WO2006046040A1 - Pharmaceutical compounds - Google Patents

Pharmaceutical compounds Download PDF

Info

Publication number
WO2006046040A1
WO2006046040A1 PCT/GB2005/004146 GB2005004146W WO2006046040A1 WO 2006046040 A1 WO2006046040 A1 WO 2006046040A1 GB 2005004146 W GB2005004146 W GB 2005004146W WO 2006046040 A1 WO2006046040 A1 WO 2006046040A1
Authority
WO
WIPO (PCT)
Prior art keywords
thieno
pyrimidin
ylmethyl
moφholin
group
Prior art date
Application number
PCT/GB2005/004146
Other languages
French (fr)
Inventor
Stephen J. Shuttleworth
Adrian J. Folkes
Irina S. Chuckowree
Nan Chi Wan
Timothy C. Hancox
Stewart J. Baker
Sukhjit Sohal
Mohammed A. Latif
Original Assignee
Piramed Limited
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 Piramed Limited filed Critical Piramed Limited
Priority to EP05798278.7A priority Critical patent/EP1812446B1/en
Priority to ES05798278.7T priority patent/ES2554689T3/en
Priority to JP2007537401A priority patent/JP5077946B2/en
Publication of WO2006046040A1 publication Critical patent/WO2006046040A1/en
Priority to US11/893,625 priority patent/US7872003B2/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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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 pyrimidine derivatives and their use as inhibitors of phosphatidylinositol 3-kinase (P 13 K).
  • Phosphatidylinositol (hereinafter abbreviated as "PI") is one of a number of phospholipids found in cell membranes. In recent years it has become clear that PI plays an important role in intracellular signal transduction. In the late 1980s, a PI3 kinase (PI3K) was found to be an enzyme which phosphorylates the 3-position of the inositol ring of phosphatidylinositol (D. Whitman et al, 1988, Nature, 332, 664).
  • PI3K was originally considered to be a single enzyme, but it has now been clarified that a plurality of subtypes are present in PI3K. Each subtype has its own mechanism for regulating activity.
  • Three major classes of PI3Ks have been identified on the basis of their in vitro substrate specificity (B. Vanhaesebroeck,1997, Trend in Biol. Sci, 22, 267).
  • Substrates for class I PI3Ks are PI, PI 4-phosphate (PI4P) and PI 4,5-biphosphate (PI (4,5)P2).
  • Class I PI3Ks are further divided into two groups, class Ia and class Ib, in terms of their activation mechanism.
  • Class Ia PBKs include PI3K pi 10a, pi lO ⁇ and pi lO ⁇ subtypes, which transmit signals from tyrosine kinase-coupled receptors.
  • Class Ib PI3K includes a pi lO ⁇ subtype activated by a G protein-coupled receptor.
  • PI and PI(4)P are known as substrates for class II PDKs.
  • Class II PDKs include PDK C2 ⁇ , C2 ⁇ and C2 ⁇ subtypes, which are characterized by containing C2 domains at the C terminus.
  • the substrate for class III PDKs is PI only.
  • the class Ia subtype has been most extensively investigated to date.
  • the three subtypes of class Ia are heterodimers of a catalytic 110 kDa subunit and regulatory subunits of 85 kDa or 55 kDa.
  • the regulatory subunits contain SH2 domains and bind to tyrosine residues phosphorylated by growth factor receptors with a tyrosine kinase activity or oncogene products, thereby inducing the PBK activity of the pi 10 catalytic subunit which phosphorylates its lipid substrate.
  • the class Ia subtypes are considered to be associated with cell proliferation and carcinogenesis.
  • WO 01/083456 describes a series of condensed heteroaryl derivatives which have activity as inhibitors of PB K and which suppress cancer cell growth.
  • the present invention provides a compound which is a fused pyrimidine of formula (I):
  • A represents a thiophene or furan ring; n is 1 or 2;
  • R 1 is a group of formula:
  • n 0 or 1
  • R 30 is H or Ci-C 6 alkyl
  • R 4 and R 5 form, together with the N atom to which they are attached, a 5- or 6- membered saturated N-containing heterocyclic group which includes 0 or 1 additional heteroatoms selected from N, S and O, which may be fused to a benzene ring and which is unsubstituted or substituted; or one of R 4 and R 5 is alkyl and the other is a 5- or 6-membered saturated N-containing heterocyclic group as defined above or an alkyl group which is substituted by a 5- or 6-membered saturated N- containing heterocyclic group as defined above; R 2 is selected from: (a)
  • R 6 and R 7 form, together with the nitrogen atom to which they are attached, a morpholine, thiomorpholine, piperidine, piperazine, oxazepane or thiazepane group which is unsubstituted or substituted;
  • Y is a C 2 - C 4 alkylene chain which contains, between constituent carbon atoms of the chain and/or at one or both ends of the chain, 1 or 2 heteroatoms selected from O, N and S, and which is unsubstituted or substituted; and R 3 is selected from: (a) a group of the following formula:
  • B is a phenyl ring which is unsubstituted or substituted and Z is selected from H, -OR, -SR, CH 2 OR, -CO 2 R, CF 2 OH, CH(CF 3 )OH, C(CF 3 ) 2 OH, -(CH 2 ) q OR, -(CH 2 ) q NR 2 , -C(O)N(R) 2 , -NR 2 , -NRC(O)R, -S(O) 1n N(R) 2 , -OC(O)R, OC(O)N(R) 2 , -NRS(O) m R , -RC(O)N(R) 2 , CN, halogen and -NO 2 , wherein each R is independently selected from H, Ci-C 6 alkyl, C 3 - Ci 0 cycloalkyl and a 5- to 12- membered aryl or heteroaryl group, the group being unsubstitute
  • the thiophene or furan ring A in formula (I) adopts either of the two available regiochemical orientations.
  • Formula (I) thus covers the thieno[3,2- djpyrimidines and furano[3,2-d]pyrimidines of the following formula (Ia) as well as the thieno[2,3-d]pyrimidines and furano[2,3-d]pyrimidines of the following formula (Ib):
  • each of R 1 to R 3 and n is as defined above and X is S or O.
  • the group or groups R 1 which are the same or different in a given compound when n is 2, may be bonded to either or both of the two available ring positions on the thiophene or furan ring A. Referring to structures (Ia) and (Ib) above, therefore, when n is 1 the furan or thiophene ring is mono-substituted by R 1 at the 2-position or the 3-position. When n is 2, the thiophene or furan ring is di- substituted by R 1 at positions 2 and 3.
  • an alkyl group is a straight or branched chain saturated hydrocarbon radical which is unsubstituted or substituted.
  • Ci-C 20 alkyl for instance Ci-Cio alkyl, such as Ci-C 6 alkyl or Ci-C 4 alkyl, for example methyl, ethyl, i-propyl, n-propyl, t-butyl, s-butyl or n-butyl. It may also be pentyl, hexyl, heptyl, octyl and the various branched chain isomers thereof.
  • R 20 selected from halogen, alkoxy, carbocyclyl, a 5- or 6-membered saturated N- containing heterocyclic group as defined above, OH, SR, CN, nitro, NR 2 , -COOR, - C(O)R, S(O) m R and -CONR 2 , wherein each R is H, unsubstituted alkyl or C 3 -Ci 0 cycloalkyl and m is 1 or 2.
  • haloalkyl group or a group -alk- N(R 4 )(R 5 ) wherein alk is an alkylene chain and R 4 and R 5 form, together with the N atom to which they are attached, a 5- or 6-membered saturated N-containing heterocyclic group which includes 0 or 1 additional heteroatoms selected from N, S and O, which may be fused to a benzene ring and which is unsubstituted or substituted.
  • R 20 is selected from halogen, alkoxy, carbocyclyl, a 5- or 6- membered saturated N-containing heterocyclic group as defined above, OH, CN, NR 2 , -COOR and -CONR 2 , wherein each R is H or unsubstituted alkyl as defined above. It is, for instance, a haloalkyl group or a group -alk-N(R 6 )(R 5 ) wherein alk is an alkylene chain and R 4 and R 5 form, together with the N atom to which they are attached, a 5- or 6-membered saturated N-containing heterocyclic group as defined above.
  • An alkylene group is unsubstituted or substituted, straight or branched chain saturated divalent hydrocarbon group.
  • Ci-C 8 alkylene for instance Ci-C 6 alkylene.
  • Ci-C 4 alkylene for example C 2 -C 4 alkylene, such as methylene, ethylene, i-propylene, n-propylene, t-butylene, s-butylene or n-butylene. It may also be pentylene, hexylene, heptylene, octylene and the various branched chain isomers thereof.
  • the alkylene group is substituted it is typically substituted by a group R 20 as defined above.
  • alkenyl group is an unsubstituted or substituted, straight or branched chain hydrocarbon radical having one or more double bonds. Typically it is C 2 -C 8 alkenyl, for instance C 2 -C 6 alkenyl, such as allyl, butenyl, butadienyl, pentenyl or hexenyl.
  • alkenyl group is substituted it is typically substituted by a group R 20 as defined above or by alkyl which is unsubstituted or substituted by a group R 20 as defined above.
  • alkynyl group is an unsubstituted or substituted, straight or branched chain hydrocarbon radical having one or more triple bonds. Typically it is C 2 -C 8 alkynyl, for instance C 2 -C 6 alkynyl, such as ethynyl, propynyl or butynyl.
  • alkynyl group is substituted it is typically substituted by a group R 20 as defined above or by alkyl which is unsubstituted or substituted by a group R 20 as defined above.
  • a haloalkyl group is an alkyl group as defined above, substituted by one or more halogen atoms. It can be a perhaloalkyl group, for instance trifiuoromethyl or perfluorohexyl.
  • a halogen is chlorine, fluorine, bromine or iodine. It is typically bromine or iodine.
  • An alkoxy group is typically Ci-C 6 alkoxy, for instance Ci-C 4 alkoxy, such as methoxy, ethoxy, i-propoxy, n-propoxy, t-butoxy, n-butoxy or s-butoxy. It is unsubstituted or substituted, for instance by a group R 20 as defined above or by alkyl which is unsubstituted or substituted by a group R 20 as defined above or by alkyl which is unsubstituted or substituted by a group R 20 as defined above.. Typically it is substituted by carbocyclyl, morpholino, OH, CN, NR 2 , -COOR or -CONR 2 , wherein each R is H or unsubstituted alkyl as defined above.
  • a carbocyclyl group is a non-aromatic saturated or unsaturated monocyclic hydrocarbon ring, typically having from 3 to 10 carbon atoms. It may be a C 3 -C 8 cycloalkyl group, or C 5 -Ci 0 cycloalkyl group, for instance cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl.
  • cycloalkenyl group typically C 4 -C 8 cycloalkenyl, for instance cylcopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, cyclohepadienyl, cyclooctenyl or cyclooctadienyl.
  • a carbocyclyl group may be unsubstituted or substituted, for instance by a group R 20 as defined above or by alkyl which is unsubstituted or substituted by a group R 20 as defined above.. Typically it is substituted by alkoxy, morpholino, OH, CN, NR 2 , -COOR and -CONR 2 , wherein each R is H or unsubstituted alkyl as defined above.
  • a 5- or 6-membered saturated N-containing heterocyclic group which includes 0 or 1 additional heteroatoms selected from N, S and O, which may be fused to a benzene ring and which is unsubstituted or substituted is typically selected from morpholine, piperidine, piperazine, pyrrolidine, thiomorpholine, quinoline, isoquinoline, diazepane, oxazepane and thiazepane.
  • substituents are selected from alkyl which is unsubstituted or substituted, alkoxy which is unsubstituted or substituted, -NR 2 , -N(R'")-alk-OR, -alk-OR, -O-alk-OR, -alk-C(O)NR 2 , -C(O)NR 2 , -alk-Het, -N(R)-Het, -O-Het, -N(R)-C(O)-alk-OR, -C(O)-N(R)-alk-OR, -alk-S(O) 2 R, -N(R)-alk-OR, -alk-NR'R", -N(R'")-S(O) 2 R, -N(R)-alk-OR, -alk-NR'R", -N(R'")-S(O) 2 R, -N(R)-alk-OR, -alk-
  • R is H or alkyl
  • -NR'R wherein each of R' and R" is independently H, alkyl or alkoxy
  • -SO 2 R'" wherein R'" is alkyl which is unsubstituted or substituted, for instance by NR 2 or a 5- or 6-membered saturated N- containing heterocyclic group as defined above.
  • a 5-, 6- or 7-membered saturated heterocyclic group which contains 1 or 2 heteroatoms selected from N, S and O and which is unsubstituted or substituted is typically selected from tetrahydropyran, tetrahydrothiopyran, tetrahydrofuran and tetrahydrothiofuran.
  • a 5-, 6- or 7-membered saturated heterocyclic group which contains 1 or 2 heteroatoms selected from N, S and O is substituted it may be substituted as specified above for a 5- or 6-membered saturated N-containing heterocyclic group.
  • a heteroaryl group is a heteroaryl group which contains 1 , 2 3 or 4 ring nitrogen atoms and O, 1 or 2 additional heteroatoms selected from O, N and S, which group is monocyclic or bicyclic and which is unsubstituted or substituted. It is typically a 5- to 12-membered ring.
  • heteroaryl group examples include pyrrole, pyrazole, triazole, tetrazole, indazole, thiazole, isothiazole, oxazole, isooxazole, indole, isoindole, l,3-dihydro-indol-2-one, pyridine-2-one, pyridine, pyridin-3-ol, imidazole, 1,3-dihydro-benzimidazolone, benzimidazole, benzothiazole, benzothiadiazole, quinoline, isoquinoline, quinoxaline, pyrazolopyridine, aminopyrazolinone, imidazopyridine, pyrimidine, pyridazine, pyrazine and isatin groups.
  • Preferred examples include indazole, indole, pyrazole and tetrazole groups. These groups may be unsubstituted or substituted, for instance by a group R 20 as specified above or by alkyl which is unsubstituted or substituted by a group R 20 as defined above.
  • a 5- or 6-membered N containing heteroaryl group which may be fused to a benzene ring is typically selected from pyrrole, pyrazole, triazole, tetrazole, indazole, thiazole, isothiazole, oxazole, isooxazole, indole, isoindole, 1,3- dihydro-indol-2-one, pyridine-2-one, pyridine, pyridin-3-ol, imidazole, 1,3-dihydro- benzimidazolone, benzimidazole, benzothiazole, benzothiadiazole, quinoline, isoquinoline, quinoxaline, pyrazolopyridine, aminopyrazolinone, imidazopyridine, pyrimidine, pyridazine and pyrazine.
  • heteroaryl group When such a heteroaryl group is substituted it may be substituted by a group R 20 as defined above or by alkyl which is unsubstituted or substituted by a group R 20 as defined above.
  • R 1 m is 0 or 1 , typically 1.
  • R 30 is typically H.
  • R 4 and R 5 typically form, together with the N atom to which they are attached, a saturated N-containing heterocyclic group selected from morpholine, thiomorpholine, piperidine, piperazine, pyrrolidine, quinoline, isoquinoline, diazepane, oxazepane and thiazepane.
  • the heterocylic group formed by R 4 and R 5 is unsubstituted or substituted, for instance by the examples of substituent groups listed above, such as a group R 20 as defined above or by alkyl which is unsubstituted or substituted by a group R 20 as defined above.
  • the ring formed by R 6 and R 7 is typically morpholine which is unsubstituted or substituted, for instance by a group R 20 as specified above or by alkyl which is unsubstituted or substituted by a group R 20 as defined above.
  • It may alternatively be a group selected from tetrahydropyran, tetrahydrothiopyran, tetrahydrofuran and tetrahydrothiofuran, each of which is unsubstituted or substituted, for instance, for instance by a group R 20 as specified above or by alkyl which is unsubstituted or substituted by a group R 20 as defined above.
  • the ring formed by R 6 and R 7 When the ring formed by R 6 and R 7 is substituted it may be substituted on either a ring heteroatom or a ring carbon atom, for instance by a group R 20 as defined above.
  • the alkylene chain represented by Y forms, together with the carbon atoms to which it is attached, a saturated 5-, 6- or 7- membered heterocyclic ring which contains 1 or 2 heteroatoms selected from O, N and S and which is unsubstituted or substituted.
  • the heterocyclic ring include tetrahydropyran, tetrahydrofuran, tetrahydrothiopyran, tetrahydrothiofuran and morpholine.
  • the phenyl ring B is unsubstituted (apart from group Z) or substituted.
  • the group Z is the sole substituent.
  • it When it is substituted it typically comprises, in addition to group Z, one or more substituents selected from halo, alkyl, alkenyl, alkynyl, CN, NO 2 , OR', SR', NR' 2> C(O)R', SOR', SO 2 R', SO 2 NR' 2 , NC(O)R' and CO 2 R', wherein each R' is independently H or Ci-C 6 alkyl.
  • Group Z is bonded to any available ring position on the phenyl ring B. Thus it may be situated at the 2-, 3-, A-, 5- or 6- position of the phenyl ring. Typically it is bonded at position 3 or 4. Z is most typically other than H, such that moiety -BZ is a substituted phenyl ring.
  • a typical example of Z is a group OR as defined above, in particular OH. In this embodiment the OR group, or OH group, is typically bonded at ring position 3 or 4 of phenyl ring B.
  • -BZ is a 3-hydroxyphenyl or 4- hydroxyphenyl group, or an isostere thereof, other than an indole or indazole group which is unsubstituted or substituted.
  • An isostere as used herein is a functional group which possesses binding properties which are the same as, or similar to, the 3-hydroxyphenyl or 4- hydroxyphenyl group in the context of the structure of formula (I). Isosteres of
  • 3-hydroxyphenyl and 4-hydroxyphenyl groups are encompassed within definitions (b) and (c) above for R 3 .
  • R 3 the benzene ring is unsubstituted or substituted. If it is substituted it may be substituted by one or more substituents selected from a group Z , R 20 as defined above, alkyl which is unsubstituted or substituted by R 20 as defined above, and any of the groups specified above as an additional substituent on the phenyl ring B.
  • both the benzene ring and the heteroaryl group are unsubstituted.
  • Examples of the groups included in definitions (b) and (c) for R 3 include pyrrole, pyrazole, triazole, tetrazole, thiazole, isothiazole, oxazole, isooxazole, isoindole, l,3-dihydro-indol-2-one, pyridine-2-one, pyridine, pyridin-3-ol, imidazole, 1,3-dihydro-benzimidazolone, benzimidazole, benzothiazole, benzothiadiazole, quinoline, isoquinoline, quinoxaline, pyrazolopyridine, aminopyrazolinone, imidazopyridine, pyrimidine, pyridazine, pyrazine and isatin groups.
  • Preferred examples include pyrazole and tetrazole groups. These groups may be unsubstituted or substituted, for instance by a group Z , R 20 or alkyl which is unsubstituted or substituted by a group R 20 as defined above. Typically these groups are isosteres. More specifically, groups included in definitions (b) and (c) for R 3 as defined above include the following structures, which are typically isosteres as defined above:
  • each R 10 is independently selected from H, Ci-C 6 alkyl, Ci-C 6 alkoxy, Ci-C 6 acyl, -C(O)NR 5 R", -S(O) t NR'R", aryl, heteroaryl, sulphonyl and halogen, whrein R' and R" are each independently H or CpC 6 alkyl and t is 1 or 2; each R 1 ' is independently selected from -OR 10 and -N(R 10 ) 2 , wherein R 10 is as defined above; each R 12 is independently H, F or CF 3 ; each W is independently selected from CR 10 and N, wherein R 10 is as defined above; and W is selected from O, S and NR 12 wherein R 12 is as defined above. with the proviso that formulae (Ii) and (lii) are other than indole and indazole.
  • Specific examples of compounds of the invention include:
  • the compounds of formula (I) may exist in the form of geometrical isomers or tautomers depending on the kinds of substituent groups, and these isomers in separated forms or mixtures thereof may be used in the present invention. Where the compounds have asymmetric carbon atoms, optical isomer forms may exist based on such carbon atoms. All of the mixtures and the isolated forms of these optical isomers may be used in the present invention.
  • a suitable synthetic strategy for producing compounds of formula (I) in which m is 1 employs the precursor carboxaldehyde of formula (II):
  • a and R 2 are as defined above.
  • the synthesis comprises performing, in either order, a palladium-mediated (Suzuki-type) cross- coupling reaction and a reductive amination.
  • the present invention therefore further provides a process for producing a compound of formula (I) as defined above in which m is 1, which process comprises: (a) treating a compound of formula (II):
  • a and R 2 are as defined above, with a boronic acid or ester thereof of formula R 3 B(OR 15 ) 2 , in which R 3 is as defined above and each R 15 is H or Ci-C 6 alkyl or the two groups OR 15 form, together with the boron atom to which they are attached, a pinacolato boronate ester group, in the presence of a Pd catalyst; and treating the resulting compound of formula (III): wherein A, R 2 and R 3 are as defined above, with an amine of formula NHR 4 R 5 in which R 4 and R 5 are as defined above, in the presence of a suitable reducing agent; or
  • A, R 2 , R 4 and R 5 are as defined above, with a boronic acid or ester thereof of formula R 3 B(OR 15 ) 2 , in which R 3 is as defined above and each R 15 is H or Ci-C 6 alkyl or the two groups OR 15 form, together with the boron atom to which they are attached, a pinacolato boronate ester group, in the presence of a Pd catalyst.
  • the palladium catalyst may be any that is typically used for Suzuki-type cross-couplings, such as PdCl 2 (PPh 3 ) 2.
  • the reducing agent is typically a borohydride, for instance NaBH(OAc) 3 , NaBH 4 or NaCNBH 4 , in particular NaBH(OAc) 3.
  • the invention further provides a process for producing a compound of formula (I) in which m is 1 and R 3 is a 3- or 4-hydroxyphenyl group, which process comprises: (a) treating a compound of formula (V): wherein OR' is bonded at position 3 or 4 of the phenyl ring to which it is attached, R' is a hydroxy protecting group and A and R 2 are as defined above, with an amine of formula NHR 4 R 5 wherein R 4 and R 5 are as defined above, in the presence of a suitable reducing agent; and
  • the reducing agent is typically a borohydride, for instance as specified above.
  • hydroxy protecting groups are known in the art, for instance as described in "Protective Groups for Organic Chemistry", Third Edition, T.W. Greene and P.G.M. Wuts, John Wiley & Sons, 1999.
  • a hydroxy group can be protected as an acetal, a substituted acetal, an ester, a xanthate, an ether or a silyl ether.
  • the acetal is preferably tetrahydropyran.
  • the silyl ether is preferably trimethylsilyl ether, t-butyl dimethylsilyl ether, triiso-propylsilyl ether or t- butyldiphenyl-silyl ether.
  • a compound of formula (V) as defined above may be produced by a process which comprises treating a compound of formula (VI):
  • A, R 2 and R' are as defined above, with a lithiating agent followed by N,N'- dimethylformamide (DMF).
  • the reaction is typically conducted by adding a solution of the lithiating agent in a non-polar organic solvent, for instance a hydrocarbon solvent such as hexane, to a suspension of the compound of formula (VI) in an organic solvent such as tetrahydrofuran (THF). If THF is used the addition takes place at a low temperature, of about -78°C.
  • the lithiating agent is typically an alkyllithium, for instance n-butyllithium.
  • a compound of formula (VI) as defined above may be produced by a process which comprises treating a compound of formula (VII):
  • R' and R 15 are as defined above, in the presence of a palladium catalyst.
  • the reaction is conducted under conventional conditions for a Suzuki-type cross-coupling reaction, for instance as described above.
  • a compound of formula (II) as defined above wherein R 2 is -NR 6 R 7 may be prepared by a process which comprises treating a compound of formula (IX):
  • a compound of formula (IX) as defined above may be produced by a process which comprises treating a compound of formula (X):
  • a compound of formula (X) may be prepared by the process described in
  • CH CfH 2 may be prepared by a process which comprises submitting a compound of formula (XI):
  • the compound of formula (XIII) may be converted to the corresponding carboxaldehyde by treatment with a lithiating agent followed by N,N'- dimethylformamide (DMF), for instance under the conditions described above for the conversion of a compound of formula (VI) to a compound of formula (V).
  • the lithiating agent is typically as defined above.
  • the resulting carboxaldehyde may then be converted into a desired final compound of formula (I) as defined above, in which m is 1 , by treatment with an amine of formula NHR 4 R 5 in which R 4 and R 5 are as defined above, in the presence of a suitable reducing agent, for instance a borohydride as specified above, in particular NaBH(OAc) 3 .
  • a compound of formula (I) as defined above in which m is 0 may be prepared by a Buchwald-type palladium-mediated nitrogen insertion reaction. Such a process may comprise treating a compound of formula (XIV):
  • A, R 2 and R 3 are as defined above and W is a halo group selected from Br and I, with an amine of formula NHR 4 R 5 in which R 4 and R 5 are as defined above, in the presence of a palladium catalyst.
  • a compound of formula (XIV) may be produced by treating a compound of formula (XV):
  • A, R 2 and R 3 are as defined above, with a lithiating agent and a halogen selected from bromine and iodine.
  • the lithiating agent is typically an alkyllithium, for instance butyllithium.
  • the halogen is typically iodine, which gives rise to a compound of formula (XIV) in which W is I.
  • a compound of formula (I) as defined above in which m is 0 may also be prepared by an SNAr displacement reaction, for instance under the conditions described by D. Prim and G. Kirsch in Tetrahedron 55 (21), 6511-6526, 1999.
  • Such a process comprises treating a compound of formula (XIV) as defined above in which W is Br with an amine of formula NHR 4 R 5 in which R 4 and R 5 are as defined above in H 2 O under reflux for 12 hours.
  • a compound of formula (I) as defined above in which m is 0 may alternatively be prepared by treating a compound of formula (XIV) as defined above in which W is I with an amine of formula NHR 4 R 5 in which R 4 and R 5 are as defined above in 1,4-dioxane in the presence of Cul/En and K 3 PO 4 .
  • the reaction is conducted at about 11O 0 C for 24 hours. This procedure is described by Kang S-K et al in Synlett, (3), 427-430, 2002.
  • a fused pyrimidine of formula (I) may be converted into a pharmaceutically acceptable salt, and a salts may be converted into the free compound, by conventional methods.
  • pharmaceutically acceptable salts include acid addition salts with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulphuric acid, nitric acid and phosphoric acid; and organic acids such as formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, methanesulfonic acid, ethanesulfonic acid, aspartic acid and glutamic acid.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulphuric acid, nitric acid and phosphoric acid
  • organic acids such as formic acid, acetic acid, trifluoroace
  • the salts include the salts of alkali and alkaline earth metals and ammonium, for instance the salts of sodium, potassium, magnesium, calcium and ammonium.
  • the latter are prepared by treating the free fused pyrimidine of formula (I), or an acid addition salt thereof, with the corresponding metal base or ammonia.
  • the compounds of formula (I) and their salts may exist as hydrates or solvates.
  • Compound of the present invention have been found in biological tests to be inhibitors of PB kinase.
  • the compounds are selective for class Ia PB kinases over class Ib and typically exhibit at least a 20-fold selectivity for class Ia over class Ib PB kinases.
  • the compounds are selective for the pi 10a isoform.
  • a compound of the present invention may thus be used as an inhibitor of PI3 kinase, in particular of a class Ia PB kinase.
  • a compound of the present invention can be used to treat a disease or disorder arising from abnormal cell growth, function or behaviour associated with PB kinase. Examples of such diseases and disorders are discussed by Drees et al in Expert Opin. Ther.
  • Patents (2004) 14(5):703 - 732 These include cancer, immune disorders, cardiovascular disease, viral infection, inflammation, metabolism/endocrine disorders and neurological disorders. Exampes of metabolism/endocrine disorders include diabetes and obesity. Examples of cancers which the present compounds can be used to treat include leukaemia, brain tumours, renal cancer, gastric cancer and cancer of the skin, bladder, breast, uterus, lung, colon, prostate, ovary and pancreas.
  • a human or animal patient suffering from an immune disorder, cancer, cardiovascular disease, viral infection, inflammation, a metabolism/endocrine disorder or a neurological disorders may thus be treated by a method comprising the administration thereto of a compound of the present invention as defined above.
  • the condition of the patient may thereby be improved or ameliorated.
  • the compounds of the invention exhibit physicochemical and pharmacokinetic properties which make them particularly well adapted for drug use. This is shown for instance in the results of the biological assays described in Example 11, which follows.
  • a compound of the present invention can be administered in a variety of dosage forms, for example orally such as in the form of tablets, capsules, sugar- or film-coated tablets, liquid solutions or suspensions or parenterally, for example intramuscularly, intravenously or subcutaneously.
  • the compound may therefore be given by injection or infusion.
  • the dosage depends on a variety of factors including the age, weight and condition of the patient and the route of administration. Daily dosages can vary within wide limits and will be adjusted to the individual requirements in each particular case. Typically, however, the dosage adopted for each route of administration when a compound is administered alone to adult humans is 0.0001 to 50 mg/kg, most commonly in the range of 0.001 to 10 mg/kg, body weight, for instance 0.01 to 1 mg/kg. Such a dosage may be given, for example, from 1 to 5 times daily.
  • a suitable daily dose is from 0.0001 to 1 mg/kg body weight, preferably from 0.0001 to 0.1 mg/kg body weight.
  • a daily dosage can be administered as a single dosage or according to a divided dose schedule.
  • a compound is formulated for use as a pharmaceutical or veterinary composition also comprising a pharmaceutically or veterinarily acceptable carrier or diluent.
  • the compositions are typically prepared following conventional methods and are administered in a pharmaceutically or veterinarily suitable form.
  • the compound may be administered in any conventional form, for instance as follows:
  • compositions intended for oral use may be prepared according to any method known in the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavouring agents, colouring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, dextrose, saccharose, cellulose, corn starch, potato starch, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, maize starch, alginic acid, alginates or sodium starch glycolate; binding agents, for example starch, gelatin or acacia; lubricating agents, for example silica, magnesium or calcium stearate, stearic acid or talc; effervescing mixtures; dyestuffs, sweeteners, wetting agents such as lecithin, polysorbates or lauryl sulphate.
  • inert diluents such as calcium carbonate, sodium carbonate, lactose, dextrose, saccharose, cellulose
  • the tablets may be uncoated or they may be coated by known techniques to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period.
  • a time delay material such as glyceryl monostearate or glyceryl distearate may be employed.
  • Such preparations may be manufactured in a known manner, for example by means of mixing, granulating, tableting, sugar coating or film coating processes.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is present as such, or mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil.
  • an inert solid diluent for example, calcium carbonate, calcium phosphate or kaolin
  • water or an oil medium for example, peanut oil, liquid paraffin, or olive oil.
  • Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions.
  • excipients are suspending agents, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinylpyrrolidone gum tragacanth and gum acacia; dispersing or wetting agents may be naturally-occurring phosphatides, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides for example polyoxyethylene sorbitan monooleate.
  • the said aqueous suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate, one or more colouring agents, such as sucrose or saccharin.
  • preservatives for example, ethyl or n-propyl p-hydroxybenzoate
  • colouring agents such as sucrose or saccharin.
  • Oily suspension may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
  • the oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol.
  • Sweetening agents such as those set forth above, and flavouring agents may be added to provide a palatable oral preparation. These compositions may be preserved by this addition of an antioxidant such as ascorbic acid.
  • Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavouring and colouring agents, may also be present.
  • the pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions.
  • the oily phase may be a vegetable oil, for example olive oil or arachis oils, or a mineral oil, for example liquid paraffin or mixtures of these.
  • Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally occuring phosphatides, for example soy bean lecithin, and esters or partial esters derived from fatty acids an hexitol anhydrides, for example sorbitan mono-oleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate.
  • the emulsion may also contain sweetening and flavouring agents.
  • Syrups and elixirs may be formulated with sweetening agents, for example glycerol, sorbitol or sucrose.
  • a syrup for diabetic patients can contain as carriers only products, for example sorbitol, which do not metabolise to glucose or which only metabolise a very small amount to glucose.
  • Such formulations may also contain a demulcent, a preservative and flavouring and coloring agents;
  • sterile injectable aqueous or oleaginous suspensions This suspension may be formulated according to the known art using those suitable dispersing of wetting agents and suspending agents which have been mentioned above.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non- toxic paternally- acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
  • Suitable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution, hi addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid find use in the preparation of injectables;
  • a suitable non-irritating excipient which is solid at ordinary temperature but liquid at the rectal temperature and will therefore melt in the rectum to release the drug.
  • Such materials are cocoa butter and poly-ethylene glycols
  • Ammoniacal methanol was prepared by bubbling ammonia (30- 32g) through methanol (19OmL) cooled in ice- water. To the solution was added 3-(3-methoxy- benzoylamino)-thiophene-2-carboxylic acid methyl ester (18.00g, 61.8mmol) and the mixture heated at 90 0 C for 44 hours in a steel bomb at 50psi. The solvent was then removed in vacuo and isopropanol (30OmL) added followed by 2M aqueous sodium hydroxide (4.0eq., 124mL). The solution was heated under reflux for 15 hours and then cooled in ice-water.
  • 2-Dimethylamino-l- ⁇ 4-[2-(3-hydroxy-phenyI)-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-6-ylmethyl]-piperazin-l-yl ⁇ -ethanone was prepared from 2-dimethylamino-l-piperazin-l-yl-ethanone. This in turn was prepared from 1-BOC- piperazine and bromoacetyl bromide in dichloromethane, followed by reaction with dimethylamine hydrochloride and subsequent deprotection of the BOC-group using HCl (2M solution in ether) in dichloromethane.
  • Example 4 1 - (4- [2-(3-Hydroxy-phen vD-4-morpholin-4-yI-thieno [3,2- d1pyrimidin-6-ylmethvn-piperazin-l-vU-2-methoxy-ethanone (37)
  • a mixture of 3-(4-morpholin-4-yl-6-piperazin-l-ylmethyl-thieno[3,2- d]pyrimidin-2-yl)-phenol, trifluoroacetic acid salt (50mg, 0.08mmol), dichloromethane (ImL), triethylamine (45 ⁇ L) and methoxyacetyl chloride (19 ⁇ L, 2.5eq.) was stirred at room temperature. After 3 hours the reaction mixture was diluted with dichloromethane, washed with sodium bicarbonate solution, dried
  • Example 5 l- ⁇ (2R,6S)-4-[2-(3-hvdroxy-phenyl)-4-morpholin-4-yl-thienor3,2- d1pyrimidin-6-ylmethyll-2,6-dimethyI-piperazin-l-yl ⁇ -ethanone (53) To a solution of 2-[3-(tert-butyl-dimethyl-silanyloxy)-phenyl]-6-((3R,5S)- 3,5-dimethyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine
  • the title compound 52 was prepared by analogy with the process of Example 5, using methane sulfonyl chloride.
  • reaction mixture was quenched with silica, evaporated in vacuo and purified by chromatography to give ⁇ l-[2-(3-hydroxy-phenyl)-4- mo ⁇ holin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]-piperidin-4-yl ⁇ -carbamic acid tert-butyl ester as a white solid (179 mg, 88%).
  • Example 8 N- ⁇ l-[2-(3-Hvdroxy-phenyl)-4-morpholin-4-yl-thieno[3,2- dlpyrimidin-6-vIinethyll-piperidin-4-vU-inethanesulfonamide
  • EC 50 values were calculated using a sigmoidal dose response curve fit. All the compounds tested had an EC 50 S of 5OuM or less in the range of cell lines utilized.
  • Plasma Protein Binding Solutions of test compound (5um, 0.5% final DMSO concentration) were prepared in buffer and 10% plasma (v/v in buffer).
  • a 96 well HT dialysis plate was assembled so that each well was divided in two by a semi-permeable cellulose membrane.
  • the buffer solution was added to one side of the membrane and the plasma solution to the other side; incubations were then conducted at 37 0 C over 2 hours in triplicate.
  • the cells were subsequently emptied, and the solutions for each batch of compounds were combined into two groups (plasma-free and plasma- containing) then analysed by LC-MSMS using two sets of calibration standards for plasma- free (6 points) and plasma-containing solutions (7 points).
  • Compounds of the invention were evaluated for their ability to modulate rubidium efflux from HEK-294 cells stably expressing hERG potassium channels using established flux methodology.
  • Cells were prepared in medium containing RbCl and were plated into 96-well plates and grown overnight to form monolayers.
  • the efflux experiment was initiated by aspirating the media and washing each well with 3 x lOO ⁇ L of pre-incubation buffer (containing low [K + ]) at room temperature. Following the final aspiration, 50 ⁇ L of working stock (2x) compound was added to each well and incubated at room temperature for 10 minutes. 50 ⁇ L of stimulation buffer (containing high [K+]) was then added to each well giving the final test compound concentrations.
  • Example 12 Tablet composition Tablets each weighing 0.15 g and containing 25 mg of a compound of the invention were manufactured as follows:
  • the active compound, lactose and half of the corn starch were mixed. The mixture was then forced through a sieve 0.5 mm mesh size. Corn starch (10 g) is suspended in warm water (90 ml). The resulting paste was used to granulate the powder. The granulate was dried and broken up into small fragments on a sieve of
  • Active compound 200 mg Hydrochloric Acid Solution 0.1M or
  • the compound of the invention was dissolved in most of the water (35° 40° C) and the pH adjusted to between 4.0 and 7.0 with the hydrochloric acid or the sodium hydroxide as appropriate.
  • the batch was then made up to volume with water and filtered through a sterile micropore filter into a sterile 10 ml amber glass vial (type 1) and sealed with sterile closures and overseals.
  • the active compound was dissolved in the glycofurol.
  • the benzyl alcohol was then added and dissolved, and water added to 3 ml.
  • the mixture was then filtered through a sterile micropore filter and sealed in sterile 3 ml glass vials (type
  • the compound of the invention was dissolved in a mixture of the glycerol and most of the purified water. An aqueous solution of the sodium benzoate was then added to the solution, followed by addition of the sorbital solution and finally the flavour. The volume was made up with purified water and mixed well.

