Z (ES+) 357.2, Retention time 2.04.
1H NMR (500 MHz, DMSO-d6) δ 11.44 (s, 1H), 8.39 - 8.27 (m, 1H), 7.59 (dd, J = 8.4, 1.4 Hz, 1 H), 7.52 - 7.44 (m, 1H), 7.13 (s, 1 H), 6.65 (s, 1H), 3.69 - 3.56 (m, 4H), 2.49 - 2.44 (m,4H), 2.28 - 2.21 (m, 1H), 2.21 - 2.14 (m, 1H), 1.94 - 1.81 (m, 2H), 1.77 - 1.67 (m, 2H), 1.51 - 1.34 (m, 4H). Example 66. 5-(4-{4-oxo-3H,4H-pyrido[3,2-dJpyrimidin-2-yl}piperazin-l-yl)pyridine-2- carboxamide (64)
Figure imgf000096_0001
[00276] 5-(4-{4-oxo-3H,4H-pyrido[3,2-d]pyrimidin-2-yl}piperazin-l-yl)pyridine-2- carboxamide was prepapred in a manner analogous to that described above for 6-(4-{4-oxo- 3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)pyridine-3-carboxamide: 11.7 mg (2.6%) as off-white solid. METCR1600 (high pH 7 min) M/Z (ES+) 352, Retention time 2.5.
1H NMR (500 MHz, DMSO-d6) δ 8.43 (dd, J = 4.2, 1.4 Hz, 1H), 8.33 (d, J = 2.8 Hz, 1H), 7.87 (d, J = 8.8 Hz, 1H), 7.80 - 7.76 (m, 1H), 7.70 - 7.66 (m, 1H), 7.56 (dd, J = 8.3, 4.2 Hz, 1H), 7.45 (dd, J = 8.8, 3.0 Hz, 1H), 7.31 (s, 1H), 3.89 - 3.79 (m, 4H), 3.50 - 3.41 (m, 4H).
Example 67.
Figure imgf000096_0002
tert-buty 1 4-(6-cyanopyridin-3-yl)piperazine- 1 -carboxylate
[00277J The tert-butyl 4-(6-cyanopyridin-3-yl)piperazine-l -carboxylate required for the synthesis was prepared as follows: A mixture of 5-chloro-2-cyanopyridine (1 g, 7.22 mmol), 1- Boc-piperazine (1.61 g, 8.66 mmol), RuPhos (0.34 g, 0.72 mmol) and Cs2C03 (7.05 g, 21.65 mmol) in anhydrous THF (44 mL) was degassed with nitrogen for 10 min with sonication.
RuPhos Precatalyst (0.59 g, 0.72 mmol) was then added and the mixture heated to 85 °C overnight. The reaction mixture was partitioned between EtOAc (50 ml) and water (100 ml). The two phases were separated and the aqueous layer extracted with EtOAc (50 ml). The combined organic extracts were washed with brine (100 ml), dried over MgS04, filtered and concentrated in vacuo. The compound is purified using column chromatography, eluting at a gradient of 17- 92% EtOAc in heptane to yield 1.74g, 77% of the title compound as a yellow powder. METCR1673 Generic 2 minutes M/Z (ES+) 289, Retention time 1.26.
Example 68.5-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yI}piperazin-l-yl)pyrazine-2- carboxamide (65)
Figure imgf000097_0001
[00278] 5-(4-{4-oxo-3H,4H-trdeno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)pyrazine-2- carboxamide was prepapred in a manner analogous to that described above for 6-(4-{4-oxo- 3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)pyridine-3-carboxamide: 22.8 mg (7.9%) as yellow powder. METCR1600 (high pH 7 min) M/Z (ES+) 358.1, Retention time 2.55.
1H NMR (500 MHz, DMSO-d6) δ 1 1.50 (s, 1H), 8.65 (d, J = 1.3 Hz, 1H), 8.31 (d, J = 1.3 Hz, 1H), 8.02 (d, J = 5.2 Hz, 1H), 7.74 (brs, 1H), 7.37 (brs, 1H), 7.11 (d, J = 5.2 Hz, 1H), 3.83 - 3.79 (m, 4H), 3.77 - 3.72 (m, 4H).
Example 69.
Figure imgf000097_0002
tert-butyl 4-(5-cyanopyrazin-2-yl)piperazine-l-carboxylate
[00279] The tert-butyl 4-(5-cyanopyrazin-2-yl)piperazine-l -carboxylate required for the synthesis was prepared in the following manner: To a solution of 5-chloropyrazine-2-carbonitrile (200 mg, 1.43 mmol) and tert-butyl piperazine-l-carboxylate (266.95 mg, 1.43 mmol) in anhydrous DMA (5 ml) under N2 was added DIPEA (748.95 μΐ, 4.3 mmol). The mixture was stirred and heated at 150°C in a microwave for 90 mins. The mixture was allowed to cool to RT, prior to adding water until a significant amount of precipitate was observed. This was filtered under vacuum, washed with water and dried to afford 363mg, 87% of titled compound as a light brown solid. METCR1673 Generic 2 minutes M/Z (ES+) 233.9, Retention time 1.28.
1H NMR (500 MHz, DMSO-d6) δ 8.35 (d, J = 1.3 Hz, 1H), 8.12 (d, J = 1.3 Hz, 1H), 3.78 - 3.71 (m, 4H), 3.61 - 3.55 (m, 4H), 1.49 (s, 9H).
Example 70. N-(3-h drox prop l)-4-(4-{4-oxo-3H,4H-thieno [3,2-d] pyrimidin-2- yl}piperazin-l-yl)benzamide (66)
Figure imgf000098_0001
[00280] General procedure B provided 60mg (12%) of N-(3-hydroxypropyl)-4-(4-{4-oxo- 3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)benzamide as a beige powder. MET-uPLC- AB-101 (7 min, low pH) M/Z (ES+) 414.2, Retention time 2.07.
1H NMR (500 MHz, DMSO-d6) δ 11.49 (s, 1H), 8.17 (t, J = 5.6 Hz, 1H), 8.02 (d, J = 5.2 Hz, 1H), 7.75 (d, J= 8.9 Hz, 2H), 7.11 (d, J = 5.2 Hz, 1H), 7.00 (d, J = 9.0 Hz, 2H), 4.46 (t, J = 5.3 Hz, 1H), 3.78 - 3.71 (m, 4H), 3.45 (q, J = 6.2 Hz, 2H), 3.39 - 3.34 (m, 4H), 3.30 - 3.26 (m, 2H), 1.66 (p, J = 6.5 Hz, 2H).
Example 71.
Figure imgf000099_0001
tert-but l 4- {4-[(3- ydroxypropyl)carbamoyl]phenyl } piperazine- 1 -carboxylate
[00281] The tert-butyl 4-{4-[(3-hydroxypropyl)carbamoyl]phenyl}piperazine-l -carboxylate required for the synthesis was prepared as follows: 4-{4-[(tert-butoxy)carbonyl]piperazin-l- yl}benzoic acid (1 g, 3.26 mmol) was dissolved in DMF (15ml) and DIPEA (1.62 ml, 9.79 mmol) and HATU (1489.38 mg, 3.92 mmol) were added to the mixture and stirred at room temperature for 30 minutes. 3-aminopropan-l-ol (0.5 ml, 6.53 mmol) is added and the reaction is stirred for further 2 hours. Mixture is diluted with water (10ml) and extracted with ethyl acetate (10x3 ml), the organic layers are combined and washed with water (2x10ml), dried over magnesium sulphate, filtered and reduced in vacuo. The remaining residue is sonicated with heptane and DCM to remove the remaining traces of DMF, yielding l .lg, 93% of the title compound as an orange solid. METCR1673 Generic 2 minutes M/Z (ES+) 364.15, Retention time 1.12.
1H NMR (500 MHz, DMSO-d6) δ 8.16 (t, J = 5.6 Hz, 1H), 7.73 (d, J = 8.9 Hz, 2H), 6.96 (d, J = 9.0 Hz, 2H), 4.46 (t, J = 5.3 Hz, 1H), 3.45 (q, J = 6.2 Hz, 6H), 3.31 - 3.26 (m, 2H), 3.27 - 3.21 (m, 4H), 1.66 (p, J = 6.5 Hz, 2H), 1.43 (s, 9H).
Example 72. N-[3-(morpholin-4-yl)propyl]-4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2- yl}piperazin-l-yl)benzamide (67)
Figure imgf000099_0002
[00282] N-[3-(morpholin-4-yl)propyl]-4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2- yl}piperazin-l-yl)benzamide was prepapred in a manner analogous to that described above for N-(3-hydroxypropyl)-4-(4-{4-oxo-3H,4H-tWeno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)benzamide 33mg (11.2%) as a beige powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 483.2, Retention time 1.41.
1H NMR (500 MHz, DMSO-d6) 5 1 1.51 (s, 1H), 8.21 (t, J = 5.6 Hz, 1H), 8.02 (d, J = 5.2 Hz, 1H), 7.74 (d, J= 8.9 Hz, 2H), 7.11 (d, J = 5.2 Hz, 1H), 7:01 (d, J = 9.0 Hz, 2H), 3.79 - 3.72 (m, 4H), 3.60 - 3.54 (m, 4H), 3.38 - 3.34 (m, 4H), 3.29 - 3.23 (m, 2H), 2.38 - 2.30 (m, 6H), 1.71 - 1.62 (m, 2H).
Example 73. N-[3-(dimethylamino)propyl]-4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2- yl}piperazin-l-yl)benzamide (68)
Figure imgf000100_0001
[00283] N-[3-(dimethylamino)propyl]-4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2- yl}piperazin-l-yl)benzamide was prepapred in a manner analogous to that described above for N-(3-hydroxypropyl)-4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)benzamide 18.1 mg (7.5%) as brown powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 221.1, Retention time 1.38.
1H NMR (500 MHz, DMSO-d6) δ 11.47 (s, 1H), 8.31 (t, J = 5.6 Hz, 1H), 8.02 (d, J = 5.2 Hz, 1H), 7.74 (d, J = 8.9 Hz, 2H), 7.10 (d, J = 5.2 Hz, 1H), 7.01 (d, J = 9.0 Hz, 2H), 3.79 - 3.71 (m, 4H), 3.39 - 3.34 (m, 4H), 2.76 - 2.67 (m, 2H), 1.82 - 1.71 (m, 2H).
Example 74. N-methyl-4-(4-{thieno[3,2-b]pyridin-5-yl}piperazin-l-yl)benzamide (69)
Figure imgf000100_0002
[00284] General procedure B provided 22 mg (10.6%) of N-methyl-4-(4-{thieno[3,2- b]pyridin-5-yl}piperazin-l-yl)benzamide as a brown powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 353, Retention time 1.81.
1H NMR (500 MHz, DMSO-d6) δ 8.21 - 8.17 (m, 1H), 8.15 (q, J = 4.2 Hz, 1H), 7.94 (d, J = 5.4 Hz, 1H), 7.75 (d, J = 8.9 Hz, 2H), 7.30 (dd, J = 5.4, 0.5 Hz, 1H), 7.05 (d, J = 9.1 Hz, 1H), 7.02 (d, J = 9.0 Hz, 2H), 3.75 - 3.70 (m, 4H), 3.43 - 3.39 (m, 4H), 2.76 (d, J = 4.5 Hz, 3H).
Example 75. N-methyI-4-(4-{thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)benzamide (70)
Figure imgf000101_0001
[00285] General procedure B provided 43.6 mg (54.1%) of N-methyl-4-(4-{thieno[3,2- d]pyrimidin-2-yl}piperazin-l-yl)benzamide as a cream colored solid. METCR1600 (high pH 7 min) M/Z (ES+) 354.2, Retention time 2.69.
1H NMR (500 MHz, DMSO-d6) 6 9.10 (d, J = 0.6 Hz, 1H), 8.27 (d, J = 5.4 Hz, 1H), 8.19 - 8.13 (m, 1H), 7.74 (d, 2H), 7.28 (dd, J = 5.4, 0.6 Hz, 1H), 7.01 (d, J = 9.0 Hz, 2H), 3.96 - 3.89 (m, 4H), 3.37 - 3.36 (m, 4H), 2.75 (d, J = 4.5 Hz, 3H).
Example 76. N-methyl-4-(4-{7-oxo-6H,7H-thieno[3,2-b]pyridin-5-yl}piperazin-l- yl)benzamide (71)
Figure imgf000101_0002
[00286] General procedure B provided 4.6 mg (4.6%) of N-methyl-4-(4-{7-oxo-6H,7H- thieno[3,2-b]pyridin-5-yl}piperazin-l-yl)benzamide as a brown solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 369.2, Retention time 1.55.
1H NMR (500 MHz, DMSO-d6) δ 8.15 (q, J = 4.6 Hz, 1H), 7.81 (d, J = 5.3 Hz, 1H), 7.73 (d, J = 8.9 Hz, 2H), 7.19 (d, J = 5.3 Hz, 1H), 7.00 (d, J = 8.9 Hz, 2H), 3.57 - 3.53 (m, 4H), 3.17 (s, 2H), 2.75 (d, J = 4.5Hz, 3H).
Example 77. N-methyl-4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l- yl)benzamide (72)
Figure imgf000102_0001
[00287] General procedure B provided 4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2- yl}piperazin-l-yl)benzonitrile and subsequent hydrolysis and amide formation using standard conditions for such transformations gave the desired product, N-methyl-4-(4-{4-oxo-3H,4H- thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)benzamide. 34.8 mg (44.5%) as white powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 370.2, Retention time 1.89.
1H NMR (500 MHz, DMSO-d6) δ 11.50 (s, 1H), 8.14 (q, J = 4.2 Hz, 1H), 8.00 (d, J - 5.2 Hz, 1H), 7.76 - 7.70 (m, 2H), 7.10 (d, J = 5.2 Hz, 1H), 7.02 - 6.97 (m, 2H), 3.79 - 3.70 (m, 4H), 3.40 - 3.33 (m, 4H), 2.74 (d, J = 4.5 Hz, 3H).
Example 78. N,N-dimethyI-4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l- yl)benzamide (73)
Figure imgf000103_0001
[00288] N,N-dimethyl-4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l- yl)benzamide was prepared in a manner analogous to that described above for N-methyl-4-(4-{4- oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-yl)benzamide: 48.7 mg (56.9%) as white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 384, Retention time 2.09.
1H NMR (500 MHz, DMSO-d6) 5 1 1.49 (s, 1H), 8.00 (d, J = 5.2 Hz, 1H), 7.36 - 7.24 (m, 2H),
7.10 (d, J = 5.2 Hz, 1H), 7.02 - 6.95 (m, 2H), 3.80 - 3.68 (m, 4H), 3.35 - 3.31 (m, 4H), 2.95 (s,
6H).
Example 79. N-methyl-6-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yI}piperazin-l- yl)pyridazine-3-carboxamide (74)
Figure imgf000103_0002
[00289] N-methyl-6-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l- yl)pyridazine-3-carboxamide was prepared in a manner analogous to that described above for N- methyl-4-(4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yI}piperazin-l-yl)benzamide: 0.01 g (6.2%) as off white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 372.1, Retention time 1.64.
1H NMR (500 MHz, DMSO-d6) 5 11.50 (s, 1H), 8.79 (q, J = 4.6 Hz, 1H), 8.03 (d, J = 5.2 Hz, 1H), 7.87 (d, J= 9.5 Hz, 1H), 7.40 (d, J = 9.6 Hz, 1H), 7.12 (d, J = 5.2 Hz, 1H), 3.85 - 3.81 (m, 4H), 3.80 - 3.76 (m, 4H), 2.83 (d, J = 4.8 Hz, 3H). Example 80.
Figure imgf000104_0001
tert-butyl 4-(6-cyanopyridazin-3-yl)piperazine-l-carboxylate
[00290] The tert-butyl 4-(6-cyanopyridazin-3-yl)piperazine-l-carboxylate required for the synthesis was prepared as follows: 6-chloropyridazine-3-carbonitrile (600 mg, 4.3 mmol) and tert-butyl piperazine-l-carboxylate (1803.69 μΐ, 10.75 mmol) were dissolved in MeCN (20ml) and the reaction is heated in a pressure tube to 60C for one hour. Solid had precipitated during reaction and LCMS indicated no remaining starting material. Reaction is cooled down to room temperature and the solid is filtered off. This compound is washed with water and left to dry. 1033 mg (83%) as orange powder. METCR1673 Generic 2 minutes M/Z (ES+) 233.95, Retention time 1.21.