Abstract

Fused pyrimidines of formula (1); wherein A represents a thiophene or a furan ring; n is 1 or 2; R1 is a group of formula (); wherein m is 0 or 1; R30 is H or C1-C6 alkyl; R4 and R5 form, together with the N atom to which they are attached a 5- or 6- membered saturated N-containing heterocyclic group which includes 0 or 1 additional heteroatoms selected from N, S and O, which may be fused to a benzene ring and which is unsubstituted or substituted; or one of R4 and R5 is alkyl and the other is a 5- Or 6-membered saturated N-containing heterocyclic group as defined above; R2 is selected from a) formula (3) wherein R6 and R7 form, together with the nitrogen atom to which they are attached, a morpholine, thiomorpholine, piperidine, piperazine, oxazepane, or thiazepane group which is unsubstituted or substituted; and b) formula (4) wherein Y is a C2-C4 alkylene chain which contains, between constituent carbon atoms of the chain and/or at one or both ends of the chain, 1 or 2 heteroatoms selected from O, N and S, and which is unsubstituted or substituted; and R3 is selected from: (a) a group of the following formula: (5) wherein B is a phenyl ring which is unsubstituted or substituted and Z is selected from H, -OR, -SR, CH2OR, -CO2R, CF2OH, CH(CF3)OH, C(CF3)2OH, -(CH2)qOR, -(CH2)qNR2, -C(O)N(R)2, -NR2, -NRC(O)R, -S(O)mN(R)2, -OC(O)R, OC(O)N)2, -NRS(O)mR, -NRC(O)N(R)2, CN, halogen and -NO2, wherein each R is independently selected from H, C1-C6 alkyl, C3 - C10 cycloalkyl and a 5- to 12- membered aryl or heteroaryl group, the group being unsubstituted or substituted, m is 1 or 2 and q is 0, 1 or 2; (b) a heteroaryl group which contains, 1, 2, 3 or 4 ring nitrogen atoms and 0, 1 or 2 additional heteroatoms selected from O and S, which group is monocyclic or bicyclic and which is unsubstituted or substituted; and (c) a group comprising a benzene ring which is unsubstituted or substituted and which is fused to a heteroaryl group as defined above; or a pharmaceutically acceptable salt thereof; provided that R3 is not an indole group or an indazole group, which group is unsubstituted or substituted; and the pharmaceutically acceptable salt thereof have activity as inhibitors of P13K and may thus be used to treat diseases and disorders arising from abnormal cell growth, function or behaviour associated with P13 kinase such as cancer, immune disorders, cardiovascular disease, viral infection, inflammation, metabolism/endocrine disorders and neurological disorders. Processes for synthesizing the compounds are also described.

Description

PHARMACEUTICAL COMPOUNDS
Field of the Invention The present invention relates to pyrimidine derivatives and their use as inhibitors of phosphatidylinositol 3-kinase (P 13 K).
Background to the Invention
Phosphatidylinositol (hereinafter abbreviated as "PI") is one of a number of phospholipids found in cell membranes. In recent years it has become clear that PI plays an important role in intracellular signal transduction. In the late 1980s, a PI3 kinase (PI3K) was found to be an enzyme which phosphorylates the 3-position of the inositol ring of phosphatidylinositol (D. Whitman et al, 1988, Nature, 332, 664).
PI3K was originally considered to be a single enzyme, but it has now been clarified that a plurality of subtypes are present in PI3K. Each subtype has its own mechanism for regulating activity. Three major classes of PI3Ks have been identified on the basis of their in vitro substrate specificity (B. Vanhaesebroeck,1997, Trend in Biol. Sci, 22, 267). Substrates for class I PI3Ks are PI, PI 4-phosphate (PI4P) and PI 4,5-biphosphate (PI (4,5)P2). Class I PI3Ks are further divided into two groups, class Ia and class Ib, in terms of their activation mechanism. Class Ia PBKs include PI3K pi 10a, pi lOβ and pi lOδ subtypes, which transmit signals from tyrosine kinase-coupled receptors. Class Ib PI3K includes a pi lOγ subtype activated by a G protein-coupled receptor. PI and PI(4)P are known as substrates for class II PDKs. Class II PDKs include PDK C2α, C2β and C2γ subtypes, which are characterized by containing C2 domains at the C terminus. The substrate for class III PDKs is PI only.
In the PDK subtypes, the class Ia subtype has been most extensively investigated to date. The three subtypes of class Ia are heterodimers of a catalytic 110 kDa subunit and regulatory subunits of 85 kDa or 55 kDa. The regulatory subunits contain SH2 domains and bind to tyrosine residues phosphorylated by growth factor receptors with a tyrosine kinase activity or oncogene products, thereby inducing the PBK activity of the pi 10 catalytic subunit which phosphorylates its lipid substrate. Thus, the class Ia subtypes are considered to be associated with cell proliferation and carcinogenesis. WO 01/083456 describes a series of condensed heteroaryl derivatives which have activity as inhibitors of PB K and which suppress cancer cell growth.
Summary of the Invention
It has now been found that a novel class of fused pyrimidine compounds are effective inhibitors of PBK with drug-like physicochemical and pharmacokinetic properties. The compounds exhibit selectivity for class Ia PBKs over class Ib.
Accordingly, the present invention provides a compound which is a fused pyrimidine of formula (I):
Figure imgf000004_0001
wherein
A represents a thiophene or furan ring; n is 1 or 2;
R1 is a group of formula:
Figure imgf000004_0002
wherein m is 0 or 1;
R30 is H or Ci-C6 alkyl;
R4 and R5 form, together with the N atom to which they are attached, a 5- or 6- membered saturated N-containing heterocyclic group which includes 0 or 1 additional heteroatoms selected from N, S and O, which may be fused to a benzene ring and which is unsubstituted or substituted; or one of R4 and R5 is alkyl and the other is a 5- or 6-membered saturated N-containing heterocyclic group as defined above or an alkyl group which is substituted by a 5- or 6-membered saturated N- containing heterocyclic group as defined above; R2 is selected from: (a)
R0
— N
R'
wherein R6 and R7 form, together with the nitrogen atom to which they are attached, a morpholine, thiomorpholine, piperidine, piperazine, oxazepane or thiazepane group which is unsubstituted or substituted; and
(b)
Figure imgf000005_0001
wherein Y is a C2 - C4 alkylene chain which contains, between constituent carbon atoms of the chain and/or at one or both ends of the chain, 1 or 2 heteroatoms selected from O, N and S, and which is unsubstituted or substituted; and R3 is selected from: (a) a group of the following formula:
Figure imgf000005_0002
wherein B is a phenyl ring which is unsubstituted or substituted and Z is selected from H, -OR, -SR, CH2OR, -CO2R, CF2OH, CH(CF3)OH, C(CF3)2OH, -(CH2)qOR, -(CH2)qNR2 , -C(O)N(R)2, -NR2, -NRC(O)R, -S(O)1nN(R)2, -OC(O)R, OC(O)N(R)2, -NRS(O) mR , -RC(O)N(R)2, CN, halogen and -NO2, wherein each R is independently selected from H, Ci-C6 alkyl, C3 - Ci0 cycloalkyl and a 5- to 12- membered aryl or heteroaryl group, the group being unsubstituted or substituted, m is 1 or 2 and q is 0, 1 or 2;
(b) a heteroaryl group which contains 1 , 2, 3 or 4 ring nitrogen atoms and 0, 1 or 2 additional heteroatoms selected from O and S, which group is monocyclic or bicyclic and which is unsubstituted or substituted; and
(c) a group comprising a benzene ring which is unsubstituted or substituted and which is fused to a heteroaryl group as defined above; provided that R3 is not an indole group or an indazole group, which group is unsubstituted or substituted; or a pharmaceutically acceptable salt thereof.
Detailed description of the Invention
The thiophene or furan ring A in formula (I) adopts either of the two available regiochemical orientations. Formula (I) thus covers the thieno[3,2- djpyrimidines and furano[3,2-d]pyrimidines of the following formula (Ia) as well as the thieno[2,3-d]pyrimidines and furano[2,3-d]pyrimidines of the following formula (Ib):
Figure imgf000006_0001
wherein each of R1 to R3 and n is as defined above and X is S or O.
In formula (I), the group or groups R1 , which are the same or different in a given compound when n is 2, may be bonded to either or both of the two available ring positions on the thiophene or furan ring A. Referring to structures (Ia) and (Ib) above, therefore, when n is 1 the furan or thiophene ring is mono-substituted by R1 at the 2-position or the 3-position. When n is 2, the thiophene or furan ring is di- substituted by R1 at positions 2 and 3.
As specified herein, an alkyl group is a straight or branched chain saturated hydrocarbon radical which is unsubstituted or substituted. Typically it is Ci-C20 alkyl, for instance Ci-Cio alkyl, such as Ci-C6 alkyl or Ci-C4 alkyl, for example methyl, ethyl, i-propyl, n-propyl, t-butyl, s-butyl or n-butyl. It may also be pentyl, hexyl, heptyl, octyl and the various branched chain isomers thereof.
When an alkyl group is substituted it typically bears one or more substituents R20 selected from halogen, alkoxy, carbocyclyl, a 5- or 6-membered saturated N- containing heterocyclic group as defined above, OH, SR, CN, nitro, NR2, -COOR, - C(O)R, S(O)mR and -CONR2, wherein each R is H, unsubstituted alkyl or C3-Ci0 cycloalkyl and m is 1 or 2. It is, for instance, a haloalkyl group or a group -alk- N(R4)(R5) wherein alk is an alkylene chain and R4 and R5 form, together with the N atom to which they are attached, a 5- or 6-membered saturated N-containing heterocyclic group which includes 0 or 1 additional heteroatoms selected from N, S and O, which may be fused to a benzene ring and which is unsubstituted or substituted. Typically R20 is selected from halogen, alkoxy, carbocyclyl, a 5- or 6- membered saturated N-containing heterocyclic group as defined above, OH, CN, NR2 , -COOR and -CONR2, wherein each R is H or unsubstituted alkyl as defined above. It is, for instance, a haloalkyl group or a group -alk-N(R6)(R5) wherein alk is an alkylene chain and R4 and R5 form, together with the N atom to which they are attached, a 5- or 6-membered saturated N-containing heterocyclic group as defined above. An alkylene group is unsubstituted or substituted, straight or branched chain saturated divalent hydrocarbon group. Typically it is Ci-C8 alkylene, for instance Ci-C6 alkylene. Preferably it is Ci-C4 alkylene, for example C2-C4 alkylene, such as methylene, ethylene, i-propylene, n-propylene, t-butylene, s-butylene or n-butylene. It may also be pentylene, hexylene, heptylene, octylene and the various branched chain isomers thereof. When the alkylene group is substituted it is typically substituted by a group R20 as defined above. An alkenyl group is an unsubstituted or substituted, straight or branched chain hydrocarbon radical having one or more double bonds. Typically it is C2-C8 alkenyl, for instance C2-C6 alkenyl, such as allyl, butenyl, butadienyl, pentenyl or hexenyl. When the alkenyl group is substituted it is typically substituted by a group R20 as defined above or by alkyl which is unsubstituted or substituted by a group R20 as defined above.
An alkynyl group is an unsubstituted or substituted, straight or branched chain hydrocarbon radical having one or more triple bonds. Typically it is C2-C8 alkynyl, for instance C2-C6 alkynyl, such as ethynyl, propynyl or butynyl. When the alkynyl group is substituted it is typically substituted by a group R20 as defined above or by alkyl which is unsubstituted or substituted by a group R20 as defined above.. A haloalkyl group is an alkyl group as defined above, substituted by one or more halogen atoms. It can be a perhaloalkyl group, for instance trifiuoromethyl or perfluorohexyl.
A halogen is chlorine, fluorine, bromine or iodine. It is typically bromine or iodine.
An alkoxy group is typically Ci-C6 alkoxy, for instance Ci-C4 alkoxy, such as methoxy, ethoxy, i-propoxy, n-propoxy, t-butoxy, n-butoxy or s-butoxy. It is unsubstituted or substituted, for instance by a group R20 as defined above or by alkyl which is unsubstituted or substituted by a group R20 as defined above or by alkyl which is unsubstituted or substituted by a group R20 as defined above.. Typically it is substituted by carbocyclyl, morpholino, OH, CN, NR2 , -COOR or -CONR2, wherein each R is H or unsubstituted alkyl as defined above.
A carbocyclyl group is a non-aromatic saturated or unsaturated monocyclic hydrocarbon ring, typically having from 3 to 10 carbon atoms. It may be a C3-C8 cycloalkyl group, or C5-Ci0 cycloalkyl group, for instance cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl or cyclooctyl. Alternatively it may be a cycloalkenyl group, typically C4-C8 cycloalkenyl, for instance cylcopentenyl, cyclohexenyl, cyclohexadienyl, cycloheptenyl, cyclohepadienyl, cyclooctenyl or cyclooctadienyl. A carbocyclyl group may be unsubstituted or substituted, for instance by a group R20 as defined above or by alkyl which is unsubstituted or substituted by a group R20 as defined above.. Typically it is substituted by alkoxy, morpholino, OH, CN, NR2 , -COOR and -CONR2, wherein each R is H or unsubstituted alkyl as defined above.
A 5- or 6-membered saturated N-containing heterocyclic group which includes 0 or 1 additional heteroatoms selected from N, S and O, which may be fused to a benzene ring and which is unsubstituted or substituted is typically selected from morpholine, piperidine, piperazine, pyrrolidine, thiomorpholine, quinoline, isoquinoline, diazepane, oxazepane and thiazepane.
When a 5- or 6-membered saturated N-containing heterocyclic group as defined above is substituted it is typically substituted by one or more substituents, for instance 1, 2 or 3 substituents, typically by 1 or 2 substituents. Typically the substituents are selected from alkyl which is unsubstituted or substituted, alkoxy which is unsubstituted or substituted, -NR2, -N(R'")-alk-OR, -alk-OR, -O-alk-OR, -alk-C(O)NR2, -C(O)NR2, -alk-Het, -N(R)-Het, -O-Het, -N(R)-C(O)-alk-OR, -C(O)-N(R)-alk-OR, -alk-S(O)2R, -N(R)-alk-OR, -alk-NR'R", -N(R'")-S(O)2R, S(O)2R'", -alk-N(R)-alk-OR, -S(O)2-alk-OR, a second 5- or 6-membered saturated N-containing heterocyclic group as defined above, a 5- or 6-membered N-containing heteroaryl group which is unsubstituted or substituted and which may be fused to a benzene ring, -COOR, -CONR2, oxo (=0), -SO2NR2 , -SO2-alk-NR2 and -CO-alk-OR, wherein: alk is an alkylene chain as defined above; Het is a 5- or 6- membered N-containing heteroaryl group as defined herein which is unsubstituted or substituted; R is H or alkyl, or when two groups R are bonded to N they may form, together with the N atom, a saturated 5- or 6-membered N-containing heterocyclic group as defined herein which is unsubstituted or substituted; each of R' and R" is independently H, alkyl or alkoxy; and R'" is alkyl which is unsubstituted or substituted, for instance by CF3, NR2 , OR, a 5- or 6-membered saturated N- containing heterocyclic group as defined herein or a 5- or 6-membered N-containing heteroaryl group as defined herein, the said heterocyclic and heteroaryl groups being unsubstituted or substituted. It may be substituted by a group R20 as defined above or by alkyl which is unsubstituted or substituted by a group R20 as defined above. Typically a 5- or 6-membered saturated N-containing heterocyclic group as defined above is substituted by a group selected from alkyl which is unsubstituted or substituted, alkoxy which is unsubstituted or substituted, a second 5- or 6-membered saturated N-containing heterocyclic group as defined above, a 5- or 6-membered N- containing heteroaryl group which is unsubstituted or substituted and which may be fused to a benzene ring, -COOR, -CONR2, -CONR, oxo (=0), OH, -NSO2R, -
SO2NR2 or -CO(CH2)nOR wherein R is H or alkyl, -NR'R" wherein each of R' and R" is independently H, alkyl or alkoxy, and -SO2R'" wherein R'" is alkyl which is unsubstituted or substituted, for instance by NR2 or a 5- or 6-membered saturated N- containing heterocyclic group as defined above. More typically a 5- or 6-membered saturated N-containing heterocyclic group is substituted by one or more substituents selected from alkyl as defined above which is unsubstituted or substituted (for instance by R20 as defined above), haloalkyl as defined above, alkoxy as defined above which is unsubstituted or substituted, halogen, hydroxy, CN, nitro, amino, oxo (=0), and -NR'R" wherein each of R' and R" is independently H or alkyl.
A 5-, 6- or 7-membered saturated heterocyclic group which contains 1 or 2 heteroatoms selected from N, S and O and which is unsubstituted or substituted is typically selected from tetrahydropyran, tetrahydrothiopyran, tetrahydrofuran and tetrahydrothiofuran. When a 5-, 6- or 7-membered saturated heterocyclic group which contains 1 or 2 heteroatoms selected from N, S and O is substituted it may be substituted as specified above for a 5- or 6-membered saturated N-containing heterocyclic group.
A heteroaryl group is a heteroaryl group which contains 1 , 2 3 or 4 ring nitrogen atoms and O, 1 or 2 additional heteroatoms selected from O, N and S, which group is monocyclic or bicyclic and which is unsubstituted or substituted. It is typically a 5- to 12-membered ring. Examples of a heteroaryl group include pyrrole, pyrazole, triazole, tetrazole, indazole, thiazole, isothiazole, oxazole, isooxazole, indole, isoindole, l,3-dihydro-indol-2-one, pyridine-2-one, pyridine, pyridin-3-ol, imidazole, 1,3-dihydro-benzimidazolone, benzimidazole, benzothiazole, benzothiadiazole, quinoline, isoquinoline, quinoxaline, pyrazolopyridine, aminopyrazolinone, imidazopyridine, pyrimidine, pyridazine, pyrazine and isatin groups. Preferred examples include indazole, indole, pyrazole and tetrazole groups. These groups may be unsubstituted or substituted, for instance by a group R20 as specified above or by alkyl which is unsubstituted or substituted by a group R20 as defined above.
A 5- or 6-membered N containing heteroaryl group which may be fused to a benzene ring is typically selected from pyrrole, pyrazole, triazole, tetrazole, indazole, thiazole, isothiazole, oxazole, isooxazole, indole, isoindole, 1,3- dihydro-indol-2-one, pyridine-2-one, pyridine, pyridin-3-ol, imidazole, 1,3-dihydro- benzimidazolone, benzimidazole, benzothiazole, benzothiadiazole, quinoline, isoquinoline, quinoxaline, pyrazolopyridine, aminopyrazolinone, imidazopyridine, pyrimidine, pyridazine and pyrazine. When such a heteroaryl group is substituted it may be substituted by a group R20 as defined above or by alkyl which is unsubstituted or substituted by a group R20 as defined above. In R1 , m is 0 or 1 , typically 1. R30 is typically H. R4 and R5 typically form, together with the N atom to which they are attached, a saturated N-containing heterocyclic group selected from morpholine, thiomorpholine, piperidine, piperazine, pyrrolidine, quinoline, isoquinoline, diazepane, oxazepane and thiazepane. The heterocylic group formed by R4 and R5 is unsubstituted or substituted, for instance by the examples of substituent groups listed above, such as a group R20 as defined above or by alkyl which is unsubstituted or substituted by a group R20 as defined above.
In definition (a) of R2 in formula (I), the ring formed by R6 and R7 is typically morpholine which is unsubstituted or substituted, for instance by a group R20 as specified above or by alkyl which is unsubstituted or substituted by a group R20 as defined above.. It may alternatively be a group selected from tetrahydropyran, tetrahydrothiopyran, tetrahydrofuran and tetrahydrothiofuran, each of which is unsubstituted or substituted, for instance, for instance by a group R20 as specified above or by alkyl which is unsubstituted or substituted by a group R20 as defined above.. When the ring formed by R6 and R7 is substituted it may be substituted on either a ring heteroatom or a ring carbon atom, for instance by a group R20 as defined above.
In definition (b) of R2 in formula (I), the alkylene chain represented by Y forms, together with the carbon atoms to which it is attached, a saturated 5-, 6- or 7- membered heterocyclic ring which contains 1 or 2 heteroatoms selected from O, N and S and which is unsubstituted or substituted. Examples of the heterocyclic ring include tetrahydropyran, tetrahydrofuran, tetrahydrothiopyran, tetrahydrothiofuran and morpholine. When the heterocyclic ring is substituted it is typically substituted by one or more substituents, for instance 1, 2 or 3 substituents, selected from halogen, alkyl, haloalkyl (for instance trifluoromethyl), alkoxy, OH, CN, NR2 , oxo (=0), -COOR and -CONR2, wherein each R is H or unsubstituted alkyl as defined above.
In the definition (a) for R3 the phenyl ring B is unsubstituted (apart from group Z) or substituted. When it is unsubstituted the group Z is the sole substituent. When it is substituted it typically comprises, in addition to group Z, one or more substituents selected from halo, alkyl, alkenyl, alkynyl, CN, NO2 , OR', SR', NR'2> C(O)R', SOR', SO2 R', SO2NR'2 , NC(O)R' and CO2 R', wherein each R' is independently H or Ci-C6 alkyl.
Group Z is bonded to any available ring position on the phenyl ring B. Thus it may be situated at the 2-, 3-, A-, 5- or 6- position of the phenyl ring. Typically it is bonded at position 3 or 4. Z is most typically other than H, such that moiety -BZ is a substituted phenyl ring. A typical example of Z is a group OR as defined above, in particular OH. In this embodiment the OR group, or OH group, is typically bonded at ring position 3 or 4 of phenyl ring B. Typically -BZ is a 3-hydroxyphenyl or 4- hydroxyphenyl group, or an isostere thereof, other than an indole or indazole group which is unsubstituted or substituted. An isostere as used herein is a functional group which possesses binding properties which are the same as, or similar to, the 3-hydroxyphenyl or 4- hydroxyphenyl group in the context of the structure of formula (I). Isosteres of
3-hydroxyphenyl and 4-hydroxyphenyl groups are encompassed within definitions (b) and (c) above for R3 .
In definition (b) for R3 the heteroaryl group is unsubstituted or substituted. If it is substituted it may be substituted by one or more substituents selected from a group Z, R20 as defined above, alkyl which is unsubstituted or substituted by a R20 as defined above, any group specified above as an additional substituent on the phenyl ring B, and an oxo group (=0). Typically, if substituted, the heteroaryl group is substituted by OH, NH2 or an oxo group. In one embodiment the heteroaryl group is unsubstituted.
In definition (c) for R3 the benzene ring is unsubstituted or substituted. If it is substituted it may be substituted by one or more substituents selected from a group Z , R20 as defined above, alkyl which is unsubstituted or substituted by R20 as defined above, and any of the groups specified above as an additional substituent on the phenyl ring B. The heteroaryl group to which the benzene ring is fused is itself unsubstituted or substituted, for instance by a group Z , R20 or alkyl which is unsubstituted or substituted by a group R20 as defined above; by any group specified above as an option for an additional substituent on the phenyl ring B; or by an oxo group (=0). In one embodiment both the benzene ring and the heteroaryl group are unsubstituted.
Examples of the groups included in definitions (b) and (c) for R3 include pyrrole, pyrazole, triazole, tetrazole, thiazole, isothiazole, oxazole, isooxazole, isoindole, l,3-dihydro-indol-2-one, pyridine-2-one, pyridine, pyridin-3-ol, imidazole, 1,3-dihydro-benzimidazolone, benzimidazole, benzothiazole, benzothiadiazole, quinoline, isoquinoline, quinoxaline, pyrazolopyridine, aminopyrazolinone, imidazopyridine, pyrimidine, pyridazine, pyrazine and isatin groups. Preferred examples include pyrazole and tetrazole groups. These groups may be unsubstituted or substituted, for instance by a group Z , R20 or alkyl which is unsubstituted or substituted by a group R20 as defined above. Typically these groups are isosteres. More specifically, groups included in definitions (b) and (c) for R3 as defined above include the following structures, which are typically isosteres as defined above:
Figure imgf000014_0001
1 i 1 Jj
Figure imgf000014_0002
1iii 1 iv
Figure imgf000014_0003
2 iv 2 v
Figure imgf000015_0001
3iv 3v 3 vi
Figure imgf000015_0002
3vii 3 viii 3ix
Figure imgf000016_0001
4i
Figure imgf000016_0002
5ii
Figure imgf000016_0003
Figure imgf000016_0004
Figure imgf000016_0005
6v wherein each R10 is independently selected from H, Ci-C6 alkyl, Ci-C6 alkoxy, Ci-C6 acyl, -C(O)NR5R", -S(O)tNR'R", aryl, heteroaryl, sulphonyl and halogen, whrein R' and R" are each independently H or CpC6 alkyl and t is 1 or 2; each R1 ' is independently selected from -OR10 and -N(R10 )2, wherein R10 is as defined above; each R12 is independently H, F or CF3; each W is independently selected from CR10 and N, wherein R10 is as defined above; and W is selected from O, S and NR12 wherein R12 is as defined above. with the proviso that formulae (Ii) and (lii) are other than indole and indazole. Specific examples of compounds of the invention include:
3-(4-Morpholin-4-yl-6-morpholin-4-ylmethyl-thieno[3,2-d]pyrimidin-2-yl)-phenol;
3-(4-Morpholin-4-yl-6-pyrrolidin-l-ylmethyl-thieno[3,2-d]pyrimidin-2-yl)-phenol;
3-[6-(4-Methyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-2- yl]-phenol; 3-[6-(6,7-Dimethoxy-3,4-dihydro-lH-isoquinolin-2-ylmethyl)-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-2-yl]-phenol ;
3-(6-[ 1 ,4']Bipiperidinyl- r-ylmethyl-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-2-yl)- phenol;
3-[4-Morpholin-4-yl-6-(4-pyrimidin-2-yl-piperazin-l-ylmethyl)-thieno[3,2- d]pyrimidin-2-yl]-phenol;
3-[6-(4-Cyclohexylmethyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol;
4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazine-1-carboxylic acid tert-butyl ester; 3-(4-Morpholin-4-yl-6-piperazin-l-ylmethyl-thieno[3,2-d]pyrimidin-2-yl)-phenol , trifluoroacetic acid salt;
3-{6-[4-(2-Methoxy-ethyl)-piperazin-l-ylmethyl]-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl} -phenol;
3-[6-(4-Methyl-[l,4]diazepan-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin- 2-yl]-phenol; {4-[2-(3-Hydroxy-phenyl)-4-morpholin- 4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin-1-yl} -acetic acid ethyl ester;
1 - {4- [2-(3 -Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3 ,2-d]pyrimidin-6-ylmethyl] - piperazin- 1 -yl} -ethanone; 3-{6-[4-(2-Hydroxy-ethyl)-piperazin-l-ylmethyl]-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-2-yl} -phenol;
3-(4-Morpholin-4-yl-6-thiomorpholin-4-ylmethyl-thieno[3,2-d]pyrimidin-2-yl)- phenol;
3-[6-(4-Ethyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-2-yl]- phenol;
3 -(6- { [Methyl-( 1 -methyl-piperidin-4-yl)-amino]-methyl } -4-morpholin-4-yl- thieno[3,2-d]pyrimidin-2-yl)-phenol;
3-[6-(4-Dimethylamino-piperidin-l-ylmethyl)-4-niorpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol ; 4- [2-(3 -Hydroxy-phenyl)-4-morpholin-4-yl-thieno [3 ,2-d]pyrimidin-6-ylmethyl]- piperazin-2-one;
3-[6-(4-Methanesulfonyl-piperazin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol;
{4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin- l-yl}-acetonitrile;
3-[4-Mθφholin-4-yl-6-(4-moφholin-4-yl-piperidin-l-ylmethyl)-thieno[3,2- d]pyrimidin-2-yl]-phenol;
3-[6-(4-Amino-piperidin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-2- yl]-phenol; 3-[6-(3-Dimethylamino-pyrrolidin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol;
1 - [2-(3 -Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3 ,2-d]pyrimidin-6-ylmethyl] -A- moφholin-4-ylmethyl-piperidin-4-ol;
4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazine-1-carboxylic acid ethylamide ; N- { 1 -[2-(3-Hydroxy-phenyl)-4- moφholin-4-yl-thieno[3,2-d]pyrimidin-
6-ylmethyl] -piperidin-4-yl } -methanesulfonamide; l-{4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin- 1 -yl} -2-methoxy-ethanone; N-{1 -[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperidin-4-yl} -acetamide;
4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazine-1-carboxylic acid dimethylamide; l-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperidin-4-ol;
3-[6-(l,l-Dioxo-llambda*6*-thiomoφholin-4-ylmethyl)-4-moφholin-4-yl- thieno[3,2-d]pyrimidin-2-yl]-phenol;
4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazine-1 -sulfonic acid dimethylamide; 3-(6- {4-[(2-Methoxy-ethyl)-methyl-amino]-piperidin- 1 -ylmethyl} -4-moφholin-4-yl- thieno[3,2-d]pyrimidin-2-yl)-phenol; l-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- pyrrolidin-3-ol;
(i?,5)-3-[6-(2-Dimethylaminomethyl-pyrrolidin-l-ylmethyl)-4-moφholin-4-yl- thieno[3,2-d]pyrimidin-2-yl]-phenol;
(Λ^-S-Cό-ltMethyl-Cl-methyl-pyrrolidin^-ylmethyO-aminoJ-methyl}^- moφholin-4-yl-thieno[3,2-d]pyrimidin-2-yl)-phenol; l-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperidine-4-carboxylic acid amide; 2-{4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin- 1 -yl} -N,N-dimethyl-acetamide;
3 -(6- { [Methyl-( 1 -methyl-pyrrolidin-3 -yl)-amino]-methyl } -4-moφholin-4-yl- thieno[3,2-d]pyrimidin-2-yl)-phenol;
2-Dimethylamino- 1 - {4-[2-(3-hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-6-ylmethyl]-piperazin- 1 -yl} -ethanone; 3-[6-((3R,5S)-3,5-Dimethyl-piperazin-l- ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl] -phenol ;
3-[6-((3R,5S)-4-Methanesulfonyl-3,5-dimethyl-piperazin-l-ylmethyl)-4-morpholin-
4-yl-thieno[3,2-d]pyrimidin-2-yl]-phenol; 1 - {(2R,6S)-4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyl]-2,6-dimethyl-piperazin-l-yl}-ethanone;