1H NMR (500 MHz, DMSO-d6) δ 7.90 (d, J = 9.7 Hz, 1H), 7.35 (d, J = 9.7 Hz, 1H), 3.85 - 3.69 (m, 4H), 3.60 - 3.38 (m, 4H), 1.44 (s, 9H).
Example 81. N,3-dimethyI-4-(4-{4-oxo-3H,4H-thieno [3,2-d] pyrimidin-2-yl} piperazin-1- yl)benzamide (75)
Figure imgf000104_0002
[00291J General procedure B provided N,3-dimethyl-4-(4-{4-oxo-3H,4H-thieno[3,2- d]pyrimidin-2-yl}piperazin-l-yl)benzamide: 0.08 g (23.5%) as an off white powder. MET- uPLC-AB-101 (7 min, low pH) M/Z (ES+) 384.1 , Retention time 3.05.
1H NMR (500 MHz, DMSO-d6) δ 1 1.45 (br s, 1H), 8.24 (q, J = 4.2 Hz, 1H), 8.00 (d, J = 5.2 Hz, 1H), 7.67 (d, J = 1.9 Hz, 1H), 7.63 (dd, J = 8.3, 2.1 Hz, 1H), 7.09 (d, J = 5.2 Hz, 1H), 7.06 (d, J = 8.3 Hz, 1 H), 3.85 -3.64 (m, 4H), 3.02 - 2.89 (m, 4H), 2.75 (d, J = 4.5 Hz, 3H), 2.32 (s, 3H)
Example 82. 2-[4-(l-methylcyclohexyl)piperazin-l-yl]-4H-thieno[3,2-d] [l,3]thiazin-4-one
Figure imgf000105_0001
tert- utyl 4-(l-methylcyclohexyl)piperazine-l-carboxylate
[00292] A mixture of cyclohexanone (0.4 g, 4.08 mmol), Boc-piperazine (0. 83 g, 4.48 mmol) and 2H- l,2,3-triazole (0.28 ml, 4.89 mmol) in anhydrous toluene (3ml) was heated to reflux in a 3 neck flask with azeotropic removal of water overnight. The mixture was allowed to cool to rt and then added to a solution of 3M MeMgCl in THF (5.43 ml) via cannula over 20min, keeping the internal temperature below 24°C. The mixture was stirred at rt for 3h. The mixture was then poured slowly onto an aqueous NH4CI solution (20%, 20ml), keeping the temperature below 30°C. The layers were separated and the aqueous layer extracted with EtOAc, The combined organic extracts were washed with 2M aqueous NaOH, water, dried over Na2S04, filtered and concentrated in vacuo. Purification of the crude material by column chromatography using a gradient of 0-10% MeOH in DCM gave 0.73 g (63%) of the title compound as orange oil. METCR1673 (Generic 2 minutes, low pH) m/z 283.05 (ES+), Retention time 1 .29 min. 1H NMR (500 MHz, DMSO-d6) δ 3.29 - 3.23 (m, 1H), 2.42 - 2.31 (m, 4H), 1.71 - 1.62 (m, 4H), 1.56 (ddd, J =15.0, 9.2, 4.5 Hz, 1H), 1.48 - 1.41 (m, 1H), 1.39 (s, 9H), 1.31 - 1.23 (m,3H), 1.22 - 1.15 (m, 2H), 0.78 (s, 3H).
1- (l-methylcycIohexyl)piperazine
[00293] A stirred mixture of tert-butyl 4-(l-methylcyclohexyl)piperazine-l-carboxylate (0.73 g, 2.58 mmol), MeOH (10 ml) and 5M HC1 in MeOH (12.91 ml) was stirred at rt overnight. The volatiles were then evaporated in vacuo and the residue re-dissolved in 4N HC1 in dioxane. The mixture was stirred at rt for 4 h and the solvents then removed in vacuo. The resulting residue was triturated with EtOAc and the solid obtained partitioned between 2M NaOH and DCM. The organic phase was dried over Na2S04, filtered and concentrated in vacuo to give 0.27 g (58%) of the title compound as a yellow gum. METCR1673 (Generic 2 minutes, low pH) m/z 183.05 (ES+), Retention time: solvent front.
1H NMR (250 MHz, DMSO-d6) δ 2.71 - 2.57 (m, 4H), 2.38 - 2.26 (m, 4H), 1.75 - 1.04 (m, 10H), 0.78 (s, 3H).
3-{[4-(l-methylcycIohexyl)piperazine-l-carbothioyl]amino}thiophene-2-carboxylate
[00294] l-(l-methylcyclohexyl)piperazine (0.15 g, 0.83 mmol) and methyl 3- isothiocyanatothiophene-2-carboxylate (0.15 g, 0.75 mmol) were dissolved in anhydrous DCM and stirred at rt under N2 for 4 h. The reaction mixture was then washed with 1M aqueous HC1, water, dried over Na2S04, filtered and concentrated in vacuo. The crude material was slurried in DCM and the solid obtained collected by filtration to give 0.11 g (40%) of the title compound as a white solid. METCR1673 (Generic 2 minutes, low pH) m/z 385.05 (ES+), Retention time 0.97 min.
1H NMR (500 MHz, DMSO-d6) δ 10.19 (s, 1H), 8.14 (d, J = 5.4 Hz, 1H), 7.87 (d, J = 5.4 Hz, 1H), 4.87 - 4.72 (m,2H), 3.85 (s, 3H), 3.80 - 3.72 (m, 2H), 3.68 (dd, J = 13.2 Hz, 2H), 3.26 - 3.15 (m, 2H), 1.94- 1.86 (m, 2H), 1.78 - 1.68 (m, 4H), 1.66 - 1.60 (m, 1H), 1.46 - 1.35 (m, 2H), 1.32 (s, 3H), 1.18 - 1.04 (m, 1H).
2- [4-(l-methylcyclohexyl)piperazin-l-yl]-4H-thieno[3,2-d] [l,3]thiazin-4-one
[00295] Methyl 3 - { [4-( 1 -methylcyclohexy l)piperazine- 1 -carbothioyljamino }thiophene-2- carboxylate (0.1 1 g, 0.29 mmol) was stirred in H2SO4 (3 ml) at rt for 3 h and then poured onto ice. The pH of the mixture was adjusted to pH 9 with 6N aqueous NaOH and the mixture extracted with EtOAc. The organic phase was dried over Na2S04, filtered and concentrated in vacuo. The material was re-dissolved in H2SO4 and the mixture stirred at for a further 3h. The crude material was isolated as per the same quench and work-up procedure above to give the 18 (18%) of the title compound as a yellow solid. MET-uPLC-AB-101 (7 min, low pH) m/z (ES+) 350.1, Retention time 1.77 min.
lH NMR (500 MHz, DMSO- 6) δ 7.99 (d, J= 5.3 Hz, 1H), 7.10 (d, J= 5.3 Hz, 1 H), 3.91 - 3.72 (m, 4H), 2.90 - 2.66 (m, 4H), 1.81 - 1.68 (m, 4H), 1 .51 - 1.30 (m, 6H), 0.99 (s, 3H).
Example 83. 4-[4-(l-benzothiophen-5-yl)piperazin-l-yl]benzamide (77)
Figure imgf000107_0001
4-[4-(l-benzothiophen-5-yl)piperazin-l-yl] benzoic acid
[00296] A sealed tube was charged with 5-bromo-l-benzothiophene (147.55 μΐ, 1.03 mmol), 4-(piperazin-l -yJ)benzoic acid (255.51 mg, 1.24 mmol), RuPhos (24.09 mg, 0.05 mmol), CS2CO3 (198.26 μΐ, 2.48 mmol) and anhydrous THF (7 ml). The reaction mixture was degassed by bubbling nitrogen with sonication for 30 min, prior to adding RuPhos Precatalyst (42.17 mg, 0.05 mmol) and the reaction mixture stirred at 70°C for 40 h. The mixture was allowed to cool to rt and the precipitate filtered and washes with water. The solid was triturated with heptane and DCM, and then filtered to give the title compound 0.07 g (17%) as a white solid. METCR1673 (Generic 2 minute, low pH), m/z (ES+) 250, Retention time 1.41 min.
l H NMR (500 MHz, DMSO-d6) δ 7.10 (d, J = 8.8 Hz, 2H), 6.97 (d, J = 8.8 Hz, 1H), 6.71 (d, J = 5.4 Hz, 1 H), 6.65 (d, J = 2.2 Hz, 1H), 6.49 (d, J = 5.4 Hz, 1H), 6.40 (dd, J = 8.8, 2.3 Hz, 1H), 6.20 (d, J = 8.8 Hz, 2H), 2.70 - 2.63 (m, 4H), 2.58 - 2.55 (m, 4H), 2.43 - 2.40 (m, 1H), 2.19 - 2.16 (m, 1H).
4-[4-(l-benzothiophen-5-yl)piperazin-l-yl]benzamide