3-{6-[4-(3-Dimethylamino-propane-l-sulfonyl)-piperazin-l-ylmethyl]-4-morpholin-
4-yl-thieno[3,2-d]pyrimidin-2-yl}-phenol;
3-[6-(4-Methoxy-piperidin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-2- yl] -phenol;
3-{4-Moφholin-4-yl-6-[4-(3-morpholin-4-yl-propane-l-sulfonyl)-piperazin-l- ylmethyl]-thieno[3,2-d]pyrimidin-2-yl}-phenol;
{4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin- 1 -yl} -moφholin-4-yl-methanone; 4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazine-1-carboxylic acid (2-methoxy-ethyl)-methyl-amide;
1 - {3-[6-(4-Methyl-piperazin- 1 -ylmethyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-
2-yl]-phenyl}-ethanol; .
3-[6-(4-Methyl-piperazin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-2- yl] -phenyl } -methanol ;
2-Chloro-5-[6-(4-methyl-piperazin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol;
2,3-Difluoro-5-[6-(4-methyl-piperazin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol; 4-Fluoro-3 - [6-(4-methyl-piperazin- 1 -ylmethyl)-4-moφholin-4-yl-thieno [3 ,2- d]pyrimidin-2-yl]-phenol; 2-( 1 H-Indazol-6-yl)-6-(4-methyl-piperazin- 1 -ylmethyl)-4-moφholin-4-yl- thieno[3,2-d] pyrimidin-2-yl]-phenol; and
6-(4-Methyl-piperazin- 1 -ylmethyl)-4-moφholin-4-yl-2-( 1 H-pyrazol-4-yl)- thieno[3,2-d]pyrimidine; and the pharmaceutically acceptable salts thereof. The compounds of formula (I) may exist in the form of geometrical isomers or tautomers depending on the kinds of substituent groups, and these isomers in separated forms or mixtures thereof may be used in the present invention. Where the compounds have asymmetric carbon atoms, optical isomer forms may exist based on such carbon atoms. All of the mixtures and the isolated forms of these optical isomers may be used in the present invention.
A suitable synthetic strategy for producing compounds of formula (I) in which m is 1 employs the precursor carboxaldehyde of formula (II):
Figure imgf000021_0001
wherein A and R2 are as defined above. Starting from this precursor the synthesis comprises performing, in either order, a palladium-mediated (Suzuki-type) cross- coupling reaction and a reductive amination. The present invention therefore further provides a process for producing a compound of formula (I) as defined above in which m is 1, which process comprises: (a) treating a compound of formula (II):
Figure imgf000021_0002
wherein A and R2 are as defined above, with a boronic acid or ester thereof of formula R3B(OR15)2 , in which R3 is as defined above and each R15 is H or Ci-C6 alkyl or the two groups OR15 form, together with the boron atom to which they are attached, a pinacolato boronate ester group, in the presence of a Pd catalyst; and treating the resulting compound of formula (III):
Figure imgf000022_0001
wherein A, R2 and R3 are as defined above, with an amine of formula NHR4R5 in which R4 and R5 are as defined above, in the presence of a suitable reducing agent; or
(b) treating a compound of formula (II) as defined above with an amine of formula NHR4R5 wherein R4 and R5 are as defined above, in the presence of a suitable reducing agent; and treating the resulting compound of formula (IV):
Figure imgf000022_0002
wherein A, R2 , R4 and R5 are as defined above, with a boronic acid or ester thereof of formula R3B(OR15)2 , in which R3 is as defined above and each R15 is H or Ci-C6 alkyl or the two groups OR15 form, together with the boron atom to which they are attached, a pinacolato boronate ester group, in the presence of a Pd catalyst.
Both the animation step and the Pd-mediated cross-coupling step take place under conventional conditions. The palladium catalyst may be any that is typically used for Suzuki-type cross-couplings, such as PdCl2(PPh3)2. The reducing agent is typically a borohydride, for instance NaBH(OAc)3, NaBH4 or NaCNBH4, in particular NaBH(OAc)3.
The invention further provides a process for producing a compound of formula (I) in which m is 1 and R3 is a 3- or 4-hydroxyphenyl group, which process comprises: (a) treating a compound of formula (V):
Figure imgf000023_0001
wherein OR' is bonded at position 3 or 4 of the phenyl ring to which it is attached, R' is a hydroxy protecting group and A and R2 are as defined above, with an amine of formula NHR4R5 wherein R4 and R5 are as defined above, in the presence of a suitable reducing agent; and
(b) removing the hydroxy protecting group.
The reducing agent is typically a borohydride, for instance as specified above.
Examples of hydroxy protecting groups are known in the art, for instance as described in "Protective Groups for Organic Chemistry", Third Edition, T.W. Greene and P.G.M. Wuts, John Wiley & Sons, 1999. For instance, a hydroxy group can be protected as an acetal, a substituted acetal, an ester, a xanthate, an ether or a silyl ether. The acetal is preferably tetrahydropyran. The silyl ether is preferably trimethylsilyl ether, t-butyl dimethylsilyl ether, triiso-propylsilyl ether or t- butyldiphenyl-silyl ether. These protecting groups are removed by conventional techniques.
A compound of formula (V) as defined above may be produced by a process which comprises treating a compound of formula (VI):
Figure imgf000023_0002
wherein A, R2 and R' are as defined above, with a lithiating agent followed by N,N'- dimethylformamide (DMF). The reaction is typically conducted by adding a solution of the lithiating agent in a non-polar organic solvent, for instance a hydrocarbon solvent such as hexane, to a suspension of the compound of formula (VI) in an organic solvent such as tetrahydrofuran (THF). If THF is used the addition takes place at a low temperature, of about -78°C. The lithiating agent is typically an alkyllithium, for instance n-butyllithium.
A compound of formula (VI) as defined above may be produced by a process which comprises treating a compound of formula (VII):
Figure imgf000024_0001
wherein A and R2 are as defined above, with a boronic acid of formula (VIII):
Figure imgf000024_0002
wherein R' and R15 are as defined above, in the presence of a palladium catalyst. The reaction is conducted under conventional conditions for a Suzuki-type cross-coupling reaction, for instance as described above.
A compound of formula (II) as defined above wherein R2 is -NR6R7 may be prepared by a process which comprises treating a compound of formula (IX):
Figure imgf000024_0003
wherein A, R6 and R7 are as defined above, with a lithiating agent followed by N,N'- dimethylformamide (DMF). The reaction is typically carried out as described above for the production of a compound of formula (V).
A compound of formula (IX) as defined above may be produced by a process which comprises treating a compound of formula (X):
Figure imgf000025_0001
with an amine of formula NHR6R7 , wherein R6 and R7 are as defined above, in an organic solvent. The solvent is typically an alcohol, such as methanol. The reaction is generally conducted at room temperature. A compound of formula (X) may be prepared by the process described in
Reference Example 1 for the preparation of 2,4-dichloro-thieno[3,2-d]pyrimidine, or by analogy with such a process.
A compound of formula (II) as defined above wherein R2 is of formula
CH CfH2 may be prepared by a process which comprises submitting a compound of formula (XI):
Figure imgf000025_0002
wherein A and R3 are as defined above, to palladium-mediated cross-coupling with a compound of formula (XII):
Figure imgf000025_0003
(xπ) wherein L is H or a group selected from halo, -OSO2CF3, -B(OR) 2, -Sn(R)3 and - Si(R)3 wherein R is H or alkyl as defined above, followed by reduction, to yield a compound of the following formula (XIII):
Figure imgf000026_0001
wherein A, R3 and Y are as defined above.
The compound of formula (XIII) may be converted to the corresponding carboxaldehyde by treatment with a lithiating agent followed by N,N'- dimethylformamide (DMF), for instance under the conditions described above for the conversion of a compound of formula (VI) to a compound of formula (V). The lithiating agent is typically as defined above. The resulting carboxaldehyde may then be converted into a desired final compound of formula (I) as defined above, in which m is 1 , by treatment with an amine of formula NHR4R5 in which R4 and R5 are as defined above, in the presence of a suitable reducing agent, for instance a borohydride as specified above, in particular NaBH(OAc)3.
A compound of formula (I) as defined above in which m is 0 may be prepared by a Buchwald-type palladium-mediated nitrogen insertion reaction. Such a process may comprise treating a compound of formula (XIV):
Figure imgf000026_0002
wherein A, R2 and R3 are as defined above and W is a halo group selected from Br and I, with an amine of formula NHR4R5 in which R4 and R5 are as defined above, in the presence of a palladium catalyst.
A compound of formula (XIV) may be produced by treating a compound of formula (XV):
Figure imgf000027_0001
wherein A, R2 and R3 are as defined above, with a lithiating agent and a halogen selected from bromine and iodine. The lithiating agent is typically an alkyllithium, for instance butyllithium. The halogen is typically iodine, which gives rise to a compound of formula (XIV) in which W is I.
A compound of formula (I) as defined above in which m is 0 may also be prepared by an SNAr displacement reaction, for instance under the conditions described by D. Prim and G. Kirsch in Tetrahedron 55 (21), 6511-6526, 1999. Such a process comprises treating a compound of formula (XIV) as defined above in which W is Br with an amine of formula NHR4R5 in which R4 and R5 are as defined above in H2O under reflux for 12 hours.
A compound of formula (I) as defined above in which m is 0 may alternatively be prepared by treating a compound of formula (XIV) as defined above in which W is I with an amine of formula NHR4R5 in which R4 and R5 are as defined above in 1,4-dioxane in the presence of Cul/En and K3PO4. The reaction is conducted at about 11O0C for 24 hours. This procedure is described by Kang S-K et al in Synlett, (3), 427-430, 2002.
A fused pyrimidine of formula (I) may be converted into a pharmaceutically acceptable salt, and a salts may be converted into the free compound, by conventional methods. Examples of pharmaceutically acceptable salts include acid addition salts with inorganic acids such as hydrochloric acid, hydrobromic acid, hydroiodic acid, sulphuric acid, nitric acid and phosphoric acid; and organic acids such as formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, methanesulfonic acid, ethanesulfonic acid, aspartic acid and glutamic acid. In the case of compounds of the invention bearing a free carboxy substituent, the salts include the salts of alkali and alkaline earth metals and ammonium, for instance the salts of sodium, potassium, magnesium, calcium and ammonium. The latter are prepared by treating the free fused pyrimidine of formula (I), or an acid addition salt thereof, with the corresponding metal base or ammonia. The compounds of formula (I) and their salts may exist as hydrates or solvates.
Compound of the present invention have been found in biological tests to be inhibitors of PB kinase. The compounds are selective for class Ia PB kinases over class Ib and typically exhibit at least a 20-fold selectivity for class Ia over class Ib PB kinases. In particular, the compounds are selective for the pi 10a isoform. A compound of the present invention may thus be used as an inhibitor of PI3 kinase, in particular of a class Ia PB kinase. Accordingly, a compound of the present invention can be used to treat a disease or disorder arising from abnormal cell growth, function or behaviour associated with PB kinase. Examples of such diseases and disorders are discussed by Drees et al in Expert Opin. Ther. Patents (2004) 14(5):703 - 732. These include cancer, immune disorders, cardiovascular disease, viral infection, inflammation, metabolism/endocrine disorders and neurological disorders. Exampes of metabolism/endocrine disorders include diabetes and obesity. Examples of cancers which the present compounds can be used to treat include leukaemia, brain tumours, renal cancer, gastric cancer and cancer of the skin, bladder, breast, uterus, lung, colon, prostate, ovary and pancreas.
A human or animal patient suffering from an immune disorder, cancer, cardiovascular disease, viral infection, inflammation, a metabolism/endocrine disorder or a neurological disorders may thus be treated by a method comprising the administration thereto of a compound of the present invention as defined above. The condition of the patient may thereby be improved or ameliorated. In addition to possessing biochemical potency the compounds of the invention exhibit physicochemical and pharmacokinetic properties which make them particularly well adapted for drug use. This is shown for instance in the results of the biological assays described in Example 11, which follows. In particular the compounds possess high aqueous solubility at physiological pH; many have a solubility of at least 40 μM and a significant number have a solubility of greater than 100 μM. High solubility at physiological pH is desirable since it promotes bioavailability. The compounds also possess high metabolic stability, as shown in particular by the hepatocyte clearance assay described in Example 11 in which most of the tested compounds were shown to have low hepatocyte clearance. Low hepatocyte clearance correlates with a low rate of liver metabolism. It can therefore be seen that the compounds of the present invention possess improved physicochemical and pharmacokinetic properties whilst retaining biochemical potency as inhibitors of PI3 kinase. A compound of the present invention can be administered in a variety of dosage forms, for example orally such as in the form of tablets, capsules, sugar- or film-coated tablets, liquid solutions or suspensions or parenterally, for example intramuscularly, intravenously or subcutaneously. The compound may therefore be given by injection or infusion. The dosage depends on a variety of factors including the age, weight and condition of the patient and the route of administration. Daily dosages can vary within wide limits and will be adjusted to the individual requirements in each particular case. Typically, however, the dosage adopted for each route of administration when a compound is administered alone to adult humans is 0.0001 to 50 mg/kg, most commonly in the range of 0.001 to 10 mg/kg, body weight, for instance 0.01 to 1 mg/kg. Such a dosage may be given, for example, from 1 to 5 times daily. For intravenous injection a suitable daily dose is from 0.0001 to 1 mg/kg body weight, preferably from 0.0001 to 0.1 mg/kg body weight. A daily dosage can be administered as a single dosage or according to a divided dose schedule. A compound is formulated for use as a pharmaceutical or veterinary composition also comprising a pharmaceutically or veterinarily acceptable carrier or diluent. The compositions are typically prepared following conventional methods and are administered in a pharmaceutically or veterinarily suitable form. The compound may be administered in any conventional form, for instance as follows:
A) Orally, for example, as tablets, coated tablets, dragees, troches, lozenges, aqueous or oily suspensions, liquid solutions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or elixirs. Compositions intended for oral use may be prepared according to any method known in the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents selected from the group consisting of sweetening agents, flavouring agents, colouring agents and preserving agents in order to provide pharmaceutically elegant and palatable preparations.
Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets. These excipients may be for example, inert diluents, such as calcium carbonate, sodium carbonate, lactose, dextrose, saccharose, cellulose, corn starch, potato starch, calcium phosphate or sodium phosphate; granulating and disintegrating agents, for example, maize starch, alginic acid, alginates or sodium starch glycolate; binding agents, for example starch, gelatin or acacia; lubricating agents, for example silica, magnesium or calcium stearate, stearic acid or talc; effervescing mixtures; dyestuffs, sweeteners, wetting agents such as lecithin, polysorbates or lauryl sulphate. The tablets may be uncoated or they may be coated by known techniques to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate may be employed. Such preparations may be manufactured in a known manner, for example by means of mixing, granulating, tableting, sugar coating or film coating processes. Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is present as such, or mixed with water or an oil medium, for example, peanut oil, liquid paraffin, or olive oil.
Aqueous suspensions contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients are suspending agents, for example, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethyl-cellulose, sodium alginate, polyvinylpyrrolidone gum tragacanth and gum acacia; dispersing or wetting agents may be naturally-occurring phosphatides, for example lecithin, or condensation products of an alkylene oxide with fatty acids, for example polyoxyethylene stearate, or condensation products of ethylene oxide with long chain aliphatic alcohols, for example heptadecaethyleneoxycetanol, or condensation products of ethylene oxide with partial esters derived from fatty acids and a hexitol such as polyoxyethylene sorbitol monooleate, or condensation products of ethylene oxide with partial esters derived from fatty acids and hexitol anhydrides for example polyoxyethylene sorbitan monooleate.
The said aqueous suspensions may also contain one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate, one or more colouring agents, such as sucrose or saccharin.
Oily suspension may be formulated by suspending the active ingredient in a vegetable oil, for example arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol.
Sweetening agents, such as those set forth above, and flavouring agents may be added to provide a palatable oral preparation. These compositions may be preserved by this addition of an antioxidant such as ascorbic acid. Dispersible powders and granules suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those already mentioned above. Additional excipients, for example sweetening, flavouring and colouring agents, may also be present.
The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, for example olive oil or arachis oils, or a mineral oil, for example liquid paraffin or mixtures of these. Suitable emulsifying agents may be naturally-occurring gums, for example gum acacia or gum tragacanth, naturally occuring phosphatides, for example soy bean lecithin, and esters or partial esters derived from fatty acids an hexitol anhydrides, for example sorbitan mono-oleate, and condensation products of the said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsion may also contain sweetening and flavouring agents. Syrups and elixirs may be formulated with sweetening agents, for example glycerol, sorbitol or sucrose. In particular a syrup for diabetic patients can contain as carriers only products, for example sorbitol, which do not metabolise to glucose or which only metabolise a very small amount to glucose.
Such formulations may also contain a demulcent, a preservative and flavouring and coloring agents;
B) Parenterally, either subcutaneously, or intravenously, or intramuscularly, or intrasternally, or by infusion techniques, in the form of sterile injectable aqueous or oleaginous suspensions. This suspension may be formulated according to the known art using those suitable dispersing of wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non- toxic paternally- acceptable diluent or solvent, for example as a solution in 1,3-butane diol.
Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution, hi addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglycerides. In addition fatty acids such as oleic acid find use in the preparation of injectables; C) By inhalation, in the form of aerosols or solutions for nebulizers;
D) Rectally, in the form of suppositories prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperature but liquid at the rectal temperature and will therefore melt in the rectum to release the drug. Such materials are cocoa butter and poly-ethylene glycols; E) Topically, in the form of creams, ointments, jellies, collyriums, solutions or suspesions.
The invention will be further described in the Examples which follow:
Example 1: 3-[6-(4-Methyl-piperazin-l-ylmethv0-4-morpholin-4-yl- thieno[3,2-dlpyrimidin-2-yll-phenoI (9)
The synthetic route to compound 9 is shown in the following scheme.
S
Figure imgf000034_0001
Figure imgf000035_0001
Preparation of 3-(3-Methoxy-benzoylamino)-thioρhene-2-carboxylic acid methyl ester (1)
To a solution of methyl-3-amino-2-thiophenecarboxylate (8.0Og, 50.9mmol) in acetonitrile (10OmL) was added potassium carbonate (l.leq., 56.0mmol, 7.73g) followed by w-anisoyl chloride (1.05eq., 53.4mmol, 7.5ImL) and the mixture heated under reflux for 1 hour. A white precipitate forms. The mixture was cooled in ice- water, diluted with water (10OmL) and the white solid collected by filtration and dried to yield compound 1 (12.78 g). Upon standing, an additional quantity of product precipitated from the water-acetonitrile solution; this was collected in the same fashion. (Total yield: 14.08 g, 95%).
1H NMR (400 MHz, CDCl3) 3.89 (s, 3H), 3.93 (s, 3H), 7.12 (dd, IH, J=2.2, 8.4), 7.42 (t, 1H, J=7.9), 7.54 (d,lH, J=5.5), 7.58 (m, IH), 8.29 (d,lH, J=5.5), 11.17 (brs, IH).
Preparation of 2-(3-Methoxy-phenyl')-3H-thieno|~3,2-d1pyrimidin-4-one ( 2)
Ammoniacal methanol was prepared by bubbling ammonia (30- 32g) through methanol (19OmL) cooled in ice- water. To the solution was added 3-(3-methoxy- benzoylamino)-thiophene-2-carboxylic acid methyl ester (18.00g, 61.8mmol) and the mixture heated at 900C for 44 hours in a steel bomb at 50psi. The solvent was then removed in vacuo and isopropanol (30OmL) added followed by 2M aqueous sodium hydroxide (4.0eq., 124mL). The solution was heated under reflux for 15 hours and then cooled in ice-water. The mixture was acidified to pH 1 by the addition of 4 M hydrochloric acid and the white precipitate collected by filtration, washed with water and dried to give compound 2 (15.20 g, 95%). 1H NMR (400 MHz, J6-DMSO) 3.86 (s, 3H), 7.14 (dd, IH, J=2.2, 8.2), 7.45 (m, 2H), 7.70 (s, IH), 7.74 (d, IH, J=7.8), 8.22 (d, IH, J=5.2), 12.70 (brs, IH),
Preparation of 2-(3-Hvdroxy-phenyl)-3H-thieno[3,2-dlpyrimidin-4-one (3)
To a suspension of 2-(3-methoxy-phenyl)-3H-thieno[3,2-d]pyrimidin-4-one (30.31g, 0.12 mol) in acetic acid (17OmL) was added 48% aqueous hydrogen bromide (17OmL) and the mixture heated under reflux for 48 hours. The mixture was then cooled in ice-water and diluted with water (20OmL). The grey solid was collected by filtration, washed with water and dried to give compound 3 (28.58 g, 100%). 1H NMR (400 MHz, J6-DMSO) 6.87 (m, IH), 7.23 (t, IH, J=8.0), 7.36 (d, IH, J=5.2), 7.45 (m, 2H), 8.13 (d, IH, J=5.2), 9.67 (brs, IH), 12.52 (brs, IH).
Preparation of Acetic acid 3-(4-oxo-3,4-dihvdro-thieno[3,2-d1pyrimidin-2-yl)-phenyl ester (4) A mixture of 2-(3-hydroxy-phenyl)-3H-thieno[3,2-d]pyrimidin-4-one
(28.58g, 0.12mol) and sodium acetate (l.leq., 0.13mol, 10.56g) in acetic anhydride (28OmL) was heated under reflux for 1.5 hours. The mixture was then cooled in ice- water and the grey solid collected by filtration, washed with water and dried to yield compound 4 (27.69 g, 82%). 1H NMR (400 MHz, J6-DMSO) 2.32 (s, 3H), 7.36 (dd, IH, J=2.0, 8.0), 7.47 (d, IH, J=5.2), 7.59 (d, IH, J=8.0), 7.93 (s, IH), 8.04 (d, IH, J=7.9), 8.22 (d, IH, J=5.2), 12.75 (brs, IH).
Preparation of 3-(4-Chloro-thienof3,2-d]pyrirnidin-2-yl)-phenol and Acetic acid 3- (4-chloro-thienor3,2-d1pyrimidin-2-yl)-phenyl ester (5) To the pyrimidone (26.65g, 93.1mmol) was added phosphorus oxychloride (13OmL, 15eq.) and the dark mixture heated under reflux for 3 hours. Most of the phosphorus oxychloride was removed by distillation at reduced pressure and the residue transferred to a conical flask and cooled in ice-water. The mixture was quenched by the addition of ice-water and then saturated aqueous sodium hydrogen carbonate added (60OmL). The solid was collected by filtration and re-suspended in aqueous sodium hydrogen carbonate, such that the pH was greater than 7. The solid was collected by filtration, washed thoroughly with water and dried to yield a mixture of the chloropyrimidines 5 in the ratio 1.5 : 1 (24.0 g). 3-(4-Chloro-thieno[3,2-d]pyrimidin-2-yl)-phenol: 1H NMR (400 MHz, J6-DMSO) 6.94 (dd, IH, J=I.6, 7.6). 7.35 (t, IH, J=7.8), 7.76 (d, IH, J=5.4), 7.87 (m, 2H), 8.58 (d, 1H, J=5.4), 9.67 (brs, IH).
Acetic acid 3-(4-chloro-thieno[3,2-d]pyrimidin-2-yl)-phenyl ester: 1H NMR(400 MHz, CDCl3) 2.35 (s, 3H), 7.24 (m, IH), 7.52 (t, IH, J=8.0), 7.61 (d, IH, J=5.4), 8.04 (d, IH, J=5.4), 8.28 (t, IH, J=2.0), 8.43 (dd, IH, J=I.7, 7.9).
Preparation of 3-(4-Mθφholin-4-yl-thieno[3.2-dlpyrimidin-2-yl)-phenol (6)
To a mixture of the chloropyrimidines (24.Og) in n-butanol, (22OmL) was added morpholine (3.5eq., 0.33mol, 28.3mL) and the mixture heated at 1000C for 2 hours. The solvent was removed in vacuo and water added to the residue. The pale brown solid was collected by filtration and dried in a vacuum oven to give compound
6 as a pale brown solid (20 .3 g, 70%).
1H NMR (400 MHz, ^6-DMSO) 3.81 (t, 4H, j=4.8), 4.01 (t, 4H, J=4.8), 6.86 (m,
IH), 7.27 (t, IH, J=8.0), 7.51 (d, IH, j=5.6), 7.86 (m, 2H), 8.25 (d, IH, j=5.6), 9.46 (brs, IH).
Preparation of 2-[3-(tgrt-Butyl-dimethyl-silanyloxy')-phenyl1-4-moφholin-4-yl- thieno[3,2-dlpyrimidine (7)
A mixture of 3-(4-moφholin-4-yl-thieno[3,2-d]pyrimidin-2-yl)-phenol (17.28g, 55.1mmol), imidazole (1.7eq., 93.7mmol, 6.38g) and tert-butyldimethylsilyl chloride (1.5eq., 82.7mmol. 12.47g) in DMF (45mL) was heated at 500C for 5 hours. The reaction mixture was then cooled and partitioned between water (40OmL) and ether (3 * 40OmL). The combined organic layers were washed sequentially with saturated aqueous sodium hydrogen carbonate (30OmL), brine (30OmL), separated and dried (MgSO4). The crude product was evaporated onto silica and purified by chromatography (5%→20% ethyl acetate-petrol) to yield compound 7 as a white solid (19.6O g, 83%).
1H NMR (400 MHz, CDCl3) 0.15 (s, 6H), 0.92 (s, 9H), 3.80 (t, 4H, J=4.8), 3.98 (t, 4H, J=4.8), 6.83 (td, IH, J=I.5, 8.3), 7.22 (t, IH, J=7.9), 7.41 (d, IH, J=5.5), 7.64 (d, IH, J-5.5), 7.84 (t, IH, J=2.0), 7.95 (d, IH, J=7.8).
Preparation of 4-Moφholin-4-yl-2-[3-(l , 1 ,2,2-tetramethyl-propylsilanyloxy)- phenyl]-thieno[3,2-d1pyrimidine-6-carbaldehyde (8)
To a solution of 4-morpholin-4-yl-2-[3-(l,l,2,2-tetramethyl- propylsilanyloxy)-phenyl]-thieno[3,2-d]pyrimidine (2.26g, 5.29mmol) in dry THF (4OmL) cooled to -78 0C was added nBuLi (2.5 M solution in hexanes, 2.75mL, 1.3eq.). After stirring for 20 minutes, dry N,7V-dimethylformamide (617μL, 1.5eq.) was added, and the reaction mixture was stirred for 20 minutes at -78 0C and then warmed slowly to room temperature. After a further 30 minutes at room temperature the reaction mixture was quenched with ice/brine and then extracted exhaustively with chloroform. The combined organic extracts were dried (MgSO4) and the solvent removed in vacuo to yield compound 8 as a yellow solid (2.38g, 99%). Preparation of 3-[6-(4-Methyl-piperazin-l-ylmethyl)-4-moφholin-4-yl-thienor3,2- dlpyrimidin-2-yll-phenol (9) A mixture of 4-moφholin-4-yl-2-[3-(l,l,2,2-tetramethyl-propylsilanyloxy)- phenyl]-thieno[3,2-d]pyrimidine-6-carbaldehyde (6.67g, 14.66mmol), N- methylpiperazine (2.1 ImL, 1.3 equivalents) and acetic acid (838μL, l.Oeq.) was stirred in 1,2-dichloroethane (7OmL) at room temperature. To this was added sodium triacetoxyborohydride (3.42 g, 1.1 equivalents) and the reaction mixture was stirred for 3 days. The reaction mixture was then quenched with aqueous sodium bicarbonate solution, extracted exhaustively with chloroform, dried (MgSO4), and the solvent removed in vacuo to yield a yellow oil. This was purified using flash chromatography (silica, ethyl acetate to ethyl acetate/methanol) to yield 6-(4-methyl- piperazin- 1 -ylmethyl)-4-morpholin-4-yl-2-[3-(l , 1 ,2,2-tetramethyl-propylsilanyloxy)- phenyl]-thieno[3,2-d]ρyrimidine (6.95 g, 88%).
To a solution of 6-(4-methyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-2-[3- (l,l,2,2-tetramethyl-propylsilanyloxy)-phenyl]-thieno[3,2-d]pyrimidine (6.95g, 12.89mmol) in THF (10OmL) cooled to 0 0C was added a 1.0 M solution of tetrabutyl ammonium fluoride in THF (14.2mL, l.leq.). After 30 minutes the solvent was removed in vacuo and the residue was purified using flash chromatography (silica, 8% methanol in dichloromethane) and then triturated using ethyl acetate/methanol to yield the title compound as a white solid (4.62g, 85%).
1H NMR (400 MHz, CDCl3) 2.27 (s, 3H, CH3), 2.49 (br s, 8H, 4 x CH2), 3.79 (s, 2H, CH2), 3.82-3.84 (m, 4H, 2 x CH2), 4.0-4.03 (m, 4H, 2 x CH2), 6.88-6.91 (m, H, ArH), 7.24-7.28 (m, 2H, 2 x ArH part under CDCl3), 7.69-7.70 (m, H, ArH), 7.81 (d, H, ArH, J=7.8Hz); MS (ESI+) 426(MH+).
Example 2 Further Compounds of the Invention
The following compounds of the invention were prepared by analogy with the procedure described in Example 1, using the appropriate amine. The amines are commercially available compounds unless stated otherwise. Characterising data are also given for the compounds of the invention.
10: 3-(4-Morpholin-4-yl-6-morphoIin-4-yImethyl-thieno[3,2-d]pyrimidin-2- yl)-phenol was prepared using morpholine.
1H NMR (400 MHz, CDCl3) 2.55 (m, 4H), 3.75-3.79 (m, 4H), 3.82 (s, 2H), 3.93- 3.96 (m, 4H), 4.04-4.00(m, 4H), 6.95 (d, 2H), 7.36-7.30 (m, 2H), 7.99-7.91 (m, 2H); MS (ESI+) 413 (MH+). 11 : 3-(4-Morpholin-4-yl-6-pyrrolidin-l-ylmethyl-thieno[3,2-d]pyrimidin-2- yl)-phenol was prepared using pyrrolidine.
1H NMR (400 MHz, CDCl3) 1.87-1.90 (m, 4H), 2.65-2.70 (m, 4H), 3.82-3.88 (m, 4H), 3.99 -4.03 (m, 6H), 6.95 (d, 2H), 7.30-7.36 (m, 2H), 7.80 (s, IH), 7.99 (d, IH); MS (ESI+) 397 (MH+).
12: 3-[6-(6,7-Dimethoxy-3,4-dihydro-lH-isoquinolin-2-ylmethyl)-4- morpholin-4-yl-thieno[3,2-d]pyrimidin-2-yI]-phenol was prepared using 6,7- dimethoxy-l,2,3,4-tetrahydroisoquinoline. 1H NMR (400 MHz, CDCl3) 2.86-2.88 (m, 4H), 3.70 (s, 2H), 3.83 (s, 3H), 3.85-3.89 (m, 7H), 3.97-4.02 (m, 6H), 5.11 (br s, IH), 6.51 (s, IH), 6.60 (s, IH), 6.95 (d, IH), 7.35-7.30 (m, 2H), 7.92 (s, IH), 8.01 (d, IH); MS (ESl+) 519 (MH+).
13: 3-(6-[l,4']Bipiperidinyl-l'-ylmethyl-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl)-phenoI was prepared using 4-piperidinopiperidine.
1H NMR (400 MHz, CDCl3) 1.42-1.48 (m, 2H), 1.54-1.68 (m, 6H), 1.82-1.84 (m, 2H), 2.01-2.05 (m, 2H), 2.28-2.30 (m, IH), 2.48-2.52 (m, 4H), 2.97-2.99 (m, 2H), 3.79 (s, 2H), 3.85-3.89 (m, 4H), 3.99-4.02 (m, 4H), 6.90 (d, IH), 7.29-7.38 (m, 2H), 7.90 (s, IH), 7.99 (d, IH); MS (ESI+) 494 (MH+).
14: 3- [4-Morpholin-4-yl-6-(4-pyrimidin-2-yl-piperazin-l -ylmethyl)- thieno|3,2-d]pyrimidin-2-yl]-phenol was prepared usingl-(2-pyrimidyl)piperazine. 1H NMR (400 MHz, CDCl3) 2.58-2.62 (m, 4H) 3.85-3.89 (m, 10H), 3.99-4.02 (m, 4H), 5.90 (br s, IH), 6.48 (t, IH), 6.90 (d, IH), 7.29-7.38 (m, 2H), 7.90 (s, IH), 7.99 (d, IH), 8.30 (d, 2H); MS (ESI+) 490 (MH+).
15: 3-[6-(4-CyclohexyImethyl-piperazin-l-ylmethyl)-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-2-yl]-phenoI was prepared using 1- (cyclohexylmethyl)piperazine. 1H NMR (400 MHz, CDCl3) 0.83-0.95 (2H, m), 1.17-1.26 (3H, m), 1.42-1.50 (IH, m), 1.68-1.77 (5H, m), 2.14 (2H, d, J=7.1Hz), 2.40-2.50 (4H, br m), 2.52-2.64 (4H, broad), 3.81 (2H, s), 3.87 (4H, t, J=4.6Hz), 4.03 (4H, t, J=4.6Hz), 5.23-5.28 (IH, broad, OH), 6.92-6.94 (IH, m), 7.31 (IH, s), 7.33 (IH, t, J=7.9Hz), 7.91 (IH, s), 8.00 (IH, d, J=7.8); MS (ESI+) 508.2 (MH+).
16: 3-[6-(4-IsopropyI-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol was prepared using 1-isopropylpiperazine. 1H NMR (400 MHz, J6-DMSO) 0.99 (d, 6H), 2.65 (septet, IH) 3.82-3.88 (m, 6H), 3.94-3.99 (m, 4H), 6.90 (d, IH), 7.27-7.30 (m, IH), 7.40 (s, IH), 7.84-7.88 (m, 2H), 9.55 (lH,s), 8H hidden under DMSO peak @ 2.52; MS (ESf) 454 (MH+).
17: 4- [2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno [3,2-d] pyrimidin-6- ylmethyl]-piperazine-l-carboxylic acid tert-butyl ester was prepared using 1- BOC-piperazine.
1H NMR (400 MHz, CDCl3) 1.48 (s, 9H) 2.48-2.52 (m, 4H), 3.48-3.51 (m, 4H), 3.79 (s, 2H), 3.85-3.89 (m, 4H), 3.99-4.02 (m, 4H) 6.90 (d, IH), 7.29-7.38 (m, 2H), 7.76- 7.99 (m, 2H); MS (ESI+) 512 (MH+).
19: 3-{6-[4-(2-Methoxy-ethyl)-piperazin-l-ylmethyl]-4-morpholin-4-yI- thieno[3,2-d]pyrimidin-2-yI}-phenol was prepared using l-(2- methoxyethyl)piperazine.
1H NMR (400 MHz, CDCl3) 2.58-2.68 (m, 10H), 3.35 (s, 3H), 3.52 (t, 2H, J=5.6Hz), 3.82 (s, 2H), 3.87 (t, 4H, J=4.6Hz), 4.03 (t, 4H, J=4.6Hz), 5.60-5.68 (brs, IH), 6.92- 6.94 (m, IH), 7.31 (s, IH), 7.33 (t, IH, J=7.9), 7.91 (s, IH), 8.00 (d, IH, J=7.8Hz). MS (ESl+) 470.1 (MH+).
20: 3-[6-(4-Methyl-[l,4]diazepan-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol was prepared using 1-methylhomopiperazine. 1H NMR (400 MHz, CDCl3) 1.85-1.89 (m, 2H), 2.40 (s, 3H), 2.65-2.74 (m, 4H), 2.82-2.86 (m, 4H), 3.88 (t, 4H, J=4.6Hz), 3.95 (s, 2H), 4.04 (t, 4H, J=4.6Hz), 5.90- 5.98 (br s, IH), 6.92-6.94 (m, IH), 7.31 (s, IH), 7.33 (t, lH, J=7.9Hz), 7.91 (s, IH), 8.00 (d, IH, J=7.8Hz); MS (ESI+) 440.1 (MH+).
21 : {4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidiii-6- ylmethyl]-piperazin-l-yl}-acetic acid ethyl ester was prepared from 1- ethoxycarbonylmethyl)piperazine.
1H NMR (400 MHz, CDCl3) 1.27 (t, 3H, J=7.1Hz), 2.60-2.69 (br, 8H), 3.23 (s, 2H), 3.83 (s, 2H), 3.89 (t, 4H, J=4.6Hz), 4.03 (t, 4H, J=4.6Hz), 4.20 (q, 2H, J=4.6Hz),
5.12 (brs, IH), 6.92-6.94 (m, IH), 7.31 (s, IH), 7.33 (t, 1H, J=7.9), 7.91 (s, IH), 8.00 (d, IH, J=7.8Hz); MS (ESI+) 498.1 (MH+)
22: l-{4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yI-thieno[3,2-d]pyrimidin-6- ylmethyl]-piperazin-l-yl}-ethanone was prepared from 1 -acetylpiperazine.
1H NMR (400 MHz, CDCl3) 2.09 (s, 3H, CH3), 2.52-2.55 (m, 4H, 2 x CH2), 3.49- 3.51 (m, 2H, CH2), 3.66-3.68 (m, 2H, CH2), 3.83 (s, 2H, CH2), 3.86-3.90 (m, 4H, 2 x CH2), 4.03-4.06 (m, 4H, 2 x CH2), 5.88 (br s, H, OH), 6.94 (d, H, ArH, J=7.66Hz), 7.31-7.35 (m, 2H, 2 x ArH), 7.93 (s, H, ArH), 7.98 (d, H, ArH, J=7.77Hz); MS (ESI+) 454.0 (MH+).
23: 3-{6-[4-(2-Hydroxy-ethyl)-piperazin-l-yImethyl]-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-2-yl}-phenol was prepared from N-(2- hydroxyethyl)piperazine. 1H NMR (400 MHz, CDCl3) 2.63 (br m, 1OH, 2 x CH2), 3.65 (m, 2H, CH2), 3.84 (s, 2H, CH2), 3.87-3.90 (m, 4H, 2 x CH2), 4.04 -4.06 (m, 4H, 2 x CH2), 6.93 (d, H, ArH, J=ICOHz), 7.32-7.36 (m, 2H, 2 x ArH), 7.91 (s, H, ArH), 8.01 (d, H, ArH, J=7.74Hz); MS (ESf) 456.1 (MH+). 24: 3-(4-Morpholin-4-yl-6-thiomorpholin-4-ylmethyl-thieno[3,2-d]pyrimidin-
2-yl)-phenol was prepared from thiomorpholine.
1H NMR (400 MHz, CDCl3) 2.70-2.72 (m, 4H, 2 x CH2), 2.80-2.82 (m, 4H, 2 x CH2), 3.83 (s, 2H, CH2), 3.87-3.90 (m, 4H, 2 x CH2), 5.77 (sbr, H, OH), 6.93 (d, H, ArH, J=7.84Hz), 7.30-7.35 (m, 2H, 2 x ArH), 7.93 (s, H, ArH), 7.98 (d, H, ArH, J=7.84Hz); MS (ESf) 429.2 (MH+).
25: 3-[6-(4-Ethyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidiii-2-yl]-phenol was prepared from 1-ethylpiperazine. 1H NMR (400 MHz, CDCl3) 1.11 (t, 3H, J=7.1Hz), 2.45-2.49 (br m, 2H), 2.49-2.72 (br, 8H), 3.83 (s, 2H), 3.88 (t, 4H, J=4.6Hz), 4.03 (t, 4H, J=4.6Hz), 6.24-6.32 (br, IH), 6.91-6.94 (m, IH), 7.28 (s, IH), 7.33 (t, IH, J=7.9Hz), 7.94 (s, IH), 7.98 (d, IH, J=7.8Hz); MS (ESf) 440.2 (MH+).
26: 3-(6-{[Methyl-(l-methyl-piperidin-4-yl)-amino]-methyl}-4-morpholin-4- yl-thieno[3,2-d]pyrimidin-2-yl)-phenol was prepared from l-methyl-4- (methylamino)piperidine.
1H NMR (400 MHz, CDC13) 1.80-1.88 (3H, br m), 1.97-2.08 (IH, br m), 2.17 (3H, s), 2.33 (3H, s), 2.33-2.39 (2H, br m), 2.48-2.56 (IH, br m), 2.99-3.10 (2H, br m), 3.83 (2H, s), 3.86-3.89 (4H, m), 4.02-4.04 (4H, m), 6.93 (IH, m), 7.21 (IH, s), 7.32 (IH, t, J=I.9), 7.93 (IH, s), 8.00 (IH, d, J=7.8); MS (ESl+) 454.1 (MH+).
27: 3-[6-(4-Dimethylamino-piperidin-l-ylmethyl)-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-2-yl]-phenoI was prepared from 4- dimethylaminopiperidine.
1H NMR (400 MHz, CDCl3) 1.58-1.68 (br m, 2H), 1.87-1.95 (br m, 2H), 2.10 (br t, 2H, J=10.7Hz), 2.34 (br m, IH), 2.37 (br s, 6H), 3.02 (br m, 2H), 3.80 (s, 2H), 3.87 (t, 4H, J=4.7Hz), 4.03 (t, 4H, J=4.9Hz), 6.92-6.94 (m, IH), 7.31 (s,lH), 7.33 (t, IH, J=7.9Hz), 7.91 (s, IH), 8.00 (d, lH, J=7.8Hz); MS (ESl+) 454.1 (MH+). 28: 4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyl]-piperazin-2-one was prepared from piperazin-2-one. 1H NMR (400 MHz, CDCl3) 2.77 (t, 2H, J=5.3Hz), 3.39 (2H, s), 3.42 (br t, 2H), 3.89 (t, 4H, J=5.0Hz), 3.92 (s, 2H), 4.05 (t, 4H, J=5.0Hz), 5.48 (br s, IH), 5.86 (bs,lH), 6.92-6.94 (m, IH), 7.31 (s, IH), 7.33 (t, IH, J=7.9Hz), 7.91 (s, IH), 8.00 (d, IH, J=7.8Hz); MS (ESI+) 426.1 (MH+).
29: 3-[6-(4-Methanesulfonyl-piperazin-l-yImethyl)-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-2-yl]-phenol was prepared from 1- methanesulfonylpiperazine hydrochloride. This in turn was prepared by reaction of
N-BOC-piperazine with methane sulfonyl chloride in dichloromethane, followed by cleavage of the BOC protecting group using HCl (2M) in dichloromethane.
1H NMR (400 MHz, CDCl3) 2.59 (m, 4H, 2 x CH2), 2.72 (s, 3H, CH3), 3.20 (m, 4H,
2 x CH2), 3.80 (m, 6H, 3 x CH2), 3.94-3.97 (m, 4H, 2 x CH2), 6.87 (d, H, ArH, J=7.76Hz), 7.19-7.27 (m, 2H, 2 x ArH), 7.88 (m, 2H, 2 x ArH); MS (ESl+) 490.0
(MH+).
30: {4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyl]-piperazin-l-yl}-acetonitrile was prepared from piperazin-1-yl- acetonitrile hydrochloride. This in turn was prepared from prepared by reaction of N-
BOC-piperazine with bromoacetonitrile in MeCN, followed by cleavage of the BOC protecting group using HCl (2M) in dichloromethane.
1H NMR (400 MHz, CDCl3 ) 2.64-2.68 (m, 8H, 4 x CH2), 3.56 (s, 2H, CH2), 3.84 (s,
2H, CH2), 3.90-3.92 (m, 4H, 2 x CH2), 4.05-4.08 (m, 4H, 2 x CH2), 6.93-6.95 (m, H, ArH), 7.32-7.36 (m, 2H, 2 x ArH), 7.94 (s, H, ArH), 8.0 (d, H, ArH, J=7.8Hz). MS
(ESI+) 451.1 (MH+).
31 : 3-[4-Morpholin-4-yl-6-(4-morpholin-4-yl-piperidin-l-yImethyl)- thieno[3,2-d]pyrimidin-2-yl]-phenol was prepared from 4-morpholinopiperidine. 1H NMR (400 MHz, CDCl3 )1.56-1.65 (m, 2H, CH2), 1.83-1.86 (m, 2H, CH2), 2.08- 2.14 (m, 2H, CH2), 2.19-2.24 (m, H, CH), 2.55 (m, 4H, 2 x CH2), 3.02-3.05 (m, 2H, CH2), 3.72-3.74 (m, 4H, 2 x CH2), 3.79 (s, 2H, CH2), 3.85-3.88 (m, 4H, 2 x CH2), 4.0-4.03 (m, 4H, 2 x CH2), 6.52 (br s, H, OH), 6.91 (m, H, ArH), 7.29-7.34 (m, 2H, 2 x ArH), 7.91 (s, H, ArH), 7.98 (d, H, ArH, J=7.8Hz); MS (ESf) 496.2 (MH+).
33: 3-[6-(3-DimethyIamino-pyrrolidin-l-ylmethyl)-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-2-yl]-phenol was prepared from 3-(dimethylamino) pyrrolidine. 1H NMR (400 MHz, J6-DMSO) 9.52 (s,lH), 7.85 (m, 2H), 7.37 (s, IH), 7.28 (t, IH, J=7.6Hz), 6.87 (d, IH), 3.96 (m, 6H), 3.82 (m, 4H), 2.89 (m, 2H), 2.79 (m, IH), 2.68 (m, IH), 2.24 (s, 6H), 1.95 (m, IH), 1.74 (m, IH); MS (ESI+) 440.1 (MH+).
34: l-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyI]-4-morpholin-4-ylmethyl-piperidin-4-ol was prepared from 4-morpholin- 4-ylmethyl-piperidin-4-ol dihydrochloride. This was prepared as follows: trimethylsulfoxonium iodide (1.Og, 4.54mmol, 1.03eq.,) and sodium hydride (190 mg, 4.75mmol, 1.08eq.,) were stirred in dry DMSO (5mL) under nitrogen. After 30 min. a solution of t-butyl 4-oxo-l-piperidine carboxylate (875mg, 4.39mmol, leq.) in DMSO was added and the resultant heated at 60 0C under argon for 4 hours. The mixture was then cooled, diluted with water and extracted with EtOAc (3 x 3OmL). The combined extracts were dried (Na2SO4) and purified by flash chromatography (silica gel, 20% EtOAc/Hexanes) to give l-oxa-6-aza-spiro[2.5]octane-6-carboxylic acid tert-butyl ester as an off-white solid (661.6 mg, 71%). l-Oxa-6-aza-spiro[2.5]octane-6-carboxylic acid tert-butyl ester (lOOmg,
0.47mmol) and morpholine (42μL, 0.48mmol, 1.03eq) were stirred in EtOH at RT overnight. Solvent was removed in vacuo and the residue purified by flash chromatography (silica gel, 4% MeOH/DCM) to give 4-hydroxy-4-morpholin-4- ylmethyl-piperidine-1-carboxylic acid tert-butyl ester as a white solid (91.4 mg, 67%). To a solution of 4-hydroxy-4-morpholin-4-ylmethyl-piperidine-l-carboxylic acid tert-butyl ester (91.4mg, 0.32mmol) in MeOH (3mL) was added 2 M HCl in ether (480μL, 0.96mmol, 3eq.). The mixture was stirred at RT for 4 days then concentrated in vacuo to give a gum which was used without further purification. 1H NMR (400 MHz, CDCl3) 1.56-1.62 (m, 4H, 2 x CH2, part HOD), 2.35 (s, 2H, CH2), 2.51 (m, 2H, CH2), 2.61-2.64 (m, 4H, 2 x CH2), 2.74 (m, 2H, CH2), 3.12 (br s, H, OH), 3.69-3.71 (m, 4H, 2 x CH2), 3.84 (s, 2H, CH2), 3.87-3.89 (m, 4H, 2 x CH2), 4.02-4.05 (m, 4H, 2 x CH2), 5.2 (br s, H, OH), 6.94 (d, H, ArH, J=7.55Hz), 7.31 (s, H, ArH), 7.35 (t, H, ArH, J=7.9Hz), 7.9 (s, H, ArH), 8.02 (d, H, ArH, J=7.75Hz); MS (ESf) 526.2 (MH++0Me)
35: 4-[2-(3-Hydroxy-phenyl)-4-morphoIin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyl]-piperazine-l-carboxylic acid ethylamide was prepared from piperazine- 1-carboxylic acid ethylamide. This in turn was prepared by reaction of N-BOC- piperazine with ethyl isocyanate in dichloromethane, followed by cleavage of the BOC protecting group using HCl (2M solution in ether) in dichloromethane. 1H NMR (400 MHz, CDCl3) 1.14 (t, 3H, J=7.3Hz), 2.55 (t, 4H, J=5.0Hz), 3.25-3.32 (m, 2H), 3.41 (t, 4H, J=5.0Hz), 3.84 (s, 2H), 3.89 (t, 4H, J=4.Hz), 4.05 (t, 4H, J=4.8Hz), 4.35 (t, IH), 5.22-5.28 (br m, IH), 6.92-6.94 (m, IH), 7.31 (s, IH), 7.33 (t, IH, J=7.9Hz), 7.91 (s, IH), 8.00 (d, IH, J=7.8Hz); MS (ESI+) 483.1 (MH+).
39: 4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyl]-piperazine-l-carboxylic acid dimethylamide was prepared from piperazine- 1-carboxylic acid dimethylamide. 1H NMR (400 MHz, J6-DMSO) 9.50 (IH, br s), 7.85-7.80 (2H, m), 7.38 (IH, s), 7.30-7.27 (IH, m), 6.85 (IH, d), 3.99-3.96 (4H, m), 3.90 (2H, s), 3.81-3.78 (4H, m), 3.17-3.15 (4H, m), 2.72 (6H, s), 2.55-2.52 (4H, m); MS (ESI+) 483 (MH+).
40: l-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyl]-piperidin-4-ol was prepared from 4-hydroxypiperidine. 1H NMR (400 MHz, J6-DMSO) 1.40-1.47 (m, 2H), 1.72-1.75 (m, 2H), 2.16-2.21 (m, 2H), 2.75-2.78 (m, 2H), 3.48-3.50 (m, IH), 3.79-3.82 (m, 6H), 3.96-3.98 (m, 4H), 4.57 (d, IH, J=4.1Hz), 6.85 (dd, IH), 7.26 (t, IH, J=8.1Hz), 7.35 (s, IH), 7.83-7.85 (m, 2H), 9.48 (s, IH); MS (ESf) 427.1 (MH+).
41: 3-[6-(l,l-Dioxo-thiomorpholin-4-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol was prepared from thiomorpholine 1,1 -dioxide, which in turn was prepared according to the literature (Journal of Medicinal Chemistry, 1994, 37, 913-923). 1H NMR (400 MHz, J6-DMSO) 9.50 (br s, IH), 7.85-7.80 (m, 2H), 7.40 (lH,s), 7.30-7.27 (m, IH), 6.85 (d, IH), 4.08 (s, 2H), 4.00-3.95 (m, 4H),3.81-3.78 (m, 4H), 3.23-3.20 (m, 4H), 3.02-2.98 (m, 4H); MS (ESI+) 461 (MH+).
42: 4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyl]-piperazine-l-sulfonic acid dimethylamide was prepared using piperazine-1 -sulfonic acid dimethylamide.
1H NMR (400 MHz, J6-DMSO) 9.50 (IH, br s), 7.85-7.80 (2H, m), 7.40 (IH, s), 7.30-7.27 (IH, m), 6.85 (lH,d), 4.02-3.98 (4H, m), 3.95 (2H, s), 3.81-3.78 (4H, m), 3.23-3.20 (4H, m), 2.75 (6H, s), 2.55-2.52 (4H, m); MS (ESI+) 519.4 (MH+).
43: 3-(6-{4-[(2-Methoxy-ethyl)-methyl-amino]-piperidin-l-ylmethyl}-4- morpholin-4-yl-thieno[3,2-d]pyrimidin-2-yl)-phenol was prepared from (2- methoxy-ethyl)-methyl-piperidin-4-yl-amide. This in turn was prepared from tert- butyl-4-oxo-l-piperidine carboxylate and N-(2-methoxyethyl)methylamine via reductive amination, followed by cleavage of the BOC protecting group using HCl (2M solution in ether) in dichloromethane.
1H NMR (400 MHz, J6-DMSO) 1.40-1.49 (m, 2H), 1.64-1.67 (m, 2H), 2.02-2.07 (m, 2H), 2.20 (s, 3H), 2.29-2.35 (m, IH), 2.56 (t, 2H, J=6.2Hz), 2.93-2.96 (m, 2H), 3.23 (s, 3H), 3.36 (t, 2H, J=6.2Hz), 3.79-3.82 (m, 6H), 3.96-3.98 (m, 4H), 6.85 (dd, IH), 7.26 (t, lH, J=8.1Hz), 7.35 (s, IH), 7.83-7.85 (m, 2H), 9.48 (s, IH); MS (ESI+) 498.2 (MH+)
44: l-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidiii-6- ylmethyl]-pyrrolidin-3-oI was prepared from 3-pyrrolidinol.
1H NMR (400 MHz, J6-DMSO) 1.59 (m, IH), 2.00 (m, IH), 2.45 (m, IH), 2.50 (m, IH), 2.69 (m, IH), 2.84 (m, IH), 3.80 (m, 4H), 3.96 (m, 6H), 4.24 (s, IH), 4.74 (br s, IH), 6.85 (m, IH), 7.26 (t, IH, J=7.9Hz), 7.34 (s, IH), 7.83 (br s,2H), 9.45 (s, IH); MS (ESf) 413.1 (MH+).
45: (R,S)-3-[6-(2-Dimethylaminomethyl-pyrrolidin-l-ylmethyl)-4-morpholin- 4-yl-thieno[3,2-d]pyrimidin-2-yl]-phenol was prepared from (R, 5)-Dimethyl-(l- methyl-pyrrolidin-2-ylmethyl)-amine, which was prepared by standard methods (J. Med. Chem. 2002, 45, 721). 1H NMR (400 MHz, CDCl3) 1.64-1.78 (m, 3H), 2.01 (m, IH), 2.28 (s, 6H, & m, IH), 2.34 (dd, IH, J=12.3Hz, 7.3Hz), 2.53 (dd, IH, J=12.3Hz, 4.7Hz), 3.10 (m, IH), 2.70 (m, IH), 3.71 (d, IH, J=14.6Hz), 3.88 (t, 4H, J=4.8Hz), 4.03 (t, 4H, J=4.8Hz), 4.42 (d, IH, J=14.7), 6.92 (dd, IH, J=7.7Hz, 2.3Hz), 7.26 (s, IH), 7.33 (t, IH, J=7.9Hz), 7.91 (t, IH, J=I.9Hz), 8.00 (d, IH, J=7.8Hz); MS (ESl+) 454.4 (MH+).
46: (R,S)-3-(6-{[Methyl-(l-methyl-pyrrolidin-2-ylmethyl)-amino]-methyl}-4- morphoIin-4-yl-thieno[3,2-d]pyrimidin-2-yl)-phenol was prepared from (R1S)- methyl-(l -methyl -pyrrolidin-2-ylmethyl)-amine prepared in turn as follows: 1- methyl-2-pyrrolecarboxaldehyde (1.Og, 9.16mmol) was added to a solution of methyl amine in methanol (2.3 Ig in 1OmL) and the mixture stirred at room temperature for 3.5 hours. The solution was cooled in ice-water and sodium borohydride added (2.0eq., 18.3mmol, 693mg) and then stirred at room temperature for 17 hours. The solvent was evaporated and the residue partitioned between saturated aqueous sodium hydrogen carbonate (3OmL) and dichloromethane (3 x 3OmL). The combined organic layers were washed with brine, separated and dried (MgSO4). Hydrogenation over platinum oxide (Chem. Pharm. Bull, 1990, 38, 930-935) furnished the desired compound.
1H NMR (400 MHz, CDCl3) 1.59-1.76 (m, 3H), 1.97 (m, IH), 2.17 (dd, IH, J= 17.4Hz, 9.5Hz), 2.23 (s, 3H), 2.34-2.40 (m, 2H), 2.40 (s, 3H), 2.65 (dd, IH, J=I 1.5Hz, 4.1Hz), 3.05 (t, IH, J=7.5Hz), 7.85 (d, IH, J=7.9Hz), 3.72-3.77 (m, 6H), 3.89 (t, 4H, J=4.8Hz), 6.79 (dt, IH, J=6.2Hz, 1.2Hz), 7.22 (m, 2H), 7.79 (t, IH, J=I.9Hz); MS (ESI+) 454.4 (MH+).
47 : 1 - [2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno [3,2-d] pyrimidin-6- ylmethylj-piperidine^-carboxylic acid amide was prepared from isonipecotamide. 1H NMR (400 MHz, </6-DMSO) 1.55-1.71 (m, 4H), 2.03-2.10 (m, 3H), 2.91-2.94 (m, 2H), 3.79-3.83 (m, 6H), 3.96-3.99 (m, 4H), 6.73 (br s, IH), 6.85 (d, IH), 7.23-7.28 (m, 2H), 7.36 (s, IH), 7.83-7.85 (m, 2H), 9.48 (s, IH); MS (ESI+) 454.1 (MH+).
48: 2-{4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yI-thieno[3,2-d]pyrimidin-6- ylmethyl]-piperazin-l-yl}-iV,7V-dimethylacetamide was prepared from N5N- dimethyl-2-piperazin-l-yl-acetamide dihydrochloride. This in turn was prepared by reaction of N-BOC-piperazine with 2-chloro-iV>jV-dimethylacetamide in dichloromethane, followed by cleavage of the BOC protecting group using HCl (2M) in dichloromethane.
1H NMR (400 MHz, CDCl3) 2.61 (br s, 8H, 4 x CH2),2.94 (s, 3H, CH3), 3.07 (s, 3H, CH3), 3.19 (s, 2H, CH2), 3.81 (s, 2H, CH2), 3.86-3.88 (m, 4H, 2 x CH2), 4.02-4.04 (m, 4H, 2 x CH2), 5.61 (br s, H, OH), 6.92 (d, H, ArH, J=7.56Hz), 7.28 (s, H, ArH), 7.31 (t, H, ArH, J=6.9Hz), 7.9 (s, H, ArH), 7.98 (d, H, ArH, J=7.82Hz); MS (ESI+) 497.1 (MH+).
49: 3-(6-{[MethyI-(l-methyl-pyrrolidin-3-yI)-amino]-methyl}-4-morpholin-4- yl-thieno[3,2-d]pyrimidin-2-yl)-phenol was prepared from vV,7V'-dimethyl-3- aminopyrrolidine. 1H NMR (400 MHz, J6-DMSO) 1.72 (m, IH), 1.94 (m, IH), 2.19 (s, 3H), 2.25 (s, 3H), 2.42 (m, IH), 2.58 (m, 2H), 3.18 (m, 2H), 3.79 (m, 4H), 3.86 (s, 2H), 3.96 (m, 4H), 6.85 (dd, IH, J=6.9Hz), 7.26 (t, IH, J=7.7Hz), 7.35 (s, IH), 7.82 (s, IH), 7.84 (s, IH), 9.46 (s, IH); MS (ESI+) 440.1 (MH+).
50: 2-Dimethylamino-l-{4-[2-(3-hydroxy-phenyI)-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-6-ylmethyl]-piperazin-l-yl}-ethanone was prepared from 2-dimethylamino-l-piperazin-l-yl-ethanone. This in turn was prepared from 1-BOC- piperazine and bromoacetyl bromide in dichloromethane, followed by reaction with dimethylamine hydrochloride and subsequent deprotection of the BOC-group using HCl (2M solution in ether) in dichloromethane.
1H NMR (400 MHz, CDCl3) 2.28 (s, 6H), 2.53-2.55 (m, 4H), 3.12 (s, 2H), 3.65-3.67 (4H, br m), 3.83 (2H, s), 3.87-3.90 (4H, m), 4.04-4.06 (4H, m), 6.92 (IH, m), 7.31 (IH, s), 7.33 (t, IH, J=7.9Hz), 7.93 (s, IH), 8.00 (d, IH, J=7.8Hz); MS (ESI+) 497.1 (MH+).
51 : 3-[6-((3R,5S)-3,5-Dimethyl-piperazin-l-ylmethyl)-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-2-yl]-phenol was prepared from 2,6-dimethylpiperazine. 1H NMR (400 MHz, J6-DMSO) 0.97 (s, 6H), 1.74 (br m, 2H), 2.80-2.82 (br m, 2H), 2.90 (br m, 2H), 3.79-3.81 (m, 4H), 3.83 (s, 2H), 3.95-3.97 (m, 4H), 6.84-6.88 (m, IH), 7.26 (t, IH, J=8.0Hz), 7.37 (s, IH), 7.82-7.85 (m, 2H), 9.46 (br s, IH); MS (ESr+) 440.1 (MH+).
54: 3-{6-[4-(3-DimethyIamino-propane-l-suIfonyl)-piperazin-l-ylmethyl]-4- morpholin-4-yl-thieno[3,2-d]pyrimidin-2-yl}-phenol was prepared from dimethyl- [3-(piperazine-l-sulfonyl)-propyl]-amine . This was prepared as follows: to a solution of N-BOC-piperazine (1.55g, 8.32mmol) in dry DCM was added triethylamine (1.28mL, 9.18mmol, l.leq.) then 3-chloropropane sulfonyl chloride (1.ImL, 9.05mmol, 1.09eq.). The mixture was stirred at rt overnight then diluted with CH2Cl2 and extracted washed with water (3 x 4OmL). The CH2Cl2 was dried (Na2SO4) then purified by flash chromatography (silica gel, 4% MeOH/CH2Cl2) to yield 4-(3-chloro-propane-l-sulfonyl)-piperazine-l-carboxylic acid tert-buty\ ester as a white solid (2.68g, 98%).
A mixture of 4-(3-chloro-propane-l-sulfonyl)-piperazine-l-carboxylic acid tert-butyl ester (152.6mg, 0.47mmol), dimethyamine hydrochloride ( 403.5 mg, 4.95 mmol, 10.6eq.), potassium carbonate (72.7mg, 0.53mmol, 1.13eq.) and triethylamine (690μL, 4.95mmol, 10.6eq) in acetonitrile (1OmL) plus a catalytic amount of potassium iodide was heated at 80°C in a sealed tube overnight. The solvent was then removed in vacuo. The residue diluted with water and extracted with CH2Cl2 (3 x 2OmL). The combined fractions were dried (Na2SO4) to yield 4-(3-dimethylamino- propane-l-sulfonyl)-piperazine-l-carboxylic acid tert-butyl ester as an orange solid (176 mg).
To a solution of 4-(3-dimethylamino-propane-l-sulfonyl)-piperazine-l- carboxylic acid tert-butyl ester (167mg, 0.5mmol) in dry CH2Cl2 (8mL)was added 2 M HCl in ether (8mL, lόmmol, 32eq.). The mixture was stirred at RT overnight. Solvent was then removed in vacuo and the residue used directly (163.5 mg, 100%). 1H NMR (400 MHz, CDCl3) 1.96-2.02 (m, 2H, 2 x CH2), 2.26 (s, 6H, 2 x CH3), 2.43 (t, 2H, CH2, J=6.8Hz), 2.54-2.56 (m, 4H, 2 x CH2), 2.95-2.99 (m, 2H, CH2), 3.26- 3.28 (m, 4H, 2 x CH2), 3.74 (s, 2H, CH2), 3.80-.88 (m, 4H, 2 x CH2), 4.01-4.03 (m, 4H, 2 x CH2), 6.88 (d, H, ArH, J=7.74Hz), 7.24 (s, H, ArH), 7.30 (t, H, ArH,
J=7.88Hz), 7.92 (m, H, ArH), 7.96 (d, H, ArH, J=7.82Hz); MS (ESI+) 561.1 (MH+).
55: 3-[6-(4-Methoxy-piperidin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol was prepared from 4-methoxypiperidine. This in turn was prepared by reaction of tert-butyl-4-hydroxy- 1 -piperidine carboxylate with methyl iodide, followed by cleavage of the BOC group using standard conditions; 1H NMR (400 MHz, ^6-DMSO) 1.47 (m, 2H), 1.83 (m, 2H), 2.25(m, 2H), 2.72 (m, 2H), 3.21 (s, 3H), 3.81 (m, 6H), 3.97 (m, 4H), 6.85 (m, IH), 7.26 (t, lH, J=8.1Hz), 7.35 (s, IH), 7.83 (m, 2H), 9.46 (s, IH); MS (ESf) 441.1 (MH+). 58: 3-{4-Morpholin-4-yI-6-[4-(3-morpholin-4-yl-propane-l-sulfonyl)- piperazin-l-ylmethyl]-thieno[3,2-d]pyrimidin-2-yl}-phenol was prepared from 4- [3-(piperazine-l-sulfonyl)-propyl]-moφholine. This was prepared in an analogous manner to dimethyl-[3-(piperazine-l-sulfonyl)-propyl]-amine using morpholine as the amine.
1H NMR (400 MHz, CDCl3) 1.98-2.04 (m, 2H, CH2), 2.44-2.47 (m, 6H, 3 x CH2), 2.63-2.65 (m, 4H, 2 x CH2), 2.98-3.02 (m, 2H, CH2), 3.33-3.35 (m, 4H, 2 x CH2), 3.68-3.71 (m, 4H, 2 x CH2), 3.85 (s, 2H, CH2), 3.87-3.90 (m, 4H, 2 x CH2), 4.03- 4.06 (m, 4H, 2 x CH2), 5.29 (br s, H, OH), 6.91-6.93 (m, H, ArH), 7.31-7.35 (m, 2H, 2 x ArH), 7.91-7.92 (m, H, ArH), 7.99-8.01 (m, H, ArH); MS (ESI+) 603.2 (MH+).
57: {4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyl]-piperazin-l-yl}-morpholin-4-yl-methanone was prepared from morpholin-4-yl-piperazin-l-yl-methanone. This in turn was prepared by reaction of N-BOC-piperazine with 4-morpholino carbonyl chloride in acetonitrile, followed by cleavage of the BOC protecting group using HCl (2M) in dichloromethane. 1H NMR (400 MHz, CDCl3) 2.54-2.56 (m, 4H, 2 x CH2), 3.25-3.27 (m, 4H, 2 x CH2), 3.32-3.34 (m, 4H, 2 x CH2),3.66-3.68 (m, 4H, 2 x CH2), 3.83 (s, 2H, CH2), 3.87-3.89 (m, 4H, 2 x CH2), 4.03-4.05 (m, 4H, 2 x CH2), 5.33(br s, H, OH), 6.91- 6.93 (m, H, ArH), 7.30-7.34 (m, 2H, 2 x ArH), 7.91-7.92 (m, H, ArH), 7.99-(d, H, ArH, J=7.82Hz); MS (ESI+) 525.1 (MH+).
56: 4-[2-(3-Hydroxy-phenyI)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyl]-piperazine-l-carboxylic acid (2-methoxy-ethyl)-methyl-amide was prepared from piperazine-1-carboxylic acid (2-methoxy-ethyl)-methyl-amide. This was prepared as follows:
To 1-BOC-piperazine (500mg, 2.68mmol) in dichloromethane (5mL) and triethylamine (l.leq., 410μL) was added 4-nitrophenylchloroformate (leq., 541mg).