[00297] To a stirring solution of 4-[4-( l -benzothiophen-5-yl)piperazin-l-yl]benzoic acid (70 mg, 0.21 mmol) and HATU ( 102.24 mg, 0.27 mmol) in DMF (12 ml) under a N2 atmosphere was added DIPEA (0.1 1 ml, 0.68 mmol). The mixture was stirred at RT for lh before adding 7N Ammonia in MeOH (0.07 ml) and the reaction left stirring overnight. The mixture was concentrated in vacuo before the solid triturated with water, MeOH and DCM and residual solvent removed using a genevac system to the title compound as an off white solid. (3 mg, 4 MET-uPLC-AB-101 (7 min, low pH) m/z (ES+) 338.1, Retention time 2.91.
1H NMR (500 MHz, DMSO- 6) 6 7.84 (d, J= 8.8 Hz, 1H), 7.79 (d, J= 8.9 Hz, 2H), 7.72 (s, 1H), 7.68 (d,J= 5.4 Hz, 1H), 7.42 (d, J= 2.3 Hz, 1H), 7.34 (d, J = 5.4 Hz, 1H), 7.21 (dd, J = 8.9, 2.3 Hz, 1H), 7.05 - 7.01 (m, 3H), 3.47 - 3.43 (m, 4H).
Example 84.2-[4-(pyridin-2-yl)piperazin-l-yl]quinazolin-4-ol (78)
Figure imgf000108_0001
[00298] 2-chloroquinazolin-4-ol (100 mg, 0.55 mmol) and l-(pyridin-2-yl)piperazine (99.42 mg, 0.61 mmol) were suspended in ethanol (3 ml) in a pressure tube. The reaction was heated to 90°C for 6h. On completion the solution was filtered and a white solid was collected, which was washed with ethanol and dried under vacuum to give 81 mg (47.6%) of the title compound as a white powder without further purification. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 308.1 , Retention time 1.07.
1H NMR (250 MHz, DMSO-d6) δ 1 1.08 (s, 1H), 8.21 - 8.12 (m, 1H), 8.00 - 7.90 (m, 1H), 7.67 - 7.51 (m, 2H), 7.40 - 7.27 (m, 1H), 7.22 - 7.10 (m, 1H), 6.86 (d, J = 8.6 Hz, 1H), 6.68 (dd, J = 7.0, 4.9 Hz, 1H), 3.86 - 3.74 (m, 4H), 3.71 - 3.59 (m, 4H).
Example 85. 4-methoxy-2-[4-(pyridin-2-yl)piperazin-l-yl]quinazoline (79) and 3-methyl-2- [4-(pyridin-2-yl)piperazin-l-yl]-3,4-dihydroquinazolin-4-one (80)
Figure imgf000108_0002
[00299] 2-[4-(pyridin-2-yl)piperazin-l-yl]quinazolin-4-ol (55 mg, 0.18 mmol), prepared as described above, was dissolved in N,N-Dimethylformamide (2 ml) and NaH (60%, 8.59 mg, 0.21 mmol) was added. To this was added iodomethane (13.37 μΐ, 0.21 mmol). The reaction was stirred at r.t for 2h, and then diluted with 10ml water and extracted with EtOAc (3 x 10ml). The combined organic fractions were washed with water and brine, then dried over Na2S04, filtered and evaporated. Purification by flash column chromatography gave 4-methoxy-2-[4-(pyridin-2- yl)piperazm-l-yl]quinazoline 6 mg (10.4%) as an off-white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 322, Retention time 1.19.
1H NMR (500 MHz, DMSO-d6) 6 8.17 - 8.12 (m, 1H), 7.88 (dd, J = 8.1, 1.1 Hz, 1H), 7.66 (ddd, J = 8.5, 7.0, 1.5 Hz, 1H), 7.57 (ddd, J = 8.9, 7.1, 2.0 Hz, 1H), 7.46 (d, J = 8.3 Hz, 1H), 7.22 - 7.13(m, 1H), 6.89 (d, J = 8.6 Hz, 1H), 6.67 (dd, J = 6.7, 5.0 Hz, 1H), 4.10 (s, 3H), 4.00 - 3.90 (m, 4H), 3.66 - 3.56 (m,4H).
[00300] 3-methyl-2-[4-(pyridin-2-yl)piperazin-l-yl]-3,4-dihydroquinazolin-4-one 12 mg (20.9%) as an off-white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 322, Retention time 1.
1H NMR (500 MHz, DMSO-d6) δ 8.15 (dd, J = 4.9, 1.3 Hz, 1H), 8.04 (dd, J = 8.0, 1.3 Hz, 1H), 7.72 (ddd, J = 8.5, 7.1, 1.6 Hz, 1H), 7.64 - 7.57 (m, 1H), 7.48 (d, J = 7.8 Hz, 1H), 7.38 - 7.33 (m, 1H), 6.93 (d, J =8.5 Hz, 1H), 6.71 (dd, J = 6.8, 5.1 Hz, 1H), 3.70- 3.64 (m, 4H), 3.54 (s, 3H), 3.31 - 3.27 (m, 4H).
Example 86.
Route D
Figure imgf000110_0001
TFA
DCM
Figure imgf000110_0002
RhPhos Cs2C03 Pd(dba)2 THF
Figure imgf000110_0003
LiOH
MeOH/Water
Figure imgf000110_0004
HATU D F DIPEA Amine
Figure imgf000110_0005
[00301] tert-butyl 4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazine-l-carboxylate was prepared using general procedure B.
[00302] To a stirred suspension of sodium hydride 60% in mineral oil (0.15 g, 6.24 mmol) in anhydrous THF (5 mL) in a sealable pressure tube under nitrogen in an ice bath was added dropwise a suspension of tert-butyl 4-{4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazine-l- carboxylate (1.4 g, 4.16 mmol) in anhydrous THF (lOmL) . The mixture was stirred at rt for one hour after which iodomethane (388.61 μΐ, 6.24 mmol) was added. The tube was then sealed and the mixture heated at reflux (85 °C) overnight. On completion the precipitate was filtered under vacuum, washed with THF and air dried to give a dark yellow solid.
[00303] The THF filtrate was concentrated in vacuo and purified by reverse phase column chromatography to afford 248 mg (17.7%) of 4-{3-methyl-4-oxo-3H,4H-thieno[3,2-d]pyrimidin- 2-yl}piperazine-l-carboxylate (A) and 144 mg (10.3%) of 4-{4-methoxythieno[3,2-d]pyrimidin- 2-yl}piperazine-l-carboxylate (B) as fluffy white solids. 4-{3-methyl-4-oxo-3H,4H-thieno[3,2- d]pyrimidin-2-yl}piperazine-l-carboxylate (A): METCR1410 (2 min, low pH), m/z (ES+) 351.1, Retention time 0.94 min. 1H NMR (500 MHz, DMSO-d6) δ 8.06 (d, J = 5.4 Hz, 1H), 7.37 (d, J = 5.4 Hz, 1H), 3.69 (s, 3H), 3.51 - 3.45 (m, 4H), 3.22 - 3.17 (m, 4H), 1.43 (s, 9H). 4-{4- methoxythieno[3,2-d]pyrimidin-2-yl}piperazine-l-carboxylate (B): METCR1735 (1.5 min, high pH), m/z (ES+) 351.1, Retention time 1.95 min. 1H NMR (500 MHz, DMSO-d6) δ 8.1 1 (d, J = 5.3 Hz, 1H), 7.22 (d, J = 5.3 Hz, 1H), 4.04 (s, 3H), 3.81 - 3.70 (m, 4H), 3.46 - 3.39 (m, 4H), 1.43 (s, 9H).
[00304] The solid from the THF trituration was washed with water (to remove remaining inorganic material) and the remaining solid was dissolved in MeOH and evaporated in vacuo to afford 524 mg (34.8%) of 4-{ l-methyl-4-oxo-m,4H-thieno[3,2-d]pyrimidin-2-yl}piperazine-l- carboxylate (C) as an off white solid. 4-{ l-methyl-4-oxo-lH,4H-thieno[3,2-d]pyrimidin-2- yl}piperazine-l-carboxylate (C): METCR1735 (1.5 min, high pH), m/z (ES+) 351.1, Retention time 1.61 min. 1H NMR (500 MHz, DMSO-d6) δ 8.09 (d, J = 5.2 Hz, 1H), 7.24 (d, J = 5.2 Hz, 1H), 3.50 (s, 3H), 3.50 - 3.48 (m, 4H), 3.16 - 3.04 (m, 4H), 1.43 (s, 9H).