After stirring at room temperature for 1 hour, the reaction mixture was diluted with dichloromethane, washed with brine, dried (MgSO4), filtered and evaporated in vacuo to give piperazine-l,4-dicarboxylic acid tert-butyl ester 4-nitro-phenyl ester as a light-yellow solid (940mg, 100%).
To piperazine-l,4-dicarboxylic acid tert-butyl ester 4-nitro-phenyl ester (500mg, 1.42mmol) in anhydrous THF (5mL) was added N-(2-methoxy- ethyl)methylamine (2eq., 254mg). The reaction mixture was refluxed overnight, then evaporated in vacuo and purified by chromatography to give 4-[(2-methoxyethyl)- methyl-carbamoyl]piperazine-l-carboxylic acid tert-butyl ester as a clear oil (304mg, 71%). To 4-[(2-methoxyethyl)-methyl-carbamoyl]piperazine-l-carboxylic acid tert- butyl ester (304mg, l.Olmmol) in dichloromethane (3mL) was added HCl (2M solution in ether), the reaction mixture stirred at rt overnight and then evaporated in vacuo to give piperazine-1-carboxylic acid (2-methoxy-ethyl)-methyl-amide as HCl salt (330 mg).
1H NMR (400 MHz, CDCl3) 2.55-2.57 (m, 4H), 2.90 (s, 3H), 3.29-3.31 (m, 4H), 3.34 (s, 3H), 3.38 (t, 2H, J=5.5Hz), 3.54 (t, 2H, J=5.5Hz), 3.83 (s, 2H), 3.88-3.90 (m, 4H), 4.04-4.06 (m, 4H), 5.37 (brs,lH),6.93 (dd, IH, J=7.8Hz, 2.8Hz), 7.26-7.35 (m, 2H), 7.93 (s, IH), 8.01 (d, IH, J=7.8Hz); MS (ESI+) 527.3 (MH+).
Example 3: 3-(4-Morpholin-4-v--6-p-perazin-l-ylmethvI-thieno[3,2- dlpyrimidin-2-yl)-phenol, trifluoroacetic acid salt (18) To a solution of 4-[2-(3-hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-6-ylmethyl]-piperazine-l-carboxylic acid tert-butyl ester (95mg) in dichloromethane (1OmL) was added trifluoroacetic acid (2mL) After 3 hours the reaction mixture was concentrated in vacuo. Ether was added to the residue and the mixture was reduced in vacuo again to yield the title compound as a white foam. 1H NMR (400 MHz, 4-DMSO) 2.70-2.75 (m, 4H), 3.10-3.15 (m, 4H), 3.82-3.86 (m, 4H), 3.98-4.02 (m, 6H), 6.91 (d, IH), 7.31-7.33 (m, IH), 7.48 (s, IH), 7.79- 7.81 (m, 2H), 8.75 (br s, 2H), 9.60 (br s, IH); MS (ESI+) 412 (MH+).
Example 4: 1 - (4- [2-(3-Hydroxy-phen vD-4-morpholin-4-yI-thieno [3,2- d1pyrimidin-6-ylmethvn-piperazin-l-vU-2-methoxy-ethanone (37) A mixture of 3-(4-morpholin-4-yl-6-piperazin-l-ylmethyl-thieno[3,2- d]pyrimidin-2-yl)-phenol, trifluoroacetic acid salt (50mg, 0.08mmol), dichloromethane (ImL), triethylamine (45μL) and methoxyacetyl chloride (19μL, 2.5eq.) was stirred at room temperature. After 3 hours the reaction mixture was diluted with dichloromethane, washed with sodium bicarbonate solution, dried
(MgSO4) and the solvent removed in vacuo to yield a pale solid. This was stirred in a mixture of methanol and sodium carbonate overnight. This mixture was then diluted with chloroform, washed with brine, dried (MgSO4) and the solvent removed in vacuo to yield a gum which was purified using flash chromatography to yield the desired title compound ( 13mg)
1H NMR (400 MHz, CDCl3) 7.99 (IH, d), 7.90 (IH, s), 7.38-7.29 (2H, m), 6.90 (IH, d),5.20 (IH, br s), 4.10 (2H,s), 4.02-3.99 (4H,m), 3.89-3.85 (4H, m), 3.79 (2H, s), 3.75-3.73 (2H, m), 3.51-3.49 (2H, s), 3.40 (3H, s), 2.52-2.48 (4H, m); MS (ESI4") 484 (MH+).
Example 5: l-{(2R,6S)-4-[2-(3-hvdroxy-phenyl)-4-morpholin-4-yl-thienor3,2- d1pyrimidin-6-ylmethyll-2,6-dimethyI-piperazin-l-yl}-ethanone (53) To a solution of 2-[3-(tert-butyl-dimethyl-silanyloxy)-phenyl]-6-((3R,5S)- 3,5-dimethyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine
(80mg, 0.14mmol) in dichloromethane (5mL) was added triethylamine (40 μL, 2.0 equivalents) followed by acetyl chloride (15μL, 1.5eq.). After stirring overnight, the reaction mixture was diluted with dichloromethane, washed with aqueous sodium carbonate solution, dried (MgSO4) and the solvent removed in vacuo to yield 1- ((2R,6S)-4-{2-[3-(tert-butyl-dimethyl-silanyloxy)-phenyl]-4-moφholin-4-yl- thieno[3,2-d]pyrimidin-6-ylmethyl}-2,6-dimethyl-piperazin-l-yl)-ethanone (80mg) as a white solid. Removal of the tert-butyldimethylsilyl protecting group was performed as described above to yield the title compound. 1H NMR (400 MHz, ^6-DMSO; 373K) 1.29 (d, 6H), 1.99 (s, 3H), 2.28 (dd, 2H), 2.80 (d, 2H), 3.82-3.85 (m, 4H), 3.90 (s, 2H), 3.99-4.01 (m, 4H), 4.19-4.21 (m, 2H), 6.90 (d, IH), 7.23-7.26 (m, IH), 7.35 (s, IH), 7.84-7.88 (m, 2H), 9.10 (brs, IH). MS (ESf) 482.1 (MH+)
Example 6: 3-[6-((3R,5S)-4-Methanesulfonyl-3,5-dimethyI-pipera2in-l-
VImethyl)-4-morpholin-4-yl-thieno[3,2-d1pyrimidin-2-vn-phenol (52)
The title compound 52 was prepared by analogy with the process of Example 5, using methane sulfonyl chloride.
1H NMR (400 MHz, CDCl3/MeOD): 1.48 (s, 6H), 2.30 (br m, 2H), 2.76 (br m, 2H), 2.85 (s, 3H), 3.81 (s, 2H), 3.87 (br m, 4H), 4.04 (br m, 6H), 6.94 (br m, IH), 7.26- 7.29 (br m, 2H), 7.74 (br s, IH), 7.86 (br m, IH); MS (ESf) 518.2 (MH+).
Example 7: 3-F6-(4-Amino-piperidin-l-ylmethyl)-4-morpholin-4-yl-thienof3,2- dlpyrimidin-2-vU-phenol (32)
A mixture of (510mg, 1.12mmol), 4-(N-BOC-amino)piperidine (1.3eq., 292mg), acetic acid (leq., 65μL) and sodium triacetoxyborohydride (1.5eq., 357mg) in 1,2-dichloroethane (7mL) was stirred at RT overnight. The reaction mixture was diluted with dichloromethane, washed with saturated solution of sodium hydrogen carbonate, brine, separated and dried (MgSO4). The crude product was evaporated in vacuo and purified by chromatography to give (l-{2-[3-(tgrt-butyl-dimethyl- silanyloxy)-phenyl]-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl}-piperidin- 4-yl)-carbamic acid tert-butyl ester (570 mg, 89%). (l-{2-[3-(7ert-butyl-dimethyl-silanyloxy)-phenyl]-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-6-ylmethyl}-piperidin-4-yl)-carbamic acid tert-butyl ester (250 mg, 0.39 mmol) was dissolved in THF (4 mL), to this was added TBAF (1.4eq., 0.55mL). After 30 min. the reaction mixture was quenched with silica, evaporated in vacuo and purified by chromatography to give {l-[2-(3-hydroxy-phenyl)-4- moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]-piperidin-4-yl}-carbamic acid tert-butyl ester as a white solid (179 mg, 88%).
{l-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyl]-piperidin-4-yl}-carbamic acid tert-butyl ester (179 mg, 0.34 mmol) was dissolved in dichloromethane (3mL), to this was added HCl (2M solution in ether, ImL, excess). The reaction mixture stirred at room temperature for 1 hour and then evaporated in vacuo to give the title compound as di-HCl salt (197 mg). 1H NMR (400 MHz, J6-DMSO) 1.98-2.04 (br m, 2H), 2.11-2.18 (br m, 2H), 3.04- 3.13 (br m, 2H), 3.25-3.28 (br m, IH), 3.50-3.54 (br m, 2H), 3.83-3.87 (br m, 4H), 4.05-4.07 (br m, 4H), 4.70 (br s, 2H), 6.96 (d, IH), 7.34 (t, IH, J=7.8Hz), 7.86 (IH, s), 7.88 (d, IH), 7.95 (br s, IH), 8.30 (br s, 3H), 9.70 (br s, IH), 11.65 (br s, IH); MS (ESI+) 426.1 (MH+).
Example 8: N-{l-[2-(3-Hvdroxy-phenyl)-4-morpholin-4-yl-thieno[3,2- dlpyrimidin-6-vIinethyll-piperidin-4-vU-inethanesulfonamide
(36)
To 3-[6-(4-Amino-piperidin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol di-hydrochloride salt (60 mg, 0.12 mmol) in DMF (ImL) was added triethylamine (3eq., 50μL) and the reaction mixture stirred for 30 min. Next, methanesulfonic acid benzotriazol-1-yl ester (l.leq., 28mg) was added and the mixture stirred at RT for 3 hours. Methanesulfonic acid benzotriazol-1-yl ester was prepared according to the procedure described in Tetrahedron Lett., 1999, 40, 117- 120.
The reaction mixture was diluted with dichloromethane, washed with brine, separated and dried (MgSO4). The crude product was evaporated in vacuo, purified by chromatography to give the title compound as a white solid (39mg, 80%). 1H NMR (400 MHz, J6-DMSO) 1.47-1.51 (m, 2H), 1.81-1.85 (m, 2H), 2.12-2.20 (m, 2H), 2.83-2.87 (m, 2H), 2.91 (s, 3H), 3.12-3.15 (m, IH), 3.79-3.83 (m, 6H), 4.04- 4.06 (m, 4H), 6.85 (d, IH), 7.04 (d, IH), 7.26 (t, lH, J=8.1Hz), 7.37 (s, IH), 7.82- 7.84 (m, 2H), 9.48 (s, IH); MS (ESI+) 504.1 (MH+). Example 9 N-U-[2-(3-Hvdroxy-phenvD-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-6-ylmethyl1-piperidin-4-vU-acetamide (38) To 3-[6-(4-Amino-piperidin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-2-yl] -phenol di-hydrochloride salt (80mg, O.lόmmol) in dichloromethane (1.5mL) and triethylamine (5eq., HOμL) was added acetyl chloride (2.5eq., 29μL). After stirring at rt for 2 hours, the reaction mixture was diluted with dichloromethane, washed with brine, separated and dried (MgSO4). The crude product was evaporated in vacuo, re-dissolved in methanol (2mL), to this was added sodium carbonate. The reaction mixture was stirred at RT overnight, evaporated in vacuo, diluted with dichloromethane, washed with brine, separated and dried (MgSO4). The residue was purified by chromatography to give the title compound as a white solid (55 mg).
1H NMR (400 MHz, CDCl3) 1.45-1.54 (m, 2H), 1.94-1.97 (m, 2H), 1.97 (s, 3H), 2.22-2.27 (m, 2H), 2.90-2.93 (m, 2H), 3.79 (s, 2H), 3.80-3.83 (m, IH), 3.85-3.90 (m, 4H), 4.04-4.06 (m, 4H), 5.31 (d, IH), 6.92-6.94 (m, IH), 7.31 (s, IH), 7.33 (t, IH, J=7.9Hz), 7.91 (s, IH), 8.00 (d, IH, J=7.8Hz); MS (ESf) 468.1 (MH+).
Reference Example 1 : 2,4-Dichloro-thieno[3,2-d1pyrimidine (64)
Figure imgf000057_0001
A mixture of methyl 3-amino-2-thiophenecarboxylate (13.48 g, 85.85mmol) and urea (29.75g, 5eq.) was heated at 19O0C for 2 hours. The hot reaction mixture was then poured onto sodium hydroxide solution and any insoluble material removed by filtration. The mixture was then acidified (HCl, 2N) to yield lH-thieno [3,2- d]pyrimidine-2,4-dione (63) as a white precipitate, which was collected by filtration and air dried (9.49g, 66%).
1H NMR (400 MHz, J6-DMSO) 6.90 (IH, d, J=5.2Hz), 8.10 (IH, d, J=5.2Hz), 11.60-11.10 (2H, br, s).
A mixture of lH-thieno[3,2-d]pyrimidine-2,4-dione (9.49g, 56.49mmol) and phosphorous oxychloride (15OmL) was heated at reflux for 6 hours. The reaction mixture was then cooled and poured onto ice/water with vigorous stirring yielding a precipitate. The mixture was then filtered to yield 2,4-dichloro-thieno[3,2- d]pyrimidine (64) as a white solid (8.68 g, 75%). 1H NMR (400 MHz, CDCl3) 7.56 (IH, d, J=5.5Hz).8.13 (IH, d, J=5.5Hz).
Reference Example 2: 2-Chloro-4-morpholin-4-yl-thieno[3,2-dlpyrimidine
(65)
Figure imgf000058_0001
(65)
A mixture of 2,4-dichloro-thieno[3,2-d]pyrimidine (64), (8.68g, 42.34mmol), morpholine (8.1 ImL, 2.2eq.) and methanol (15OmL) was stirred at room temperature for 1 hour. The reaction mixture was then filtered, washed with water and methanol, to yield the title compound as a white solid (11.04 g, 100%).
1H NMR (400 MHz, ^6-DMSO) 3.74 (4H, t, J=4.9Hz), 3.90 (4H, t, J=4.9Hz), 7.40 (IH, d, J=5.6Hz), 8.30 (IH, d, J=5.6Hz). Reference Example 3: 2-Chloro-4-morpholin-4-v--thieno[3,2- dlpyrimidine-6-carbaIdehyde (66)
Figure imgf000059_0001
(66)
To a suspension of 2-chloro-4-morpholin-4-yl-thieno[3,2-d]pyrimidine (65) (1.75g, 6.85mmol) in dry THF (4OmL) at -78°C was added a 2.5M solution of nBuLi in hexane (3.3mL, 1.2eq.). After stirring for 1 hour, dry N,N-dimethylformamide (796μL, 1.5eq.) was added. The reaction mixture was stirred for 1 hour at -780C and then warmed slowly to room temperature. After a further 2 hours at room temperature the reaction mixture poured onto ice/water yielding a yellow precipitate.
This was collected by filtration and air-dried to yield the title compound (1.50 g,
77%).
1H NMR (400 MHz, J6-DMSO) 3.76 (4H, t, J=4.9Hz), 3.95 (4H, t, J=4.9Hz), 8.28 (IH, s), 10.20 (IH, s).
Reference Example 4: 4-(4,4,5,5-Tetramethyl- [1 ,3,21 dioxaborolan-2-yI)- lH-indazole (70)
Figure imgf000060_0001
(67) (68)
Figure imgf000060_0002
(70) (69)
To a solution of 2-methyl-3-nitroaniline (2.27g, 14.91mmol) in acetic acid (6OmL) was added a solution of sodium nitrite (1.13g, 1.1 eq.) in water (5mL). After 2 hours, the deep red solution was poured onto ice/ water and the precipitate collected by filtration to yield 4-nitro-lH-indazole (67) (1.98g, 81%).
A mixture of 4-nitro-lH-indazole (760mg, 4.68 mmol), palladium on charcoal (10%, cat.) and ethanol (3OmL) was stirred under a balloon of hydrogen for 4 hours. The reaction mixture was then filtered through celite, and the solvent removed in vacuo to yield lH-indazol-4-ylamine (68) (63 lmg, 100%).
An aqueous solution of sodium nitrite (337mg, 4.89mmol) in water (2mL) was added dropwise to a suspension of lH-indazol-4-ylamine (63 lmg, 4.74mmol) in 6M hydrochloric acid (7.2mL) at below O0C. After stirring for 30 minutes sodium tetrafluorobrate (724mg) was added. The reaction mixture became very thick and was filtered and washed briefly with water to yield lH-indazole-4-diazonium, tetrafluoroborate salt (69) (218mg, 20%) as a deep red solid. Dry methanol (4mL) was purged with argon for 5 minutes. To this was added IH- indazole-4-diazonium, tetrafluoroborate salt (218mg, 0.94mmol), bis-pinacolato diboron (239mg, l.Oeq.) and [l,r-bis(diphenylphosphino)ferrocene]palladium (II) chloride (20mg). The reaction mixture was stirred for 5 hours and then filtered through celite. The residue was purified using flash chromatography to yield the desired title compound (70), (117mg).
Reference Example 5: Preparation of 2-(lH-Indazol-4-vD-4-morpholin-4- yl-thieno[3,2-dlpyrimidine-6-carbaldehyde (71)
Figure imgf000061_0001
A mixture of 2-chloro-4-morpholin-4-yl-thieno[3,2-d]pyrimidine-6-carbaldehyde (66) (lOOmg, 0.35mmol), 4-(4,4,5,5-tetramethyl-[l,3,2]dioxaborolan-2-yl)-lH- indazole (70) (95mg, 0.39mmol) and sodium carbonate (112mg) were suspended in toluene (2.5mL), ethanol (1.5mL) and water (0.7mL). To this was added bis(triphenylphosphine)palladium(II) chloride (13.5mg) and the reaction vessel was flushed with argon. The reaction mixture was microwaved at 120°C for 1 hour and then partitioned between dichloromethane and water, the organic layer was washed with brine, dried over magnesium sulfate, filtered and evaporated in vacuo. The resulting residue was purified using flash chromatography to yield the title compound 71 (97mg).
Reference Example 6 Preparation of 2-(lH-Indazol-4-vD-6-(4-methyI-piperazin- l-ylmethyl)-4-morpholin-4-yl-thieno[3,2-dlpyrimidine
(59) To a mixture of 2-(lH-Indazol-4-yl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidine-6- carbaldehyde (91mg, 0.26mmol), 1-methylpiperazine (34mg, 0.36mmol) and acetic acid (15uL) in 1 ,2-dichloroethane (2mL) was added sodium triacetoxyborohydride (60mg, 0.28mmol). The reaction mixture was stirred at room temperature overnight and then basified (NaHCO3, saturated), diluted with dichloromethane, washed with brine. Organic layer was separated, dried (MgSO4), filtered and evaporated in vacuo. The residue was purified using flash chromatography to give the title compound (33 mg).
The following compounds were prepared by analogy with the procedure described above.
2-{4-[2-(lH-Indazol-4-yl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin-l-yl}-ethanol using l-(2-hydroxyethyl)piperazine. 1H NMR (400 MHz, J6-DMSO) 2.40 (br m, 2H), 2.42-2.52 (b, 8H, under DMSO peak), 3.48 (q, 2H, J=6.0Hz), 3.82-3.86 (m, 6H), 3.98-4.01 (m, 4H), 4.34 (br s, IH,), 7.44-7.48 (m, 2H), 7.65 (d, lH, J=8.3Hz), 8.21 (d, lH, J=6.8Hz), 8.87 (s, IH), 13.15 (brs, IH); MS (ESI+) 480.1 (MH+).
4- [2-(l H-Indazol-4-yl)-4-morpholin-4-yl-thieno [3,2-d]pyrimidin-6-ylmethyl] - piperazine-1-sulfonic acid dimethylamide using piperazine-1 -sulfonic acid dimethylamide.
1H NMR (400 MHz, CDCl3) 2.63-2.66 (m, 4H), 2.84 (s, 6H), 3.31-3.34 (m, 4H), 3.89 (s 2H), 3.92-3.94 (m, 4H), 4.08-4.11 (m, 4H), 7.39 (s, IH), 7.51 (t, IH, J=8.1Hz), 7.60 (d, IH, J=8.1Hz), 8.28 (d, IH, J=6.7Hz), 9.02 (s, IH), 10.12 (br s IH); MS (ESI+) 543.1 (MH+).
Example 6: Further Compounds of the Invention
The following compound were prepared by analogy with the procedure described in Reference Example 6: 176: l-{3-[6-(4-Methyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenyl}-ethanol.
As for 59, except treatment of 3-[6-(4-methyl-piperazin-l-ylmethyl)-4- morpholin-4-yl-thieno [3 ,2-d]pyrimidin-2-yl] -benzaldehyde with methyl magnesium bromide in THF/ether yielded the title compound.
1H NMR (400MHz, CDCl3): 1.49 (d, J=6.5, 3H), 2.10 (d, J=I.7Hz, IH), 2.25 (s, 3H), 2.46 (br s, 4H), 2.54 (s, br, 4H), 3.74 (s, 2H), 3.82 (t, J=4.8Hz, 4H), 3.98 (t, J=4.8, 4H), 4.94 (q, J=6.4Hz, IH), 7.23 (s, IH), 7.35-7.42 (m, 2H), 8.27 (m, IH), 8.35 (s, IH); MS (ESI+) 454.27 (MH+).
177: 3-[6-(4-Methyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenyl}-methanoI.
As for 59, except treatment of 3-[6-(4-methyl-piperazin-l-ylmethyl)-4- moφholin-4-yl-thieno[3,2-d]pyrimidin-2-yl]-benzaldehyde with sodium borohydride in ethanol at room temperature yielded the title compound.
1H NMR (400MHz, CDCl3): 2.25 (s, 3H), 2.47 (s, 4H), 2.54 (s, 4H), 3.75 (s, 2H),
3.80 (t, J=4.8Hz, 4H), 3.98 (t, J=4.8Hz, 4H), 4.71 (s, 2H), 7.23 (m, IH), 7.38 (m,
2H), 8.28 (m, IH), 8.34 (s, IH); MS (ESf) 440.23 (MH+).
178: 6-(4-Methyl-piperazin-l-yImethyl)-4-morpholin-4-yl-2-(lH-pyrazol-4- yl)-thieno [3,2-d] py rimidine. As for compound 59.
1H NMR (400 MHz, c?6-DMSO): 2.15 (s, 3H), 2.41 (m, 8H), 3.77 (m, 4H), 3.85 (s, 2H), 3.92 (m, 4H), 7.26 (s, IH), 8.31 (br s, IH), 13.05 (br s, IH); MS (ESI+) 400.21
(MH+).
Reference Example 7 2-chloro-6-(4-methyl-piperazin-l -yl methyQ-4- morpholin-4-yl-thieno[3,2-d1pyrimidine (72)
Figure imgf000064_0001
To a mixture of 2-chloro-4-morpholin-4-yl-thieno[3,2-d]pyrimidine-6- carbaldehyde (66) (147mg, 0.52mmol), 1-methyl-piperazine (1.5eq., 87μL) and acetic acid (1.05eq., 32μL) in 1 ,2-dichloroethane (3mL) was added sodium triacetoxyborohydride (l.leq., 121mg) and then stirred at room temperature overnight. The reaction mixture was diluted with dichloromethane, washed with a saturated solution of sodium hydrogen carbonate, brine, separated and dried (MgSO4). The crude product was evaporated in vacuo and purified by chromatography to give the title compound 72 as an off-white crystalline solid (51 mg, 45%).
Example 11 Biological Testing
Compounds of the invention, prepared as described in the preceding Examples, were submitted to the following series of biological assays:
(i) PI3K Biochemical Screening
Compound inhibition of PI3K was determined in a radiometric assay using purified, recombinant enzyme and ATP at a concentration of IuM. All compounds were serially diluted in 100% DMSO. The kinase reaction was incubated for 1 hour at room temperature, and the reaction was terminated by the addition of PBS. IC5O values were subsequently determined using sigmoidal dose-response curve fit (variable slope). All of the compounds exemplified had an IC50 against PI3K of 5OuM or less. (ii) Cellular Proliferation Inhibition
Cells were seeded at optimal density in a 96 well plate and incubated for 4 days in the presence of test compound. Alamar Blue™ was subsequently added to the assay medium, and cells were incubated for 6 hours before reading at 544nm excitation, 590nm emission. EC50 values were calculated using a sigmoidal dose response curve fit. All the compounds tested had an EC50S of 5OuM or less in the range of cell lines utilized.
(iii) Caco-2 Permeability
Caco-2 cells were seeded onto Millipore Multiscreen plates at 1 x 105 cells/cm2, and were cultured for 20 days. Assessment of compound permeability was subsequently conducted. The compounds were applied to the apical surface (A) of cell monolayers and compound permeation into the basolateral (B) compartment was measured. This was performed in the reverse direction (B-A) to investigate active transport. A permeability coefficient value, Papp, for each compound, a measure of the rate of permeation of the compound across the membrane, was calculated. Compounds were grouped into low (Papp </= 1.0 x 106cm/s) or high (Papp >/= 1.0 x 106cm/s) absorption potential based on comparison with control compounds with established human absorption.
For assessment of a compound's ability to undergo active efflux, the ratio of basolateral (B) to apical (A) transport compared with A to B was determined. Values of B- A/ A-B >/= 1.0 indicated the occurrence of active cellular efflux. All of the compounds tested through the Caco-2 permeability screen had Papp values >/= 1.0 x 106cm/s. One compound assessed through the bidirectional assay, PI540, had an B- A/ A-B asymmetry index of less than 1.0, indicating that the compound does not undergo active cellular efflux.
(iv) Hepatocvte Clearance Suspensions of cryopreserved human hepatocytes were used. Incubations were performed at compound concentration of ImM or 3μM at a cell density of 0.5 x 106 viable cells/mL. The final DMSO concentration in the incubation was 0.25%. Control incubations were also performed in the absence of cells to reveal any non- enzymatic degradation. Duplicate samples (50μL) were removed from the incubation mixture at 0, 5, 10, 20, 40 and 60 minutes (control sample at 60 minutes only) and added to methanol - containing internal standard (lOOμL) - to terminate the reaction. Tolbutamide, 7-hydroxycoumarin, and testosterone were used as control compounds. Samples were centrifuged and the supernatants at each time point pooled for analysis by LC-MSMS. From a plot of In peak area ratio (parent compound peak area / internal standard peak area) against time, intrinsic clearance (CLjnO was calculated as follows: CLjnt (μl/min/million cells) = V x k, where k is the elimination rate constant, obtained from the gradient of In concentration plotted against time; V is a volume term derived from the incubation volume and is expressed as uL 106 cells"1. Compounds were classified with low (CL</= 4.6μL/min/106 cells), medium
(CL >/= 4.6; </= 25.2 μl/min/106 cells) and high (>/= 25.2μl/min/106 cells) clearance. The majority of the tested compounds of the invention were determined to have low hepatocyte clearance.
(v) Cytochrome P450 Inhibition
Compounds of the invention were screened against five CYP450 targets (1 A2, 2C9, 2C19, 2D6, 3A4) at 10 concentrations in duplicate, with a top concentration of lOOuM being used. Standard inhibitors (furafylline, sulfaphenazole, tranylcypromine, quinidine, ketoconazole) were used as controls. Plates were read using a BMG LabTechnologies PolarStar in fluorescence mode. The majority of the tested compounds assessed in this assay displayed weak activity (IC50 >/=5uM) against all isoforms of CYP450.
(vi) Cytochrome P450 Induction Freshly isolated human hepatocytes from a single donor were cultured for 48 hours prior to addition of test compound at three concentrations and were incubated for 72 hours. Probe substrates for CYP3A4 and CYP 1A2 were added for 30 minutes and 1 hour before the end of the incubation. At 72 hours, cells and media were removed and the extent of metabolism of each probe substrate quantified by LC- MS/MS. The experiment was controlled by using inducers of the individual P450s incubated at one concentration in triplicate. The compounds of the invention assessed in this assay showed negligible effects on induction of cytochrome P450 enzymes.
(vii) Plasma Protein Binding Solutions of test compound (5um, 0.5% final DMSO concentration) were prepared in buffer and 10% plasma (v/v in buffer). A 96 well HT dialysis plate was assembled so that each well was divided in two by a semi-permeable cellulose membrane. The buffer solution was added to one side of the membrane and the plasma solution to the other side; incubations were then conducted at 370C over 2 hours in triplicate. The cells were subsequently emptied, and the solutions for each batch of compounds were combined into two groups (plasma-free and plasma- containing) then analysed by LC-MSMS using two sets of calibration standards for plasma- free (6 points) and plasma-containing solutions (7 points). The fraction unbound value for each compound was calculated: highly protein bound compounds (>/=90% bound) had an Fu </=0.1. The compounds of the invention assessed in this assay had Fu values >/= 0.1.
(viii) hERG channel blockage
Compounds of the invention were evaluated for their ability to modulate rubidium efflux from HEK-294 cells stably expressing hERG potassium channels using established flux methodology. Cells were prepared in medium containing RbCl and were plated into 96-well plates and grown overnight to form monolayers. The efflux experiment was initiated by aspirating the media and washing each well with 3 x lOOμL of pre-incubation buffer (containing low [K+]) at room temperature. Following the final aspiration, 50μL of working stock (2x) compound was added to each well and incubated at room temperature for 10 minutes. 50μL of stimulation buffer (containing high [K+]) was then added to each well giving the final test compound concentrations. Cell plates were then incubated at room temperature for a further 10 minutes. 80μL of supernatant from each well was then transferred to equivalent wells of a 96-well plate and analysed via atomic emission spectroscopy. Compounds were screened as lOpt duplicate IC50 curves, n=2, from a top concentration of lOOμM.
Example 12 Tablet composition Tablets, each weighing 0.15 g and containing 25 mg of a compound of the invention were manufactured as follows:
Composition for 10,000 tablets
Active compound (250 g) Lactose (800 g)
Corn starch (415g)
Talc powder (30 g)
Magnesium stearate (5 g)
The active compound, lactose and half of the corn starch were mixed. The mixture was then forced through a sieve 0.5 mm mesh size. Corn starch (10 g) is suspended in warm water (90 ml). The resulting paste was used to granulate the powder. The granulate was dried and broken up into small fragments on a sieve of
1.4 mm mesh size. The remaining quantity of starch, talc and magnesium was added, carefully mixed and processed into tablets.
Example 13: Injectable Formulation
Formulation A
Active compound 200 mg Hydrochloric Acid Solution 0.1M or
Sodium Hydroxide Solution 0. IM q.s. to pH 4.0 to 7.0
Sterile water q.s. to 10 ml
The compound of the invention was dissolved in most of the water (35° 40° C) and the pH adjusted to between 4.0 and 7.0 with the hydrochloric acid or the sodium hydroxide as appropriate. The batch was then made up to volume with water and filtered through a sterile micropore filter into a sterile 10 ml amber glass vial (type 1) and sealed with sterile closures and overseals.
Formulation B
Active Compound 125 mg
Sterile, Pyrogen-free, pH 7 Phosphate
Buffer, q.s. to 25 ml
Active compound 200 mg Benzyl Alcohol 0.10 g
Glycofurol 75 1.45 g
Water for injection q.s to 3.00 ml
The active compound was dissolved in the glycofurol. The benzyl alcohol was then added and dissolved, and water added to 3 ml. The mixture was then filtered through a sterile micropore filter and sealed in sterile 3 ml glass vials (type
1).
Example 17: Syrup Formulation
Active compound 250 mg
Sorbitol Solution 1.50 g
Glycerol 2.00 g
Sodium benzoate 0.005 g
Flavour 0.0125 ml Purified Water q.s. to 5.00 ml
The compound of the invention was dissolved in a mixture of the glycerol and most of the purified water. An aqueous solution of the sodium benzoate was then added to the solution, followed by addition of the sorbital solution and finally the flavour. The volume was made up with purified water and mixed well.

Claims

1. A compound which is a fused pyrimidine of formula (I):
Figure imgf000071_0001
wherein
A represents a thiophene or furan ring; n is 1 or 2;
R1 is a group of formula:
Figure imgf000071_0002
wherein m is 0 or 1 ;
R30 is H or Ci-C6 alkyl;
R4 and R5 form, together with the N atom to which they are attached, a 5- or 6- membered saturated N-containing heterocyclic group which includes 0 or 1 additional heteroatoms selected from N, S and O, which may be fused to a benzene ring and which is unsubstituted or substituted; or one of R4 and R5 is alkyl and the other is a 5- or 6-membered saturated N-containing heterocyclic group as defined above or an alkyl group which is substituted by a 5- or 6-membered saturated N- containing heterocyclic group as defined above; R2 is selected from: (a)
R0
— N
R' wherein R6 and R7 form, together with the nitrogen atom to which they are attached, a morpholine, thiomorpholine, piperidine, piperazine, oxazepane or thiazepane group which is unsubstituted or substituted; and (b)
Figure imgf000072_0001
wherein Y is a C2 - C4 alkylene chain which contains, between constituent carbon atoms of the chain and/or at one or both ends of the chain, 1 or 2 heteroatoms selected from O, N and S, and which is unsubstituted or substituted; and R3 is selected from:
(a) a group of the following formula:
Figure imgf000072_0002
wherein B is a phenyl ring which is unsubstituted or substituted and Z is selected from H, -OR, -SR, CH2OR, -CO2R, CF2OH, CH(CF3)OH, C(CF3)2OH, - (CH2)qOR, -(CH2)C1NR2 , -C(O)N(R)2, -NR2, -NRC(O)R, -S(O)mN(R)2, -OC(O)R, OC(O)N(R)2, -NRS(O) mR , -NRC(O)N(R)2, CN, halogen and -NO2, wherein each R is independently selected from H, C1-C6 alkyl, C3 - C10 cycloalkyl and a 5- to 12- membered aryl or heteroaryl group, the group being unsubstituted or substituted, m is 1 or 2 and q is O, 1 or 2;
(b) a heteroaryl group which contains 1, 2, 3 or 4 ring nitrogen atoms and O, 1 or 2 additional heteroatoms selected from O and S, which group is monocyclic or bicyclic and which is unsubstituted or substituted; and (c) a group comprising a benzene ring which is unsubstituted or substituted and which is fused to a heteroaryl group as defined above; or a pharmaceutically acceptable salt thereof; provided that R3 is not an indole group or an indazole group, which group is unsubstituted or substituted; or a pharmaceutically acceptable salt thereof.
2. A compound according to claim 1 wherein the fused pyrimidine is of formula (Ia):
Figure imgf000073_0001
(Ia) wherein X is O or S and R1, R2, R3 and n are as defined in claim 1.
3. A compound according to claim 1 wherein the fused pyrimidine is of formula (Ib):
Figure imgf000073_0002
(Ib)
wherein X is O or S and R1, R2, R3 and n are as defined in claim 1.
4. A compound according to claim 1 which is selected from:
3-(4-Morpholin-4-yl-6-moφholin-4-ylmethyl-thieno[3,2-d]pyrimidin-2-yl)-phenol; 3-(4-Morpholin-4-yl-6-pyrrolidin-l-ylmethyl-thieno[3,2-d]pyrimidin-2-yl)-phenol; 3-[6-(4-Methyl-piperazin-l-ylmethyl)-4- morpholin-4-yl-thieno[3,2-d]pyrimidin-
2-yl] -phenol;
3-[6-(6,7-Dimethoxy-3,4-dihydro-lH-isoquinolin-2-ylmethyl)-4-moφholin-4-yl- thieno[3,2-d]pyrimidin-2-yl]-phenol ; 3-(6-[ 1 ,4']Bipiperidinyl- 1 '-ylmethyl-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-2-yl)- phenol;
3-[4-Moφholin-4-yl-6-(4-pyrimidin-2-yl-piperazin-l-ylmethyl)-thieno[3,2- d]pyrimidin-2-yl]-phenol;
3-[6-(4-Cyclohexylmethyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol;
4- [2-(3 -Hydroxy-phenyl)-4-moφholin-4-yl-thieno [3 ,2-d]pyrimidin-6-ylmethyl] - piperazine-1-carboxylic acid tert-butyl ester;
3-(4-Moφholin-4-yl-6-piperazin-l-ylmethyl-thieno[3,2-d]pyrimidin-2-yl)-phenol , trifluoroacetic acid salt; 3-{6-[4-(2-Methoxy-ethyl)-piperazin-l-ylmethyl]-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-2-yl} -phenol;
3-[6-(4-Methyl-[l,4]diazepan-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-
2-yl]-phenol;
{4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin-1-yl} -acetic acid ethyl ester; l-{4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin- 1 -yl} -ethanone;
3-{6-[4-(2-Hydroxy-ethyl)-piperazin-l-ylmethyl]-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-2-yl} -phenol; 3 -(4-Moφholin-4-yl-6-thiomoφholin-4-ylmethyl-thieno [3 ,2-d]pyrimidin-2-yl)- phenol;
3-[6-(4-Ethyl-piperazin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-2-yl]- phenol;
3-(6- {[Methyl-(1 -methyl-piperidin-4-yl)-amino] -methyl} -4-moφholin-4-yl- thieno[3,2-d]pyrimidin-2-yl)-phenol;
3-[6-(4-Dimethylamino-piperidin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-2-yl] -phenol ; 4-[2-(3-Hydroxy-phenyl)-4-morpholin-4- yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin-2-one;
3-[6-(4-Methanesulfonyl-piperazin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol; {4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin- 1 -yl} -acetonitrile;
3-[4-Morpholin-4-yl-6-(4-moφholin-4-yl-piperidin-l-ylmethyl)-thieno[3,2- d]pyrimidin-2-yl]-phenol;
3- [6-(4- Amino-piperidin- 1 -ylmethyl)-4-morpholin-4-yl-thieno [3 ,2-d]pyrimidin-2- yl]-phenol;
3 -[6-(3 -Dimethylamino-pyrrolidin- 1 -ylmethyl)-4-morpholin-4-yl-thieno [3 ,2- d]pyrimidin-2-yl]-phenol;
1 - [2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno [3 ,2-d]pyrimidin-6-ylmethyl] -A- moφholin-4-ylmethyl-piperidin-4-ol; 4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazine-1-carboxylic acid ethylamide ;
N- { 1 -[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperidin-4-yl}-methanesulfonamide; l-{4-[2-(3-Hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin-l-yl}-2-methoxy-ethanone;
N- { 1 -[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperidin-4-yl}-acetamide;
4- [2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno [3 ,2-d]pyrimidin-6-ylmethyl] - piperazine-1-carboxylic acid dimethylamide; l-[2-(3 -Hydroxy-phenyl)-4-moφholin-4-yl-thieno [3 ,2-d]pyrimidin-6-ylmethyl] - piperidin-4-ol;
3-[6-( 1 , 1 -Dioxo- 1 lambda*6*-thiomoφholin-4-ylmethyl)-4-moφholin-4-yl- thieno[3,2-d]pyrimidin-2-yl]-phenol;
4-[2-(3 -Hydroxy-phenyl)-4-moφholin-4-yl-thieno [3 ,2-d]pyrimidin-6-ylmethyl] - piperazine-1 -sulfonic acid dimethylamide;
3-(6-{4-[(2-Methoxy-ethyl)-methyl-amino]-piperidin-l-ylmethyl}-4-moφholin-4-yl- thieno[3,2-d]pyrimidin-2-yl)-phenol; l-[2-(3-Hydroxy-phenyl)-4-morpholin-4- yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- pyrrolidin-3-ol;
(i?,iS)-3-[6-(2-Dimethylaminomethyl-pyiτolidin-l-ylmethyl)-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-2-yl]-phenol; (R,S)-3-(6- {[Methyl-(1 -methyl-pyrrolidin-2-ylmethyl)-amino]-methyl} -A- morpholin-4-yl-thieno[3,2-d]pyrimidin-2-yl)-phenol; l-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperidine-4-carboxylic acid amide;
2-{4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin- 1 -yl} -N,N-diniethyl-acetamide;
3-(6-{[Methyl-(l-methyl-pyrrolidin-3-yl)-amino]-methyl}-4-morpholin-4-yl- thieno[3,2-d]pyrimidin-2-yl)-phenol;
2-Dimethylamino-l-{4-[2-(3-hydroxy-phenyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-6-ylmethyl]-piperazin- 1 -yl} -ethanone; 3-[6-((3R,5S)-3,5-Dimethyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol;
3-[6-((3R,5S)-4-Methanesulfonyl-3,5-dimethyl-piperazin-l-ylmethyl)-4-moφholin-
4-yl-thieno[3,2-d]pyrimidin-2-yl]-phenol; l-{(2R,6S)-4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6- ylmethyl]-2,6-dimethyl-piperazin-l-yl} -ethanone;
3 - {6-[4-(3 -Dimethylamino-propane- 1 -sulfonyl)-piperazin- 1 -ylmethyl] -4-moφholin-
4-yl-thieno[3,2-d]pyrimidin-2-yl}-phenol;
3-[6-(4-Methoxy-piperidin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-2- yl]-phenol; 3-{4-Moφholin-4-yl-6-[4-(3-moφholin-4-yl-propane-l-sulfonyl)-piperazin-l- ylmethyl]-thieno[3,2-d]pyrimidin-2-yl}-phenol;
{4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazin- 1 -yl} -moφholin-4-yl-methanone;
4-[2-(3-Hydroxy-phenyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-6-ylmethyl]- piperazine-1-carboxylic acid (2-methoxy-ethyl)-methyl-amide; l-{3-[6-(4-Methyl-piperazin-l-ylmethyl)-4-moφholin-4-yl-thieno[3,2-d]pyrimidin-
2-yl]-phenyl}-ethanol; . 3-[6-(4-Methyl-piperazin-l-ylmethyl)-4- moφholin-4-yl-thieno[3,2-d]pyrimidin-
2-yl]-phenyl} -methanol;
2-Chloro-5-[6-(4-methyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl] -phenol ; 2,3-Difluoro-5-[6-(4-methyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol;
4-Fluoro-3-[6-(4-methyl-piperazin-l-ylmethyl)-4-morpholin-4-yl-thieno[3,2- d]pyrimidin-2-yl]-phenol;
2-( 1 H-Indazol-6-yl)-6-(4-methyl-piperazin- 1 -ylmethyl)-4-morpholin-4-yl- thieno[3,2-d] pyrimidin-2-yl] -phenol; and
6-(4-Methyl-piperazin- 1 -ylmethyl)-4-morpholin-4-yl-2-( 1 H-pyjazol-4-yl)- thieno[3,2-d]pyrimidine; and the pharmaceutically acceptable salts of the above-mentioned free compounds.
5. A process for producing a compound as defined in claim 1 wherein m is 1, which process comprises:
(a) treating a compound of formula (II):
Figure imgf000077_0001
whereinA and R2 are as defined in claim 1, with a boronic acid or ester thereof of formula R3B(OR15)2 , in which R3 is as defined above and each R15 is H or C1-C6 alkyl or the two groups OR15 form, together with the boron atom to which they are attached, a pinacolato boronate ester, in the presence of a Pd catalyst; and treating the resulting compound of formula (III):
Figure imgf000077_0002
wherein A, R >2 a „„nd j τ R>3 are as defined above, with an amine of formula
NHR4R5 wherein R4 and R5 are as defined in claim 1 , in the presence of a suitable reducing agent; or
(b) treating a compound of formula (II) as defined above with an amine of formula NHR4R5 wherein R4 and R5 are as defined above, in the presence of a suitable reducing agent; and treating the resulting compound of formula (IV):
Figure imgf000078_0001
wherein A, R2 , R4 and R5 are as defined above, with a boronic acid or ester thereof of formula R3B(OR15)2 , in which R3 is as defined above and each R15 is H or Ci-C6 alkyl or the two groups OR15 form, together with the boron atom to which they are attached, a pinacolato boronate ester group, in the presence of a Pd catalyst.
6. A process for producing a compound of formula (I) as defined in claim 1, in which m m i iss 11 a anndd R R33 i iss a a 33-- o orr 44--hhyyddrrooxxyypphheennyyll g grrooiup, which process comprises: (a) treating a compound of formula (V):
Figure imgf000078_0002
wherein OR' is bonded at position 3 or 4 of the phenyl ring to which it is attached, R' is a hydroxy protecting group and R2 is as defined in claim 1 , with an amine of formula NHR4R5 wherein R4 and R5 are as defined in claim 1, in the presence of a suitable reducing agent; and (b) removing the hydroxy protecting group.
7. A process for producing a compound as defined in claim 1 wherein m is 0, which process comprises treating a compound of formula (XIV):
Figure imgf000079_0001
wherein A, R2 and R3 are as defined in claim 1 and W is a halo group selected from Br and I, with an amine of formula NHR4R5 in which R4 and R5 are as defined in claim 1, in the presence of a palladium catalyst.
8. A process according to claim 5, 6 or 7 which further comprises converting the resulting compound of formula (I) into a pharmaceutically acceptable salt thereof.
9. A pharmaceutical composition which comprises a pharmaceutically acceptable carrier or diluent and, as an active ingredient, a compound as claimed in any one of claims 1 to 4.
10. A compound as defined in claim 1 for use in a method of treatment of the human or animal body by therapy.
11. Use of a compound as defined in claim 1 in the manufacture of a medicament for treating a disease or disorder arising from abnormal cell growth, function or behaviour associated with PB kinase.
12. Use according to claim 11 wherein the disease or disorder is selected from cancer, immune disorders, cardiovascular disease, viral infection, inflammation, metabolism/endocrine disorders and neurological disorders.
13. A method of treating a disease or disorder arising from abnormal cell growth, function or behaviour associated with PB kinase, which method comprises administering to a patient in need thereof a compound as defined in claim 1.
14. A method according to claim 13 wherein the disease or disorder is selected from the group consisting of cancer, immune disorders, cardiovascular disease, viral infection, inflammation, metabolism/endocrine disorders and neurological disorders.
PCT/GB2005/004146 2004-10-25 2005-10-25 Pharmaceutical compounds WO2006046040A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
EP05798278.7A EP1812446B1 (en) 2004-10-25 2005-10-25 Pharmaceutical compounds
ES05798278.7T ES2554689T3 (en) 2004-10-25 2005-10-25 Pharmaceutical compounds
JP2007537401A JP5077946B2 (en) 2004-10-25 2005-10-25 Pharmaceutical compounds
US11/893,625 US7872003B2 (en) 2004-10-25 2007-08-16 Pharmaceutical compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0423653.5A GB0423653D0 (en) 2004-10-25 2004-10-25 Pharmaceutical compounds
GB0423653.5 2004-10-25

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US11/893,625 Continuation US7872003B2 (en) 2004-10-25 2007-08-16 Pharmaceutical compounds

Publications (1)

Publication Number Publication Date
WO2006046040A1 true WO2006046040A1 (en) 2006-05-04

Family

ID=33485146

Family Applications (3)

Application Number Title Priority Date Filing Date
PCT/GB2005/004137 WO2006046035A1 (en) 2004-10-25 2005-10-25 Pharmaceutical compounds
PCT/GB2005/004129 WO2006046031A1 (en) 2004-10-25 2005-10-25 Pharmaceutical compounds
PCT/GB2005/004146 WO2006046040A1 (en) 2004-10-25 2005-10-25 Pharmaceutical compounds

Family Applications Before (2)

Application Number Title Priority Date Filing Date
PCT/GB2005/004137 WO2006046035A1 (en) 2004-10-25 2005-10-25 Pharmaceutical compounds
PCT/GB2005/004129 WO2006046031A1 (en) 2004-10-25 2005-10-25 Pharmaceutical compounds

Country Status (17)

Country Link
US (8) US7776856B2 (en)
EP (3) EP1812445B1 (en)
JP (3) JP5078016B2 (en)
KR (3) KR20070084474A (en)
CN (2) CN101087794B (en)
AU (2) AU2005298408B2 (en)
BR (2) BRPI0517461A (en)
CA (2) CA2585089C (en)
ES (3) ES2554689T3 (en)
GB (1) GB0423653D0 (en)
IL (2) IL182615A0 (en)
MX (2) MX2007004868A (en)
NO (2) NO20072116L (en)
NZ (2) NZ554585A (en)
RU (2) RU2422449C2 (en)
WO (3) WO2006046035A1 (en)
ZA (2) ZA200703312B (en)

Cited By (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008070740A1 (en) * 2006-12-07 2008-06-12 F.Hoffmann-La Roche Ag Phosphoinositide 3-kinase inhibitor compounds and methods of use
WO2008152394A1 (en) * 2007-06-12 2008-12-18 F.Hoffmann-La Roche Ag Pharmaceutical compounds
WO2009042607A1 (en) 2007-09-24 2009-04-02 Genentech, Inc. Thiazolopyrimidine p13k inhibitor compounds and methods of use
WO2009082687A1 (en) 2007-12-21 2009-07-02 Genentech, Inc. Azaindolizines and methods of use
JP2010509373A (en) * 2006-11-13 2010-03-25 イーライ リリー アンド カンパニー Thienopyrimidinone for the treatment of inflammatory diseases and cancer
JP2010512337A (en) * 2006-12-07 2010-04-22 ジェネンテック インコーポレイテッド Phosphoinositide 3-kinase inhibitor compounds and methods of use
US7750002B2 (en) 2004-10-25 2010-07-06 Piramed Limited Pharmaceutical compounds
KR20100085912A (en) * 2007-09-12 2010-07-29 제넨테크, 인크. Combinations of phosphoinositide 3-kinase inhibitor compounds and chemotherapeutic agents, and methods of use
WO2010091150A1 (en) 2009-02-05 2010-08-12 Immunogen, Inc. Novel benzodiazepine derivatives
US7781433B2 (en) 2006-04-26 2010-08-24 Piramed Limited Pharmaceutical compounds
WO2010105008A2 (en) 2009-03-12 2010-09-16 Genentech, Inc. Combinations of phosphoinositide 3-kinase inhibitor compounds and chemotherapeutic agents for the treatment of hematopoietic malignancies
WO2010110685A2 (en) 2009-03-27 2010-09-30 Pathway Therapeutics Limited Pyrimddinyl and 1,3,5-triazinyl benzimtoazole sulfonamides and their use in cancer therapy
US7846929B2 (en) 2006-04-26 2010-12-07 Genentech, Inc. Phosphoinositide 3-kinase inhibitor compounds and methods of use
JP2011500774A (en) * 2007-10-26 2011-01-06 エフ.ホフマン−ラ ロシュ アーゲー Thienopyrimidiene derivatives as PI3K inhibitors
WO2011036284A1 (en) 2009-09-28 2011-03-31 F. Hoffmann-La Roche Ag Benzoxepin pi3k inhibitor compounds and methods of use
WO2011049625A1 (en) 2009-10-20 2011-04-28 Mansour Samadpour Method for aflatoxin screening of products
WO2011089400A1 (en) 2010-01-22 2011-07-28 Centro Nacional De Investigaciones Oncológicas (Cnio) Inhibitors of pi3 kinase
JP2011522802A (en) * 2008-05-23 2011-08-04 ワイス・エルエルシー Triazine compounds as PI3 kinase and mTOR inhibitors
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
WO2011141713A1 (en) 2010-05-13 2011-11-17 Centro Nacional De Investigaciones Oncologicas (Cnio) New bicyclic compounds as pi3-k and mtor inhibitors
US8097622B2 (en) 2008-10-14 2012-01-17 Daiichi Sankyo Company, Limited Morpholinopurine derivatives
WO2012112708A1 (en) 2011-02-15 2012-08-23 Immunogen, Inc. Cytotoxic benzodiazepine derivatives and methods of preparation
WO2012151525A1 (en) 2011-05-04 2012-11-08 Rhizen Pharmaceuticals Sa Novel compounds as modulators of protein kinases
US8367663B2 (en) 2009-01-08 2013-02-05 Curis, Inc. Phosphoinositide 3-kinase inhibitors with a zinc binding moiety
WO2013174794A1 (en) 2012-05-23 2013-11-28 F. Hoffmann-La Roche Ag Compositions and methods of obtaining and using endoderm and hepatocyte cells
EP2690101A1 (en) 2007-12-19 2014-01-29 Genentech, Inc. 5-Anilinoimidazopyridines and Methods of Use
US8642607B2 (en) 2009-11-05 2014-02-04 Rhizen Pharmaceuticals Sa 4H-chromen-4-one compounds as modulators of protein kinases
WO2014031566A1 (en) 2012-08-22 2014-02-27 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US8710219B2 (en) 2011-04-01 2014-04-29 Curis, Inc. Phosphoinositide 3-kinase inhibitor with a zinc binding moiety
US8778935B2 (en) 2009-04-16 2014-07-15 Centro Nacional De Investigaciones Oncologicas (Cnio) Imidazopyrazines for use as kinase inhibitors
US8785651B2 (en) 2009-03-24 2014-07-22 Sumitomo Chemical Company, Limited Method for manufacturing a boronic acid ester compound
US8802670B2 (en) 2006-04-26 2014-08-12 F. Hoffmann-La Roche Ag Pharmaceutical compounds
WO2014134483A2 (en) 2013-02-28 2014-09-04 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
WO2014134486A2 (en) 2013-02-28 2014-09-04 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
WO2014177915A1 (en) 2013-05-01 2014-11-06 Piramal Enterprises Limited Cancer combination therapy using imidazo[4,5-c]quinoline derivatives
US8895729B2 (en) 2012-10-10 2014-11-25 Genentech, Inc. Process for making thienopyrimidine compounds
WO2014194030A2 (en) 2013-05-31 2014-12-04 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
WO2015027667A1 (en) 2013-08-30 2015-03-05 浙江医药股份有限公司新昌制药厂 2, 6-di-nitrogen-containing substituted purine derivative, and preparation method, pharmaceutical composition and use thereof
US9150579B2 (en) 2012-07-04 2015-10-06 Rhizen Pharmaceuticals Sa Selective PI3K delta inhibitors
WO2018045379A1 (en) 2016-09-02 2018-03-08 Dana-Farber Cancer Institute, Inc. Composition and methods of treating b cell disorders
US9993554B2 (en) 2010-10-21 2018-06-12 Centro Nacional De Investigaciones Oncologicas (Cnio) Use of P13K Inhibitors for the Treatment of Obesity, Steatosis and ageing
US10588894B2 (en) 2017-06-21 2020-03-17 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US10870657B2 (en) 2015-12-22 2020-12-22 SHY Therapeutics LLC Compounds for the treatment of cancer and inflammatory disease
US11234986B2 (en) 2018-09-11 2022-02-01 Curis, Inc. Combination therapy with a phosphoinositide 3-kinase inhibitor with a zinc binding moiety
US11661429B2 (en) 2019-02-06 2023-05-30 Venthera, Inc. Topical phosphoinositide 3-kinase inhibitors

Families Citing this family (95)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE448532T1 (en) * 1996-09-04 2009-11-15 Intertrust Tech Corp RELIABLE INFRASTRUCTURE SUPPORT SYSTEMS, METHODS AND TECHNIQUES FOR SECURE ELECTRONIC COMMERCIAL, ELECTRONIC TRANSACTIONS, TRADE PROCESS CONTROL AND AUTOMATION, DISTRIBUTED PROCESSING AND RIGHTS MANAGEMENT
PL2024372T3 (en) * 2006-04-26 2010-11-30 Hoffmann La Roche Thieno[3,2-d]pyrimidine derivative useful as pi3k inhibitor
WO2007122410A1 (en) * 2006-04-26 2007-11-01 F.Hoffmann-La Roche Ag Pyrimidine derivatives as pi3k inhibitors
GB0608820D0 (en) * 2006-05-04 2006-06-14 Piramed Ltd Pharmaceutical compounds
GB0611152D0 (en) 2006-06-06 2006-07-19 Ucb Sa Therapeutic agents
CN101500575A (en) * 2006-06-12 2009-08-05 沃泰克斯药物股份有限公司 Thienopyrimidines useful as modulators of ion channels
GB0612630D0 (en) 2006-06-26 2006-08-02 Novartis Ag Organic compounds
EP2158207B1 (en) * 2007-06-12 2011-05-25 F. Hoffmann-La Roche AG Thiazoliopyrimidines and their use as inhibitors of phosphatidylinositol-3 kinase
WO2009055730A1 (en) * 2007-10-25 2009-04-30 Genentech, Inc. Process for making thienopyrimidine compounds
CA2703138A1 (en) * 2007-10-26 2009-04-30 F. Hoffmann-La Roche Ag Purine derivatives useful as pi3 kinase inhibitors
WO2009085230A1 (en) 2007-12-19 2009-07-09 Amgen Inc. Inhibitors of pi3 kinase
WO2009094224A1 (en) 2008-01-25 2009-07-30 Millennium Pharmaceuticals, Inc. Thiophenes and their use as phosphatidylinositol 3-kinase (pi3k) inhibitors
EP2644204B1 (en) 2008-03-18 2017-04-19 Genentech, Inc. Combinations of an Anti-HER2 antibody-drug conjugate and pertuzumab
WO2009146406A1 (en) * 2008-05-30 2009-12-03 Genentech, Inc. Purine pi3k inhibitor compounds and methods of use
JP5599783B2 (en) 2008-05-30 2014-10-01 アムジエン・インコーポレーテツド Inhibitors of PI3 kinase
ES2432821T3 (en) * 2008-07-31 2013-12-05 Genentech, Inc. Pyrimidine compounds, compositions and methods of use
CA2750935A1 (en) 2009-01-30 2010-08-12 Millennium Pharmaceuticals, Inc. Heteroaryls and their use as pi3k inhibitors
US9090601B2 (en) 2009-01-30 2015-07-28 Millennium Pharmaceuticals, Inc. Thiazole derivatives
US8796314B2 (en) 2009-01-30 2014-08-05 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
CA2755285C (en) 2009-03-20 2014-02-11 Yunxin Y. Bo Inhibitors of pi3 kinase
EP2432321B1 (en) 2009-05-20 2017-06-14 Glaxosmithkline LLC Thiazolopyrimidinone derivatives as pi3 kinase inhibitors
WO2010136491A1 (en) * 2009-05-27 2010-12-02 F. Hoffmann-La Roche Ag Bicyclic indole-pyrimidine pi3k inhibitor compounds selective for p110 delta, and methods of use
BRPI1009022A2 (en) * 2009-05-27 2016-03-08 Hoffmann La Roche "compound, pharmaceutical composition, process for producing a pharmaceutical composition, use of a compound, method for treating a disease or disorder and kit"
US20100331305A1 (en) * 2009-06-24 2010-12-30 Genentech, Inc. Oxo-heterocycle fused pyrimidine compounds, compositions and methods of use
AU2010268058A1 (en) * 2009-07-02 2012-01-19 Novartis Ag 2-carboxamide cycloamino ureas useful as PI3K inhibitors
JP2013500257A (en) 2009-07-21 2013-01-07 ギリアード カリストガ エルエルシー Treatment of liver damage with PI3K inhibitors
MX2012002059A (en) 2009-08-20 2012-04-19 Karus Therapeutics Ltd Tricyclic heterocyclic compounds as phosphoinositide 3-kinase inhibitors.
SG10201405568UA (en) 2009-09-08 2014-11-27 Hoffmann La Roche 4-substituted pyridin-3-yl-carboxamide compounds and methods of use
US20110086837A1 (en) 2009-10-12 2011-04-14 Genentech, Inc. Combinations of a pi3k inhibitor and a mek inhibitor
US8288381B2 (en) * 2009-11-12 2012-10-16 Genentech, Inc. N-9 substituted purine compounds, compositions and methods of use
CA2780018C (en) * 2009-11-12 2015-10-20 F. Hoffmann-La Roche Ag N-7 substituted purine and pyrazolopyrimidine compounds, compositions and methods of use
WO2011080510A1 (en) 2009-12-31 2011-07-07 Centro Nacional De Investigaciones Oncológicas (Cnio) Tricyclic compounds for use as kinase inhibitors
UA110697C2 (en) 2010-02-03 2016-02-10 Сігнал Фармасьютікалз, Елелсі The use of inhibitors tor-kinase for the treatment of tumors in patients with reduced protein pampk and / or activity ampk
BR112012019635A2 (en) 2010-02-22 2016-05-03 Hoffmann La Roche pyrido [3,2-d] pyrimidine pi3k delta inhibitor compounds and methods of use
EP2558864A1 (en) 2010-04-16 2013-02-20 Genentech, Inc. Fox03a as predictive biomarker for pi3k/akt kinase pathway inhibitor efficacy
WO2012007493A1 (en) 2010-07-14 2012-01-19 F. Hoffmann-La Roche Ag Purine compounds selective for ρi3κ p110 delta, and methods of use
KR20130098334A (en) 2010-08-11 2013-09-04 밀레니엄 파머슈티컬스 인코퍼레이티드 Heteroaryls and uses thereof
EP2603216A4 (en) 2010-08-11 2013-12-18 Millennium Pharm Inc Heteroaryls and uses thereof
WO2012021611A1 (en) 2010-08-11 2012-02-16 Millennium Pharmaceuticals, Inc. Heteroaryls and uses thereof
TW201307309A (en) * 2010-10-13 2013-02-16 Millennium Pharm Inc Heteroaryls and uses thereof
SI2651951T1 (en) 2010-12-16 2015-01-30 F. Hoffmann-La Roche Ag Tricyclic pi3k inhibitor compounds and methods of use
WO2012088254A1 (en) 2010-12-22 2012-06-28 Genentech, Inc. Autophagy inducer and inhibitor combination therapy for the treatment of neoplasms
WO2012098387A1 (en) 2011-01-18 2012-07-26 Centro Nacional De Investigaciones Oncológicas (Cnio) 6, 7-ring-fused triazolo [4, 3 - b] pyridazine derivatives as pim inhibitors
CN103476767B (en) 2011-02-09 2015-06-10 弗·哈夫曼-拉罗切有限公司 Heterocyclic compounds as PI3 kinase inhibitors
BR112013024122A2 (en) 2011-03-21 2019-09-24 Hoffmann La Roche p110 delta pi3k selective benzoxazepine compounds and methods of use
CA2843887A1 (en) 2011-08-03 2013-02-07 Signal Pharmaceuticals, Llc Identification of gene expression profile as a predictive biomarker for lkb1 status
EP2573086A1 (en) * 2011-09-26 2013-03-27 AiCuris GmbH & Co. KG N-[5-(Aminosulfonyl)-4-methyl-1,3-thiazol-2-yl]-N-methyl-2-[4-(2-pyridinyl)phenyl]acetamide mesylate monohydrate
EP2573085A1 (en) 2011-09-26 2013-03-27 AiCuris GmbH & Co. KG N-[5-(aminosulfonyl)-4methyl-1,3-thiazol-2-yl]-N-methyl-2-[4-(2-pyridinyl)phenyl] acetamide mesylate monohydrate having a specific particle size distribution range and a specific surface area range
US20130096116A1 (en) 2011-10-13 2013-04-18 Genentech, Inc. Treatment of pharmacological-induced hypochlorhydria
HUE036052T2 (en) 2011-12-15 2018-06-28 Novartis Ag Use of inhibitors of the activity or function of pi3k
GB201204125D0 (en) 2012-03-08 2012-04-25 Karus Therapeutics Ltd Compounds
SG10201706196XA (en) 2012-06-08 2017-08-30 Hoffmann La Roche Mutant selectivity and combinations of a phosphoinositide 3 kinase inhibitor compound and chemotherapeutic agents for the treatment of cancer
AU2013203714B2 (en) 2012-10-18 2015-12-03 Signal Pharmaceuticals, Llc Inhibition of phosphorylation of PRAS40, GSK3-beta or P70S6K1 as a marker for TOR kinase inhibitory activity
CA2888861A1 (en) 2012-11-07 2014-05-15 Karus Therapeutics Ltd Novel histone deacetylase inhibitors and their use in therapy
EP2928488A1 (en) * 2012-12-07 2015-10-14 The General Hospital Corporation Combinations of a pi3k/akt inhibitor compound with an her3/egfr inhibitor compound and use thereof in the treatment of a hyperproliferative disorder
RU2015140573A (en) 2013-02-25 2017-03-30 Дженентек, Инк. METHODS AND COMPOSITIONS FOR DETECTION AND TREATMENT OF DRUG-RESISTANT MUTANT RESISTANT TO MEDICINES
EP2986322A1 (en) 2013-04-17 2016-02-24 Signal Pharmaceuticals, LLC Combination therapy comprising a tor kinase inhibitor and a 5-substituted quinazolinone compound for treating cancer
CA2908830C (en) 2013-04-17 2021-12-07 Signal Pharmaceuticals, Llc Treatment of cancer with dihydropyrazino-pyrazines
US9937169B2 (en) 2013-04-17 2018-04-10 Signal Pharmaceuticals, Llc Methods for treating cancer using dihydropyrazino-pyrazine compound combination therapy
PE20160041A1 (en) 2013-04-17 2016-01-28 Signal Pharm Llc PHARMACEUTICAL FORMULATIONS, PROCESS, SOLID FORMS AND METHODS OF USE RELATED TO 1-ETHYL-7- (2-METHYL-6- (1H-1,2,4-TRIAZOL-3-IL) PYRIDIN-3-IL) -3, 4-DIHYDROPIRAZINO [2,3-b] PIRAZIN-2 (1H) -ONE
JP6382948B2 (en) 2013-04-17 2018-08-29 シグナル ファーマシューティカルズ,エルエルシー Combination therapy comprising a TOR kinase inhibitor and a cytidine analog for the treatment of cancer
TW201521725A (en) 2013-04-17 2015-06-16 Signal Pharm Llc Methods for treating cancer using TOR kinase inhibitor combination therapy
TWI631950B (en) 2013-04-17 2018-08-11 標誌製藥公司 Treatment of cancer with dihydropyrazino-pyrazines
MX2015015450A (en) 2013-05-10 2016-03-17 Karus Therapeutics Ltd Novel histone deacetylase inhibitors.
DE102013008118A1 (en) 2013-05-11 2014-11-13 Merck Patent Gmbh Arylchinazoline
EP3003313A1 (en) 2013-05-29 2016-04-13 Signal Pharmaceuticals, LLC Pharmaceutical compositions of 7-(6-(2-hydroxypropan-2-yl)pyridin-3-yl)-1-((trans)-4-methoxycyclohexyl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1h)-one, a solid form thereof and methods of their use
KR101648141B1 (en) * 2013-06-11 2016-08-12 제일모직 주식회사 Compound, organic optoelectric device, and display device
CN104513254B (en) * 2013-09-30 2019-07-26 上海璎黎药业有限公司 Annelated pyrimidines class compound, intermediate, preparation method, composition and application
FR3015483B1 (en) * 2013-12-23 2016-01-01 Servier Lab NOVEL THIENOPYRIMIDINE DERIVATIVES, PROCESS FOR THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING SAME
GB201402431D0 (en) 2014-02-12 2014-03-26 Karus Therapeutics Ltd Compounds
AU2015228859A1 (en) 2014-03-13 2016-07-07 F. Hoffmann-La Roche Ag Therapeutic combinations with estrogen receptor modulators
JP2017511367A (en) 2014-04-16 2017-04-20 シグナル ファーマシューティカルズ,エルエルシー 1-ethyl-7- (2-methyl-6- (1H-1,2,4-triazol-3-yl) pyridin-3-yl) -3,4-dihydropyrazino [2,3-b] pyrazine-2 Solid forms comprising (1H) -one and coforms, compositions thereof and methods of use
NZ714742A (en) 2014-04-16 2017-04-28 Signal Pharm Llc Solid forms of 1-ethyl-7-(2-methyl-6-(1h-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1h)-one, compositions thereof and methods of their use
MA40271A (en) 2014-05-21 2017-03-29 Hoffmann La Roche Methods of treating pr-positive, luminal a breast cancer with pi3k inhibitor, pictilisib
GB201419228D0 (en) 2014-10-29 2014-12-10 Karus Therapeutics Ltd Compounds
GB201419264D0 (en) 2014-10-29 2014-12-10 Karus Therapeutics Ltd Compounds
AR104068A1 (en) 2015-03-26 2017-06-21 Hoffmann La Roche COMBINATIONS OF A 3-KINASE PHOSFOINOSYTIDE INHIBITOR COMPOSITE AND A CDK4 / 6 INHIBITOR COMPOUND FOR CANCER TREATMENT
JP6698712B2 (en) 2015-06-29 2020-05-27 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Treatment method using TASELISB
CN111848643A (en) 2015-07-02 2020-10-30 豪夫迈·罗氏有限公司 Benzoxazepine compounds and methods of use thereof
LT3317284T (en) 2015-07-02 2020-01-27 F. Hoffmann-La Roche Ag Benzoxazepin oxazolidinone compounds and methods of use
LT3325623T (en) 2015-07-23 2019-10-25 Inst Curie Use of a combination of dbait molecule and parp inhibitors to treat cancer
GB201514751D0 (en) 2015-08-19 2015-09-30 Karus Therapeutics Ltd Compounds
GB201514754D0 (en) 2015-08-19 2015-09-30 Karus Therapeutics Ltd Compounds
GB201514760D0 (en) 2015-08-19 2015-09-30 Karus Therapeutics Ltd Compounds and method of use
GB201514758D0 (en) 2015-08-19 2015-09-30 Karus Therapeutics Ltd Formulation
GB201519573D0 (en) 2015-11-05 2015-12-23 King S College London Combination
KR101919189B1 (en) * 2016-02-18 2018-11-15 경북대학교 산학협력단 Muti-substituted 4H-thiazolo[4,5-e][1,4]diazepine-5,8-dione derivatives and use thereof
WO2018162439A1 (en) 2017-03-08 2018-09-13 Onxeo New predictive biomarker for the sensitivity to a treatment of cancer with a dbait molecule
CN110996955A (en) 2017-06-22 2020-04-10 细胞基因公司 Treatment of hepatocellular carcinoma characterized by hepatitis B virus infection
AR114082A1 (en) * 2018-01-17 2020-07-22 Glaxosmithkline Ip Dev Ltd PI4KIIIb INHIBITORS
JP2021515580A (en) 2018-03-13 2021-06-24 オンクセオOnxeo DBAIT molecule against acquired resistance in the treatment of cancer
CN108997372B (en) * 2018-07-31 2020-07-28 华中科技大学同济医学院附属协和医院 Compound for positron imaging, intermediate thereof, preparation method and imaging agent
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
CN112920199B (en) * 2020-06-02 2023-02-03 四川大学 Piperazinone substituent or derivative thereof, preparation method and application thereof, and pharmaceutical composition
CN113045582B (en) * 2021-02-05 2022-12-23 中国药科大学 PARP-1/PI3K double-target inhibitor or pharmaceutically acceptable salt thereof, and preparation method and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4007187A (en) * 1973-10-02 1977-02-08 Delalande S.A. Furo (2,3d) pyrimidines
US6187777B1 (en) * 1998-02-06 2001-02-13 Amgen Inc. Compounds and methods which modulate feeding behavior and related diseases
EP1277738A1 (en) * 2000-04-27 2003-01-22 Yamanouchi Pharmaceutical Co. Ltd. Condensed heteroaryl derivatives
US20030220365A1 (en) * 1998-06-04 2003-11-27 Stewart Andrew O. Cell adhesion-inhibiting antiinflammatory compounds
WO2004017950A2 (en) * 2002-08-22 2004-03-04 Piramed Limited Phosphadidylinositol 3,5-biphosphate inhibitors as anti-viral agents