[00305] To a pale yellow solution of 4-{3-methyl-4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2- yl}piperazine-l-carboxylate (248 mg, 0.71 mmol) in DCM (10 ml) was added TFA (1.08 ml, 14.15 mmol) dropwise. The resulting brown solution was stirring for 90 mins. On completion the reaction was concentrated in vacuo, prior to passing it through a 2g NH2 cartridge. Solvent was evaporated and the resulting solid dissolved in minimal DCM which was passed through an SCX cartridge, eluting the desired product with 7 NH3 in MeOH to afford 163mg (92%) of 3- methyl-2-(piperazin-l-yl)-3H,4H-thieno[3,2-d]pyrimidin-4-one (D) as a cream solid. METCR1410 (2 min, low pH), m/z (ES+) 251.05, Retention time 0.17min. 1H NMR (500 MHz, DMSO-d6) δ 8.07 (d, J = 5.2 Hz, 1H), 7.23 (d, J = 5.2 Hz, 1H), 3.48 (s, 3H), 3.11 - 2.95 (m, 4H), 2.90 - 2.78 (m, 4H).
[00306] Compounds (E) and (F) were prepared in an analogous manner.
[00307] A mixture of 3-methyl-2-(piperazin-l-yl)-3H,4H-thieno[3,2-d]pyrimidin-4-one (161.29 mg, 0.64 mmol), methyl 5-bromopyridine-2-carboxylate (63 mg, 0.29 mmol), RuPhos (125.28 mg, 0.27 mmol) and Cs2C03 (128.89 μΐ, 1.61 mmol) in anhydrous THF (5 ml) was degassed in a sealable pressure tube prior to adding Pd2(dba)3 (245.85 mg, 0.27 mmol). The tube was then sealed and the mixture stirred at reflux (85oC) for 3 hrs. On completion the mixture allowed to cool to RT, before removing the solvent in vacuo and partitioning the residue between water (15 ml) and DCM (15 ml). The organic layer was separated and aqueous extracted further with DCM (5 x 10 ml). Combined organics were dried over Na2S04, filtering and concentrating in vacuo. Purified reverse phase column chromatography to afford 84 mg, (37%) of 5-(4-{3-methyl-4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l-:yl)pyridine-2- carboxylate (G) as a yellow solid. METCR1410 (2 min, low pH), m/z (ES+) 385.85, Retention time 0.95min. 1H NMR (250 MHz, DMSO-d6) δ 8.45 (d, J = 2.9 Hz, 1H), 8.10 (d, J = 5.2 Hz, 1H), 7.92 (d, J = 8.8 Hz, 1H), 7.43 (dd, J = 9.0, 3.0 Hz, 1H), 7.27 (d, J = 5.2 Hz, 1H), 3.82 (s, 3H), 3.62 - 3.55 (m, 4H), 3.55 (s, 3H),3.39 - 3.33 (m, 4H).
[00308] Compounds (H) and (J) were prepared in an analogous manner.
[00309] To a stirring yellow suspension of 5-(4-{3-methyl-4-oxo-3H,4H-thieno[3,2- d]pyrimidin-2-yl}piperazin-l-yl)pyridine-2-carboxylate (91%, 84 mg, 0.2 mmol) in MeOH (2.1 ml) was added a solution of LiOH (38 mg, 1.6 mmol) in Water (2.1 ml) dropwise. This was stirred at RT for 48 hrs. On completion MeOH was evaporated and the resulting aqueous was acidified to pH2-3 using 3M HC1. Precipitate was filtered, washed with water and dried under vacuum to give 54.6mg (70%) of 5-(4-{3-methyl-4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2- yl}piperazin-l-yl)pyridine-2-carboxylic acid (K) as a yellow solid. METCR1410 (2 min, low pH), m/z (ES+) 371.95, Retention time 0.79min. 1H NMR (500 MHz, DMSO-d6) δ 8.43 (d, J = 2.9 Hz, 1H), 8.10 (d, J = 5.2 Hz, 1H), 7.91 (d, J = 8.8 Hz, 1H), 7.43 (dd, J = 8.9, 3.0 Hz, 1H), 7.27 (d, J = 5.2 Hz, lH), 3.59 - 3.55 (m, 4H), 3.55 (s, 3H). note:4 piperizine protons are under water peak in NMR.
[00310] Compounds (L) and (M) were prepared in an analogous manner.
N-(3-hydroxypropyl)-5-(4-{3-methyl-4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l- yl)pyridine-2-carboxamide (81)
Figure imgf000113_0001
[00311] A mixture of 5-(4-{3-methyl-4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l- yl)pyridine-2-carboxylic acid (95%, 27.6 mg, 0.07 mmol), HATU (32.21 mg, 0.08 mmol), DMF (0.8 ml) and DIPEA (35 μΐ, 0.21 mmol) was stirred for 40 mins, prior to adding propanolamine (10.8 μΐ, 0.14 mmol) and continuing stirring for 3 hr. On completion the mixture was diluted with water (2 ml). This was extracted with DCM (3 x 3 ml) and dried over Na2S04, filtering and evaporated to give a white brown solid. Purified by HPLC preparative chromatography to afford 21.1mg (40%) of thye title compound as a white solid. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 429.1 , Retention time 2.06 min.
1H NMR (500 MHz, DMSO-d6) δ 8.49 (t, J - 6.0 Hz, 1H), 8.34 (d, J = 2.8 Hz, lH), 8.10 (d, J = 5.2 Hz, 1H), 7.87 (d, J = 8.8 Hz, 1H), 7.48 (dd, J = 8.8, 2.9 Hz, 1H), 7.27 (d, J = 5.2 Hz, 1H), 4.52 (t, J = 5.3 Hz, 1H), 3.55 (s, 3H), 3.54 - 3.50 (m, 4H), 3.48 - 3.44 (m, 2H), 1.71 - 1.62 (m, 2H).
[00312] The following compounds were prepared in an analogous manner to that described above (all reagents whose synthesis is not described in the text are of commercial origin or can be prepared by methods known in the chemical literature)
N-(3-hydroxypropyl)-5-(4-{l-methyl-4-oxo-lH,4H-thieno[3,2-d]pyrimidin-2-yl}piperazin-l- yl)pyridine-2-carboxamide (82)
Figure imgf000114_0001
[00313] 22 mg (33.5 %) as a brown gum. METCR1600 (7 min, high pH) M/Z (ES+) 429.1, Retention time 2.68 min.
1H NMR (500 MHz, DMSO-d6) δ 8.49 (t, J = 6.0 Hz, 1H), 8.34 (d, J = 2.8 Hz, 1H), 8.08 (d, J = 5.4 Hz, 1H), 7.87 (d, J = 8.8 Hz, 1H), 7.48 (dd, J = 8.8, 2.9 Hz, 1H), 7.39 (d, J = 5.4 Hz, 1H), 4.52 (t, J = 5.3 Hz, 1H), 3.74 (s, 3H), 3.53 - 3.49 (m, 4H), 3.48 - 3.44 (m, 2H), 3.43 - 3.39 (m, 4H), 3.37 - 3.33 (m, 2H), 1.69 - 1.63 (m, 2H).
N-(3-hydroxypropyl)-5-(4-{4-methoxythieno[3^-d]pyrimidin-2-yl}piperazin-l-yl)pyridine-
2-carboxamide (83)
Figure imgf000114_0002
[00314] 26mg (76.9%) as a yellow foam. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 424.2, Retention time 1.69 min.
1H NMR (500 MHz, DMSO-d6) δ 8.52 (dd, J = 4.1, 1.5 Hz, 1H), 8.48 (t, J = 6.0 Hz, 1H), 8.35 (d, J = 2.9 Hz, 1H), 7.87 (d, J = 8.7 Hz, 1H), 7.84 (dd, J = 8.6, 1.5 Hz, 1H), 7.66 (dd, J = 8.6, 4.1 Hz, 1H), 7.47 (dd, J = 8.8, 2.9 Hz, 1H), 4.52 (t, J = 5.3 Hz, 1H), 4.11 (s, 3H), 4.06 - 3.99 (m, 4H), 3.52 - 3.42 (m, 6H), 3.37 - 3.32 (m, 2H), 1.70 - 1.62 (m, 2H).
5-(4-{4-methoxythieno[3,2-d]pyrimidin-2-yI}piperazin-l-yl)-N-methylpyridine-2- carboxamide (84)
Figure imgf000115_0001
[00315] 5.2mg (18.8%) as a white powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 385.1, Retention time 2.52 min.
1H NMR (500 MHz, DMSO-d6) δ 8.41 (q, J = 4.9 Hz, 1H), 8.33 (d, J= 2.9 Hz, 1H), 8.12 (d, J = 5.3 Hz, 1H), 7.86 (d, J = 8.8 Hz, 1H), 7.46 (dd, J = 8.8, 2.9 Hz, 1H), 7.24 (d, J = 5.3 Hz, 1H), 4.06 (s, 3H), 3.99 - 3.91 (m, 4H), 3.49 - 3.43 (m, 4H), 2.79 (d, J = 4.8 Hz, 3H).
N-methyl-5-(4-{3-methyl-4-oxo-3H,4H-thieno[3,2-d]pyrimidin-2-yI}piperazin-l- yl)pyridine-2-carboxamide (85)
Figure imgf000115_0002
[00316] 1 1.4mg (45%) as a white powder. MET-uPLC-AB-101 (7 min, low pH) M/Z (ES+) 385, Retention time 2.22 min.
1H NMR (500 MHz, DMSO-d6) δ 8.43 (q, J = 4.9 Hz, 1H), 8.34 (d, J = 2.8 Hz, 1H), 8.10 (d, J = 5.2 Hz, IH), 7.87 (d, J = 8.8 Hz, 1H), 7.48 (dd, J = 8.8, 2.9 Hz, 1H), 7.27 (d, J = 5.2 Hz, 1H), 3.55 (s, 3H), 3.54 - 3.49 (m, 4H), 2.79 (d, J = 4.8 Hz, 3H).
Example 87. General Protocol for hASICla IC50 determination on QPatch Screening
Station (Sophion Bioscience).
[00317] 4 point IC50 curves were done, standard concentration range was 0.01 , 0.1, 1, 10 μΜ. If the compounds show > 50% inhibition at 10 nM, the concentration range was adapted.
Activation of hASICla was done with pH 6.75, washout/preincubation was at pH 7.4.
Four applications of pH 6.75 were done in the beginning of the experiment for current stabilization. After preincubation of the concentration at pH7.4, one activation per concentration at pH6.75 was done. Incubation between activation steps was minimum 200s.
Methods of analysis:
[00318] Compound concentrations were normalized to last initial pH 6.75 activation. Baseline current was subtracted of the peak current before normalization. IC50 value was calculated out of all valid experiments per compound, minimum was 3 valid data points per concentration.
Cell culture:
[00319] Cell line: CHO Kl hASICla #8.2
- Medium: DMEM/F12 (Sigma D8437) + 10% FCS
- Antibiotics 500 μ^ιι ΐ G418 (Life technologies, Cat.# 11811 -064)
- 5% C02, 37°C
[00320] Permanent culture
removed old medium and wash cells with Ca2+ and Mg2+ free PBS (Life technologies, Cat.# 14190-240)
added 4 ml of pre-warmed Accumax solution (Sigma, Cat.# A7089), incubated cells at 37°C for 3-4min
stopped activity by adding 16 ml of culture medium, triturated very gently (2-3x) counted cells using CasyCounter from Scharfe System
transferred cells into new T175-flask (Greiner bio-one, Cat.# 660 175) containing 30 mL of fresh media (3d: 5e5 / 4d: 2e5)
for QPatch experiments split cells Id (2e6) or 2d (8e5) before experiment without antibiotics in T75
[00321] Preparation for QPatch experiments
- Washed the cells twice with PBS (-)
To detach the cells used Accumax and incubated the suspension for 3 minutes at 37°C into the incubator
Added fresh Media to the cell suspension to stop the effect of Accumax
Centrifuged the cells 3 min with 1000 rpm
Resuspended the cells in 1,5ml SFM and transfered the suspension into a QTube in the QFuge, start experiment [00322] QPatch experiments:
Buffers:
Figure imgf000117_0001
Figure imgf000117_0002
[00323] The data is interpreted according to the following:
D > 5 μΜ;
C > 1-5 μΜ;
B ΙΟΟηΜ-ΙμΜ;
A <100nM.
Figure imgf000117_0003
6 B
7 C
8 C
9 A
10 A
11 C
12 C
13 B
14 B
15 A
16 B
17 B
18 A
19 C
20 C
21 D
22 A
23 A
24 B
25 D
26 D
27 B
28 C
29 A
30 C
31 A
32 B
33 D
34 C
35 C 36 A
37 D
38 A
39 D
40 A
41 D
42 C
43 A
44 B
45 A
46 B
47 A
48 A
49 C
50 C
51 B
52 C
53 A
54 D
55 C
56 A
57 C
58 C
59 D
60 D
61 D
62 D
63 D
64 A
65 A 0 66 A
2 67 B
3 68 B
74 69 D
75 70 D
76 71 D
77 72 A
78 73 B
79 74 A
81 75 B
82 76 B
83 77 C
84 78 D
85 79 D
85 80 D
86 81 A
86 82 D
86 83 A
86 84 B
86 85 A
Example 88. Pharmaceutical preparations
[00324] (A) Injection vials: A solution of 100 g of an active ingredient according to the invention and 5 g of disodium hydrogen phosphate in 3 1 of bidistilled water is adjusted to pH 6.5 using 2 N hydrochloric acid, sterile filtered, transferred into injection vials, is lyophilized under sterile conditions and is sealed under sterile conditions. Each injection vial contains 5 mg of active ingredient.
[00325] (B) Suppositories: A mixture of 20 g of an active ingredient according to the invention is melted with 100 g of soy lecithin and 1400 g of cocoa butter, is poured into moulds and is allowed to cool. Each suppository contains 20 mg of active ingredient. [00326] (C) Solution: A solution is prepared from 1 g of an active ingredient according to the invention, 9.38 g of NaH2P04 · 2 H20, 28.48 g of Na2HP04 · 12 H20 and 0.1 g of benzalkonium chloride in 940 ml of bidistilled water. The pH is adjusted to 6.8, and the solution is made up to 1 1 and sterilized by irradiation. This solution could be used in the form of eye drops.
[00327] (D) Ointment: 500 mg of an active ingredient according to the invention is mixed with 99.5 g of Vaseline under aseptic conditions.
[00328] (E) Tablets: A mixture of 1 kg of an active ingredient according to the invention, 4 kg of lactose, 1.2 kg of potato starch, 0.2 kg of talc and 0.1 kg of magnesium stearate is pressed to give tablets in a conventional manner in such a way that each tablet contains 10 mg of active ingredient.
[00329] (F) Coated tablets: Tablets are pressed analogously to Example E and subsequently are coated in a conventional manner with a coating of sucrose, potato starch, talc, tragacanth and dye.
[00330] (G) Capsules: 2 kg of an active ingredient according to the invention are introduced into hard gelatin capsules in a conventional manner in such a way that each capsule contains 20 mg of the active ingredient.
[00331] (H) Ampoules: A solution of 1 kg of an active ingredient according to the invention in 60 1 of bidistilled water is sterile filtered, transferred into ampoules, is lyophilized under sterile conditions and is sealed under sterile conditions. Each ampoule contains 10 mg of active ingredient.
[00332] (I) Inhalation spray: 14 g of an active ingredient according to the invention are dissolved in 10 1 of isotonic NaCl solution, and the solution is transferred into commercially available spray containers with a pump mechanism. The solution could be sprayed into the mouth or nose. One spray shot (about 0.1 ml) corresponds to a dose of about 0.14 mg.
[00333] While a number of embodiments of this invention are described herein, it is apparent that the basic examples may be altered to provide other embodiments that utilize the compounds and methods of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments that have been represented by way of example.