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1470356A1 (en) 1964-01-15 1970-04-30 Thomae Gmbh Dr K New thieno [3,2-d] pyrimidines and process for their preparation
BE754606A (en) 1969-08-08 1971-02-08 Thomae Gmbh Dr K NEW 2-AMINOALCOYLAMINO-THIENO (3,2-D) PYRIMIDINES AND THEIR MANUFACTURING PROCESSES
BE759493A (en) 1969-11-26 1971-05-25 Thomae Gmbh Dr K NEWS 2- (5-NITRO-2-FURYL) -THIENO (3,2-D) PYRIMIDINES AND METHODS FOR MAKING THEM
US3763156A (en) 1970-01-28 1973-10-02 Boehringer Sohn Ingelheim 2-heterocyclic amino-4-morpholinothieno(3,2-d)pyrimidines
RO62428A (en) 1971-05-04 1978-01-15 Thomae Gmbh Dr K PROCESS FOR THE PREPARATION OF THYENO- (3,2-D) -PYRIMIDINES
GB1570494A (en) 1975-11-28 1980-07-02 Ici Ltd Thienopyrimidine derivatives and their use as pesticides
US4196207A (en) 1977-05-23 1980-04-01 Ici Australia Limited Process for controlling eradicating or preventing infestations of animals by Ixodid ticks
CN1280580A (en) * 1997-11-11 2001-01-17 辉瑞产品公司 Thienopyrimidine and thienopyridine derivatives useful as anti-cancer agents
US6608053B2 (en) 2000-04-27 2003-08-19 Yamanouchi Pharmaceutical Co., Ltd. Fused heteroaryl derivatives
CL2003002287A1 (en) * 2002-11-25 2005-01-14 Wyeth Corp COMPOUNDS DERIVED FROM TIENO [3,2-b] -PIRIDINA-6-CARBONITRILOS AND TIENEO [2,3-b] -PIRIDINA-5-CARBONITRILS, PHARMACEUTICAL COMPOSITION, PROCEDURE OF PREPARATION AND INTERMEDIARY COMPOUNDS, AND THEIR USE IN THE TREATMENT OF CANCER, APOPLEJIA, OSTEOPOROSIS
ES2217956B1 (en) 2003-01-23 2006-04-01 Almirall Prodesfarma, S.A. NEW DERIVATIVES OF 4-AMINOTIENE (2,3-D) PIRIMIDIN-6-CARBONITRILE.
GB0423653D0 (en) 2004-10-25 2004-11-24 Piramed Ltd Pharmaceutical compounds
KR101422301B1 (en) * 2006-04-26 2014-07-30 에프. 호프만-라 로슈 아게 Pharmaceutical compounds
RU2439074C2 (en) 2006-04-26 2012-01-10 Ф. Хоффманн-Ля Рош Аг THIENO[3,2-d]PYRIMIDINE DERIVATIVE AS PHOSPHATIDYL INOSITOL-3-KINASE (PI3K) INHIBITOR
EP2205242B1 (en) * 2007-09-12 2015-04-15 Genentech, Inc. Combinations of phosphoinositide 3-kinase inhibitor compounds and chemotherapeutic agents, and methods of use

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4007187A (en) * 1973-10-02 1977-02-08 Delalande S.A. Furo (2,3d) pyrimidines
US6187777B1 (en) * 1998-02-06 2001-02-13 Amgen Inc. Compounds and methods which modulate feeding behavior and related diseases
US20030220365A1 (en) * 1998-06-04 2003-11-27 Stewart Andrew O. Cell adhesion-inhibiting antiinflammatory compounds
EP1277738A1 (en) * 2000-04-27 2003-01-22 Yamanouchi Pharmaceutical Co. Ltd. Condensed heteroaryl derivatives
WO2004017950A2 (en) * 2002-08-22 2004-03-04 Piramed Limited Phosphadidylinositol 3,5-biphosphate inhibitors as anti-viral agents

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
M. S. MANHAS, S. G. AMIN, B. DAYAL: "Heterocyclic compounds V. 2,4-Disubstituted Thienopyrimidones (1).", J. HETEROCYCL. CHEM., vol. 13, 1976, pages 633 - 638, XP002370920 *

Cited By (109)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7750002B2 (en) 2004-10-25 2010-07-06 Piramed Limited Pharmaceutical compounds
US8946217B2 (en) 2004-10-25 2015-02-03 F. Hoffmann-La Roche Ag Pharmaceutical compounds
US8153629B2 (en) 2004-10-25 2012-04-10 F. Hoffmann-La Roche Ag Pharmaceutical compounds
US8101607B2 (en) 2004-10-25 2012-01-24 F. Hoffman-La Roche Ag Pharmaceutical compounds
US7776856B2 (en) 2004-10-25 2010-08-17 F. Hoffman-La Roche Ag Pharmaceutical compounds
US9943519B2 (en) 2006-04-26 2018-04-17 Genentech, Inc. Pharmaceutical compounds
US20110105464A1 (en) * 2006-04-26 2011-05-05 Georgette Castanedo Phosphoinositide 3-kinase inhibitor compounds and methods of use
US8450315B2 (en) * 2006-04-26 2013-05-28 Genentech, Inc. Phosphoinositide 3-kinase inhibitor compounds and methods of use
US8685968B2 (en) 2006-04-26 2014-04-01 F. Hoffmann-La Roche Ag Pharmaceutical compounds
US8324206B2 (en) 2006-04-26 2012-12-04 F. Hoffmann-La Roche Ag Pharmaceutical compounds
US7781433B2 (en) 2006-04-26 2010-08-24 Piramed Limited Pharmaceutical compounds
US8802670B2 (en) 2006-04-26 2014-08-12 F. Hoffmann-La Roche Ag Pharmaceutical compounds
US8987260B2 (en) 2006-04-26 2015-03-24 F. Hoffmann-La Roche Ag Pharmaceutical compounds
US7846929B2 (en) 2006-04-26 2010-12-07 Genentech, Inc. Phosphoinositide 3-kinase inhibitor compounds and methods of use
JP2010509373A (en) * 2006-11-13 2010-03-25 イーライ リリー アンド カンパニー Thienopyrimidinone for the treatment of inflammatory diseases and cancer
EP2518074A1 (en) 2006-12-07 2012-10-31 F. Hoffmann-La Roche AG Phosphoinositide 3-kinase inhibitor compounds and methods of use
NO342697B1 (en) * 2006-12-07 2018-07-09 Hoffmann La Roche Phosphoinositide 3-kinase inhibitor compounds and methods of use
US7888352B2 (en) 2006-12-07 2011-02-15 Piramed Limited Phosphoinositide 3-kinase inhibitor compounds and methods of use
WO2008070740A1 (en) * 2006-12-07 2008-06-12 F.Hoffmann-La Roche Ag Phosphoinositide 3-kinase inhibitor compounds and methods of use
JP2010512338A (en) * 2006-12-07 2010-04-22 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Phosphoinositide 3-kinase inhibitor compounds and methods of use
US8383620B2 (en) 2006-12-07 2013-02-26 Genentech, Inc. Phosphoinositide 3-kinase inhibitor compounds and methods of use
TWI499420B (en) * 2006-12-07 2015-09-11 Hoffmann La Roche Phosphoinositide 3-kinase inhibitor compounds and methods of use
AU2007329352B2 (en) * 2006-12-07 2013-01-17 F. Hoffmann-La Roche Ag Phosphoinositide 3-kinase inhibitor compounds and methods of use
US8993563B2 (en) 2006-12-07 2015-03-31 Genentech, Inc. Phosphoinositide 3-kinase inhibitor compounds and methods of use
US9487533B2 (en) 2006-12-07 2016-11-08 Genentech, Inc. Phosphoinositide 3-kinase inhibitor compounds and methods of use
RU2468027C2 (en) * 2006-12-07 2012-11-27 Ф. Хоффманн-Ля Рош Аг Phosphoinositide-3-kinase inhibitors and methods for use thereof
JP2010512337A (en) * 2006-12-07 2010-04-22 ジェネンテック インコーポレイテッド Phosphoinositide 3-kinase inhibitor compounds and methods of use
KR101460816B1 (en) * 2006-12-07 2014-11-12 에프. 호프만-라 로슈 아게 Phosphoinositide 3-kinase inhibitor compounds and methods of use
WO2008152394A1 (en) * 2007-06-12 2008-12-18 F.Hoffmann-La Roche Ag Pharmaceutical compounds
KR20100085912A (en) * 2007-09-12 2010-07-29 제넨테크, 인크. Combinations of phosphoinositide 3-kinase inhibitor compounds and chemotherapeutic agents, and methods of use
KR101584823B1 (en) * 2007-09-12 2016-01-22 제넨테크, 인크. Combinations of phosphoinositide 3-kinase inhibitor compounds and chemotherapeutic agents, and methods of use
WO2009042607A1 (en) 2007-09-24 2009-04-02 Genentech, Inc. Thiazolopyrimidine p13k inhibitor compounds and methods of use
JP2010540458A (en) * 2007-09-24 2010-12-24 ジェネンテック, インコーポレイテッド Thiazolopyrimidine PI3K inhibitor compounds and methods of use
JP2011500774A (en) * 2007-10-26 2011-01-06 エフ.ホフマン−ラ ロシュ アーゲー Thienopyrimidiene derivatives as PI3K inhibitors
EP2690101A1 (en) 2007-12-19 2014-01-29 Genentech, Inc. 5-Anilinoimidazopyridines and Methods of Use
WO2009082687A1 (en) 2007-12-21 2009-07-02 Genentech, Inc. Azaindolizines and methods of use
JP2011522802A (en) * 2008-05-23 2011-08-04 ワイス・エルエルシー Triazine compounds as PI3 kinase and mTOR inhibitors
US8309546B2 (en) 2008-10-14 2012-11-13 Daiichi Sankyo Company, Limited Morpholinopurine derivatives
US8097622B2 (en) 2008-10-14 2012-01-17 Daiichi Sankyo Company, Limited Morpholinopurine derivatives
US8906909B2 (en) 2009-01-08 2014-12-09 Curis, Inc. Phosphoinositide 3-kinase inhibitors with a zinc binding moiety
US11261195B2 (en) 2009-01-08 2022-03-01 Curis, Inc. Phosphoinositide 3-kinase inhibitors with a zinc binding moiety
US8461157B2 (en) 2009-01-08 2013-06-11 Curis, Inc. Phosphoinositide 3-kinase inhibitors with a zinc binding moiety
US10894795B2 (en) 2009-01-08 2021-01-19 Curis, Inc. Phosphoinositide 3-kinase inhibitors with a zinc binding moiety
US11597732B2 (en) 2009-01-08 2023-03-07 Curis, Inc. Phosphoinositide 3-kinase inhibitors with a zinc binding moiety
US8367663B2 (en) 2009-01-08 2013-02-05 Curis, Inc. Phosphoinositide 3-kinase inhibitors with a zinc binding moiety
EP3360879A1 (en) 2009-02-05 2018-08-15 ImmunoGen, Inc. Benzodiazepine derivatives as cytotoxic agents
EP3100745A1 (en) 2009-02-05 2016-12-07 Immunogen, Inc. Novel benzodiazepine derivatives
WO2010091150A1 (en) 2009-02-05 2010-08-12 Immunogen, Inc. Novel benzodiazepine derivatives
WO2010105008A2 (en) 2009-03-12 2010-09-16 Genentech, Inc. Combinations of phosphoinositide 3-kinase inhibitor compounds and chemotherapeutic agents for the treatment of hematopoietic malignancies
US8785651B2 (en) 2009-03-24 2014-07-22 Sumitomo Chemical Company, Limited Method for manufacturing a boronic acid ester compound
WO2010110685A2 (en) 2009-03-27 2010-09-30 Pathway Therapeutics Limited Pyrimddinyl and 1,3,5-triazinyl benzimtoazole sulfonamides and their use in cancer therapy
US8778935B2 (en) 2009-04-16 2014-07-15 Centro Nacional De Investigaciones Oncologicas (Cnio) Imidazopyrazines for use as kinase inhibitors
EP2784078A1 (en) 2009-09-28 2014-10-01 F. Hoffmann-La Roche AG Benzoxepin pi3k inhibitor compounds and methods of use
WO2011036284A1 (en) 2009-09-28 2011-03-31 F. Hoffmann-La Roche Ag Benzoxepin pi3k inhibitor compounds and methods of use
WO2011049625A1 (en) 2009-10-20 2011-04-28 Mansour Samadpour Method for aflatoxin screening of products
US10538501B2 (en) 2009-11-05 2020-01-21 Rhizen Pharmaceuticals Sa Kinase modulators
EP3050876A2 (en) 2009-11-05 2016-08-03 Rhizen Pharmaceuticals S.A. Kinase modulators
US8642607B2 (en) 2009-11-05 2014-02-04 Rhizen Pharmaceuticals Sa 4H-chromen-4-one compounds as modulators of protein kinases
US11858907B2 (en) 2009-11-05 2024-01-02 Rhizen Pharmaceuticals Ag Kinase modulators
EP3444242A2 (en) 2009-11-05 2019-02-20 Rhizen Pharmaceuticals S.A. Novel benzopyran kinase modulators
US9421209B2 (en) 2009-11-05 2016-08-23 Rhizen Pharmaceuticals Sa Kinase modulators
US9018375B2 (en) 2009-11-05 2015-04-28 Rhizen Pharmaceuticals Sa Substituted chromenes as kinase modulators
US10442783B2 (en) 2009-11-05 2019-10-15 Rhizen Pharmaceuticals Sa 2,3-disubstituted chromen-4-one compounds as modulators of protein kinases
US9073927B2 (en) 2010-01-22 2015-07-07 Fundacion Centro Nacional De Investigaciones Oncologicas Carlos Iii Inhibitors of PI3 kinase
WO2011089400A1 (en) 2010-01-22 2011-07-28 Centro Nacional De Investigaciones Oncológicas (Cnio) Inhibitors of pi3 kinase
WO2011100403A1 (en) 2010-02-10 2011-08-18 Immunogen, Inc Cd20 antibodies and uses thereof
WO2011141713A1 (en) 2010-05-13 2011-11-17 Centro Nacional De Investigaciones Oncologicas (Cnio) New bicyclic compounds as pi3-k and mtor inhibitors
US9993554B2 (en) 2010-10-21 2018-06-12 Centro Nacional De Investigaciones Oncologicas (Cnio) Use of P13K Inhibitors for the Treatment of Obesity, Steatosis and ageing
EP3053600A1 (en) 2011-02-15 2016-08-10 ImmunoGen, Inc. Cytotoxic benzodiazepine derivatives
EP3666289A1 (en) 2011-02-15 2020-06-17 ImmunoGen, Inc. Cytotoxic benzodiazepine derivatives
WO2012128868A1 (en) 2011-02-15 2012-09-27 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
WO2012112708A1 (en) 2011-02-15 2012-08-23 Immunogen, Inc. Cytotoxic benzodiazepine derivatives and methods of preparation
US11135205B2 (en) 2011-04-01 2021-10-05 Curis, Inc. Phosphoinositide 3-kinase inhibitor with a zinc binding moiety
US10543197B2 (en) 2011-04-01 2020-01-28 Curis, Inc. Phosphoinositide 3-kinase inhibitor with a zinc binding moiety
US11654136B2 (en) 2011-04-01 2023-05-23 Curis, Inc. Phosphoinositide 3-kinase inhibitor with a zinc binding moiety
US8710219B2 (en) 2011-04-01 2014-04-29 Curis, Inc. Phosphoinositide 3-kinase inhibitor with a zinc binding moiety
US10220035B2 (en) 2011-05-04 2019-03-05 Rhizen Pharmaceuticals Sa Compounds as modulators of protein kinases
US10322130B2 (en) 2011-05-04 2019-06-18 Rhizen Pharmaceuticals Sa Substituted chromenones as modulators of protein kinases
US9775841B2 (en) 2011-05-04 2017-10-03 Rhizen Pharmaceuticals Sa Compounds as modulators of protein kinases
US11020399B2 (en) 2011-05-04 2021-06-01 Rhizen Pharmaceuticals Sa Intermediates useful in the synthesis of compounds as modulators of protein kinases
WO2012151525A1 (en) 2011-05-04 2012-11-08 Rhizen Pharmaceuticals Sa Novel compounds as modulators of protein kinases
WO2013174794A1 (en) 2012-05-23 2013-11-28 F. Hoffmann-La Roche Ag Compositions and methods of obtaining and using endoderm and hepatocyte cells
US10570142B2 (en) 2012-07-04 2020-02-25 Rhizen Pharmaceuticals Sa Selective PI3K delta inhibitors
US9669033B2 (en) 2012-07-04 2017-06-06 Rhizen Pharmaceuticals Sa Selective PI3K delta inhibitors
US10072013B2 (en) 2012-07-04 2018-09-11 Rhizen Pharmaceuticals Sa Selective PI3K delta inhibitors
US10981919B2 (en) 2012-07-04 2021-04-20 Rhizen Pharmaceuticals Sa Selective PI3K delta inhibitors
US9150579B2 (en) 2012-07-04 2015-10-06 Rhizen Pharmaceuticals Sa Selective PI3K delta inhibitors
US9475818B2 (en) 2012-07-04 2016-10-25 Rhizen Pharmaceuticals Sa Selective PI3K delta inhibitors
WO2014031566A1 (en) 2012-08-22 2014-02-27 Immunogen, Inc. Cytotoxic benzodiazepine derivatives
US8895729B2 (en) 2012-10-10 2014-11-25 Genentech, Inc. Process for making thienopyrimidine compounds
WO2014134483A2 (en) 2013-02-28 2014-09-04 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
EP3566750A2 (en) 2013-02-28 2019-11-13 ImmunoGen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
WO2014134486A2 (en) 2013-02-28 2014-09-04 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
WO2014177915A1 (en) 2013-05-01 2014-11-06 Piramal Enterprises Limited Cancer combination therapy using imidazo[4,5-c]quinoline derivatives
WO2014194030A2 (en) 2013-05-31 2014-12-04 Immunogen, Inc. Conjugates comprising cell-binding agents and cytotoxic agents
WO2015027667A1 (en) 2013-08-30 2015-03-05 浙江医药股份有限公司新昌制药厂 2, 6-di-nitrogen-containing substituted purine derivative, and preparation method, pharmaceutical composition and use thereof
US10870657B2 (en) 2015-12-22 2020-12-22 SHY Therapeutics LLC Compounds for the treatment of cancer and inflammatory disease
US11560390B2 (en) 2015-12-22 2023-01-24 SHY Therapeutics LLC Compounds for the treatment of cancer and inflammatory disease
WO2018045379A1 (en) 2016-09-02 2018-03-08 Dana-Farber Cancer Institute, Inc. Composition and methods of treating b cell disorders
US10870694B2 (en) 2016-09-02 2020-12-22 Dana Farber Cancer Institute, Inc. Composition and methods of treating B cell disorders
US10933054B2 (en) 2017-06-21 2021-03-02 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US11213515B1 (en) 2017-06-21 2022-01-04 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US11541041B1 (en) 2017-06-21 2023-01-03 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, Rasopathies, and fibrotic disease
US11026930B1 (en) 2017-06-21 2021-06-08 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US11000515B2 (en) 2017-06-21 2021-05-11 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US10940139B2 (en) 2017-06-21 2021-03-09 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US10588894B2 (en) 2017-06-21 2020-03-17 SHY Therapeutics LLC Compounds that interact with the Ras superfamily for the treatment of cancers, inflammatory diseases, rasopathies, and fibrotic disease
US11234986B2 (en) 2018-09-11 2022-02-01 Curis, Inc. Combination therapy with a phosphoinositide 3-kinase inhibitor with a zinc binding moiety
US11661429B2 (en) 2019-02-06 2023-05-30 Venthera, Inc. Topical phosphoinositide 3-kinase inhibitors

Also Published As

Publication number Publication date
KR20130065732A (en) 2013-06-19
IL182614A0 (en) 2007-07-24
CN101087795A (en) 2007-12-12
NO20072115L (en) 2007-07-24
US20090131429A1 (en) 2009-05-21
US7750002B2 (en) 2010-07-06
IL182614A (en) 2012-10-31
US20140294946A1 (en) 2014-10-02
BRPI0517461A (en) 2008-10-07
US7872003B2 (en) 2011-01-18
EP1812444A1 (en) 2007-08-01
US20120258966A1 (en) 2012-10-11
AU2005298404B2 (en) 2012-08-23
CN101087795B (en) 2011-07-27
RU2422448C2 (en) 2011-06-27
JP5078016B2 (en) 2012-11-21
US8153629B2 (en) 2012-04-10
US20080207611A1 (en) 2008-08-28
JP2008517893A (en) 2008-05-29
KR101466529B1 (en) 2014-11-27
JP5077946B2 (en) 2012-11-21
US20080207609A1 (en) 2008-08-28
MX2007004868A (en) 2007-07-20
NO20072116L (en) 2007-07-24
KR20070084474A (en) 2007-08-24
AU2005298404A1 (en) 2006-05-04
RU2007119374A (en) 2008-12-10
US8101607B2 (en) 2012-01-24
EP1812444B1 (en) 2015-08-26
ES2558867T3 (en) 2016-02-09
IL182615A0 (en) 2007-07-24
US8946217B2 (en) 2015-02-03
US20100234370A1 (en) 2010-09-16
JP2008517892A (en) 2008-05-29
RU2422449C2 (en) 2011-06-27
CA2585089A1 (en) 2006-05-04
ZA200703312B (en) 2008-09-25
WO2006046035A1 (en) 2006-05-04
ZA200703311B (en) 2008-06-25
EP1812446A1 (en) 2007-08-01
CA2585112C (en) 2013-06-11
KR20070097422A (en) 2007-10-04
ES2554689T3 (en) 2015-12-22
CN101087794B (en) 2011-05-18
CA2585112A1 (en) 2006-05-04
ES2547447T3 (en) 2015-10-06
RU2007119373A (en) 2008-12-10
EP1812446B1 (en) 2015-11-18
AU2005298408A1 (en) 2006-05-04
US20100280027A1 (en) 2010-11-04
JP2008517894A (en) 2008-05-29
NZ554585A (en) 2010-09-30
US20130317017A1 (en) 2013-11-28
CA2585089C (en) 2013-07-23
WO2006046031A1 (en) 2006-05-04
JP5048504B2 (en) 2012-10-17
AU2005298408B2 (en) 2012-08-23
US7776856B2 (en) 2010-08-17
WO2006046031A8 (en) 2007-06-07
CN101087794A (en) 2007-12-12
MX2007004867A (en) 2007-07-20
EP1812445A1 (en) 2007-08-01
GB0423653D0 (en) 2004-11-24
EP1812445B1 (en) 2015-12-30
NZ554586A (en) 2010-10-29
BRPI0517019A (en) 2008-09-30

Similar Documents

Publication Publication Date Title
EP1812446B1 (en) Pharmaceutical compounds
EP2032582A1 (en) Pyrimidine derivatives as pi3k inhibitors

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BW BY BZ CA CH CN CO CR CU CZ DK DM DZ EC EE EG ES FI GB GD GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV LY MD MG MK MN MW MX MZ NA NG NO NZ OM PG PH PL PT RO RU SC SD SG SK SL SM SY TJ TM TN TR TT TZ UG US UZ VC VN YU ZA ZM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): BW GH GM KE LS MW MZ NA SD SZ TZ UG ZM ZW AM AZ BY KG MD RU TJ TM AT BE BG CH CY DE DK EE ES FI FR GB GR HU IE IS IT LU LV MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2007537401

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2005798278

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2005798278

Country of ref document: EP