Claims

CLAIMS We claim:
1. A compound of formula I,
Figure imgf000122_0001
I
or a tautomer, or a pharmaceutically acceptable salt thereof, wherein:
X is C(R4)2, C(R4), N, N-R4, O, S, SO, or S02;
Y is C(R4), C-O-R4, or C=0; each ll is a single or double bond, as allowed by valence, wherein at least one of 'I is a double bond;
Ring A is a fused C5-6 aryl, a fused 3-8 membered saturated or partially unsaturated carbocyclic ring, a fused 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a fused 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
each R1 is independently -R, halogen, -OR, -SR, -CN, -N02, -S02R, -SOR, -C(0)R, -C02R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRS02R, or -N^;
Z is C(R4) or N;
each R2 is independently -R, halogen, -OR, -SR, -CN, -N02, -S02R, -SOR, -C(0)R, -C02R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRS02R, or -N(R)2; two R2 groups on the same atom are taken together with the atom to which they are attached to form a carbonyl, thiocarbonyl, or imine; each of which is optionally substituted;
R3 is -R, halogen, -OR, -SR, -CN, -N02, -S02R, -SOR, -C(0)R, -C02R, -C(0)N(R)2, - NRC(0)R, -NRC(0)N(R)2, -NRS02R, or -NCR^;
each R4 is independently hydrogen, Ci-6 aliphatic, C5_io aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted;
each R is independently hydrogen, 0-6 aliphatic, C5-10 aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted; or
two R groups on the same atom are taken together with the atom to which they are attached to form a C5-10 aryl, a 3-8 membered saturated or partially unsaturated carbocyclic ring, a 3-7 membered heterocylic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or a 5-6 membered monocyclic heteroaryl ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur; each of which is optionally substituted;
m is 0, 1, or 2; and
n is 0, 1, 2, 3, or 4.
2. The compound of claim 1, wherein X is CH2, CH, N, NH, N-CH3, or S.
3. The compound of any preceding claim, wherein Y is CH, C-OH, C-O-CH3, C-O-z'-Pr, or c=o.
4. The compound of any preceding claim, wherein Ring A is fused phenyl, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl;- 1,2,5- oxadiazolyl, 1,3,4-oxadiazolyl, oxazolyl, pyrimidinyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridinyl, pyrimidinyl, pyrrolyl, thiazolyl, thienyl, thiophenyl, triazinyl, 1,2,3-triazolyl,
1,2,4-triazolyl, 1,2,5-triazolyl, or 1,3,4-triazolyl.
5. The compound of any preceding claim, wherein , Ring A is
6. The compound of any preceding claim, wherein each R1 is independently -R, halogen, - OR, -SR, -CN, -N02, -S02R, -SOR, -C(0)R, -CO2R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2,
Figure imgf000124_0002
7. The compound of any preceding claim, wherein Z is N.
8. The compound of any preceding claim, wherein Z is CH.
9. The compound of any preceding claim, wherein R3 is -R, halogen, -OR, -SR, -CN, - NO2, -S02R, -SOR, -C(0)R, -CO2R, -C(0)N(R)2, -NRC(0)R, -NRC(0)N(R)2, -NRSO2R, or - N(R)2.
10. The compound of any preceding claim, wherein R3 is -C(0)R, -CO2R, -C(0)N(R)2, - NRC(0)R, -NRC(0)N(R)2, -NRSO2R, or -N(R)2.
Figure imgf000124_0003
Figure imgf000125_0001
12. The compound of claim 1, of formula I-a,
Figure imgf000126_0001
I-a;
or a pharmaceutically acceptable salt thereof.
13. The compound of claim 1, of formula I-c,
Figure imgf000126_0002
I-c;
or a pharmaceutically acceptable salt thereof.
14. The compound of claim 1, selected from Table 1.
15. A pharmaceutical composition comprising a compound of any one of claims 1-14, and a pharmaceutically acceptable adjuvant, carrier, or vehicle.
16. A method for inhibiting ASIC, or a mutant thereof, activity in a patient or in a biological sample, comprising the step of administering to said patient or contacting said biological sample with a compound of any one of claims 1-14, or a physiologically acceptable salt thereof.
17. A method for treating a ASIC-mediated disorder in a patient in need thereof, comprising the step of administering to said patient a compound of any one of claims 1-14.
18. The method of claim 17, wherein the disorder is selected from multiple sclerosis (MS), polyneuritis, multiple neuritis, amyotrophic lateral sclerosis (ALS), Alzheimer's disease, optic neuritis, and Parkinson's disease.
19. A method for treating ischemic stroke, epilepsy, multiple sclerosis, Huntington's disease, Parkinson's disease, optic neuritis, or spinal cord injury in a subject, comprising the step of administering to said subject a compound of any one of claims 1-14, or a physiologically acceptable salt thereof.
PCT/EP2016/001530 2015-09-15 2016-09-12 Compounds as asic inhibitors and uses thereof WO2017045750A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
DE102015012049.2 2015-09-15
DE102015012049.2A DE102015012049A1 (en) 2015-09-15 2015-09-15 Compounds as ASIC inhibitors and their uses

Publications (1)

Publication Number Publication Date
WO2017045750A1 true WO2017045750A1 (en) 2017-03-23

Family

ID=56979504

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2016/001530 WO2017045750A1 (en) 2015-09-15 2016-09-12 Compounds as asic inhibitors and uses thereof

Country Status (2)

Country Link
DE (1) DE102015012049A1 (en)
WO (1) WO2017045750A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1057612A (en) * 1964-01-15 1967-02-01 Thomae Gmbh Dr K Thieno-pyrimidines
DE2003714A1 (en) * 1970-01-28 1971-08-12 Thomae Gmbh Dr K Thrombocyte aggregation-inhibiting thienopyrimidine deriv.
EP0302967A2 (en) * 1987-08-10 1989-02-15 Kanebo, Ltd. Quinazoline derivative, processes for its production, and cerebral dysfunction remedying agent comprising it as active ingredient
WO2003055865A1 (en) * 2001-12-24 2003-07-10 Fujisawa Pharmaceutical Co., Ltd. Quinazolinone derivative
EP1398032A1 (en) * 2002-09-10 2004-03-17 PheneX Pharmaceuticals AG 4-Oxo-quinazolines as LXR nuclear receptor binding compounds
WO2005099688A2 (en) * 2004-04-07 2005-10-27 Takeda Pharmaceutical Company Limited Cyclic compounds
WO2006072588A1 (en) * 2005-01-07 2006-07-13 Laboratorios Del Dr. Esteve S.A. Substituted 2-amino-quinazolin-4-cn compounds for use in the treatment of cns disorders, pain, stroke, addiction and epilepsy, their preaparation and use as intermediates
WO2006100520A1 (en) * 2005-03-24 2006-09-28 Merck Sharp & Dohme Limited 2,3-substituted fused pyrimidin-4(3h)-ones as vr1 antagonists
CN101817833A (en) * 2009-02-26 2010-09-01 中国科学院广州生物医药与健康研究院 DPP-IV inhibitor
WO2011055115A1 (en) * 2009-11-04 2011-05-12 Almac Discovery Limited Akt / pkb inhibitors
WO2014111666A1 (en) * 2013-01-18 2014-07-24 Universite D'aix-Marseille Thieno[3,2-d]pyrimidin-4(3h)-one derivatives and the preparation and pharmaceutical uses thereof
EP2786998A1 (en) * 2011-12-01 2014-10-08 Shanghai Institute of Materia Medica, Chinese Academy of Sciences Thienyl [3, 2-d]pyrimidin-4-one compounds, preparation method, pharmaceutical compositions and use thereof
WO2015006508A2 (en) * 2013-07-09 2015-01-15 The Translational Genomics Research Institute Compositions and methods of screening for compounds that modulate activity at a tweak binding site on a crd of fn14

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1057612A (en) * 1964-01-15 1967-02-01 Thomae Gmbh Dr K Thieno-pyrimidines
DE2003714A1 (en) * 1970-01-28 1971-08-12 Thomae Gmbh Dr K Thrombocyte aggregation-inhibiting thienopyrimidine deriv.
EP0302967A2 (en) * 1987-08-10 1989-02-15 Kanebo, Ltd. Quinazoline derivative, processes for its production, and cerebral dysfunction remedying agent comprising it as active ingredient
WO2003055865A1 (en) * 2001-12-24 2003-07-10 Fujisawa Pharmaceutical Co., Ltd. Quinazolinone derivative
EP1398032A1 (en) * 2002-09-10 2004-03-17 PheneX Pharmaceuticals AG 4-Oxo-quinazolines as LXR nuclear receptor binding compounds
WO2005099688A2 (en) * 2004-04-07 2005-10-27 Takeda Pharmaceutical Company Limited Cyclic compounds
WO2006072588A1 (en) * 2005-01-07 2006-07-13 Laboratorios Del Dr. Esteve S.A. Substituted 2-amino-quinazolin-4-cn compounds for use in the treatment of cns disorders, pain, stroke, addiction and epilepsy, their preaparation and use as intermediates
WO2006100520A1 (en) * 2005-03-24 2006-09-28 Merck Sharp & Dohme Limited 2,3-substituted fused pyrimidin-4(3h)-ones as vr1 antagonists
CN101817833A (en) * 2009-02-26 2010-09-01 中国科学院广州生物医药与健康研究院 DPP-IV inhibitor
WO2011055115A1 (en) * 2009-11-04 2011-05-12 Almac Discovery Limited Akt / pkb inhibitors
EP2786998A1 (en) * 2011-12-01 2014-10-08 Shanghai Institute of Materia Medica, Chinese Academy of Sciences Thienyl [3, 2-d]pyrimidin-4-one compounds, preparation method, pharmaceutical compositions and use thereof
WO2014111666A1 (en) * 2013-01-18 2014-07-24 Universite D'aix-Marseille Thieno[3,2-d]pyrimidin-4(3h)-one derivatives and the preparation and pharmaceutical uses thereof
WO2015006508A2 (en) * 2013-07-09 2015-01-15 The Translational Genomics Research Institute Compositions and methods of screening for compounds that modulate activity at a tweak binding site on a crd of fn14

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 12 July 2006 (2006-07-12), XP002762883, Database accession no. 892279-59-3 *
DATABASE REGISTRY [online] CHEMICAL ABSTRACTS SERVICE, COLUMBUS, OHIO, US; 29 October 2010 (2010-10-29), XP002762882, retrieved from STN Database accession no. 1248560-73-7 *
HAMEED AFAF M ABDEL: "Synthesis of new 1,2,4-oxadiazolidin-5-ylthiophenes and thienopyrimidine derivatives by aza-Wittig reaction using a thienyl carbodiimide", CHINESE CHEMICAL LETTERS, vol. 23, no. 4, 2012, pages 411 - 414, XP028909502, ISSN: 1001-8417, DOI: 10.1016/J.CCLET.2011.12.017 *
MODICA M ET AL: "Design, synthesis and binding properties of novel and selective 5-HT"3 and 5-HT"4 receptor ligands", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, EDITIONS SCIENTIFIQUE ELSEVIER, PARIS, FR, vol. 35, no. 12, 1 December 2000 (2000-12-01), pages 1065 - 1079, XP004372825, ISSN: 0223-5234, DOI: 10.1016/S0223-5234(00)01187-9 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10287353B2 (en) 2016-05-11 2019-05-14 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385130B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-1 inhibitors
US10385131B2 (en) 2016-05-11 2019-08-20 Huya Bioscience International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors
US11535670B2 (en) 2016-05-11 2022-12-27 Huyabio International, Llc Combination therapies of HDAC inhibitors and PD-L1 inhibitors

Also Published As

Publication number Publication date
DE102015012049A1 (en) 2017-03-16

Similar Documents

Publication Publication Date Title
US10392375B2 (en) Heteroaryl compounds as IRAK inhibitors and uses thereof
US10377745B2 (en) Heterocyclic compounds as NaV channel inhibitors and uses thereof
AU2016323613B2 (en) Heteroaryl compounds as IRAK inhibitors and uses thereof
AU2016215184B2 (en) Pyridazinone macrocycles as IRAK inhibitors and uses thereof
EP3253764B1 (en) Macrocyclic compounds as irak1/4 inhibitors and uses thereof
CA2964982A1 (en) Heteroaryl compounds as irak inhibitors and uses thereof
AU2019249721A1 (en) Heteroaryl compounds as type II IRAK inhibitors and uses hereof
WO2017045750A1 (en) Compounds as asic inhibitors and uses thereof
WO2017045751A1 (en) Compounds as asic inhibitors and uses thereof
WO2019079375A1 (en) PYRIMIDINE ΤΒΚ/ΙΚΚε INHIBITOR COMPOUNDS AND USES THEREOF

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: 16769861

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 16769861

Country of ref document: EP

Kind code of ref document: A1