WO2012020227A1 - Tricyclic compounds for use as kinase inhibitors - Google Patents

Tricyclic compounds for use as kinase inhibitors Download PDF

Info

Publication number
WO2012020227A1
WO2012020227A1 PCT/GB2011/001202 GB2011001202W WO2012020227A1 WO 2012020227 A1 WO2012020227 A1 WO 2012020227A1 GB 2011001202 W GB2011001202 W GB 2011001202W WO 2012020227 A1 WO2012020227 A1 WO 2012020227A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
formula
compound
alkyl
substituents selected
Prior art date
Application number
PCT/GB2011/001202
Other languages
French (fr)
Inventor
Joaquin Pastor Fernandez
Guido Kurz
David Soilan Rodriguez
Maria Del Rosario Rico Ferreira
Miguel Angel Ortega Soret
Original Assignee
Centro Nacional De Investigaciones Oncologicas (Cnio)
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 Centro Nacional De Investigaciones Oncologicas (Cnio) filed Critical Centro Nacional De Investigaciones Oncologicas (Cnio)
Publication of WO2012020227A1 publication Critical patent/WO2012020227A1/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
    • C07D495/12Heterocyclic 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 three hetero rings
    • C07D495/14Ortho-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
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system

Definitions

  • This invention relates to novel pharmaceutically-useful compounds, which compounds are useful as inhibitors of protein or lipid kinases (such as inhibitors of the phosphoinositide 3 ⁇ kinase (PI3 kinase) family, particularly the PI3K class I sub-type.
  • the compounds may also be useful as inhibitors of the mammalian target of rapamycin (mTOR)).
  • mTOR rapamycin
  • the compounds are of potential utility in the treatment of diseases such as cancer.
  • the invention also relates to the use of such compounds as medicaments, to the use of such compounds for in vitro, in situ and in vivo diagnosis or treatment of mammalian cells (or associated pathological conditions), to pharmaceutical compositions containing them, and to synthetic routes for their production.
  • PKs protein kinases
  • a large share of the oncogenes and proto-oncogenes involved in human cancers code for PKs.
  • the enhanced activities of PKs are also implicated in many non-malignant diseases, such as benign prostate hyperplasia, familial adenomatosis, polyposis, neuro-fibromatosis, psoriasis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis glomerulonephritis and post-surgical stenosis and restenosis.
  • PKs are also implicated in inflammatory conditions and in the multiplication of viruses and parasites.
  • PKs may also play a major role in the pathogenesis and development of neurodegenerative disorders.
  • Current Opinion in Chemical Biology 1999, 3, 459 - 465 see, for instance, Current Opinion in Chemical Biology 1999, 3, 459 - 465.
  • Phosphatidylinositol 3-kinases are a family of lipid and serine/threonine kinases that catalyze the phosphorylation of the membrane lipid phosphatidylinositol (PI) on the 3'-OH of the inositol ring to produce l phosphoinositol-3-phosphate (PIP), phosphoinositol-3,4-diphosphate (PIP2) and phosphoinositol-3,4,5-triphosphate (PIP 3 ), which act as recruitment sites for various intracellular signalling proteins, which in turn form signalling complexes to relay extracellular signals to the cytoplasmic face of the plasma membrane.
  • PIP membrane lipid phosphatidylinositol
  • PIP2 phosphoinositol-3,4-diphosphate
  • PIP 3 phosphoinositol-3,4,5-triphosphate
  • PI3K isoforms categorized by their catalytic subunits, their regulation by corresponding regulatory subunits, expression patterns and signalling specific funtions ( ⁇ 110 ⁇ , ⁇ , ⁇ , ⁇ ) perform this enzymatic reaction (Exp. Cell. Res. 25 (1 ),. 239-54 (1999) by Vanhaesebroeck and Katso et al., 2001 , above).
  • the closely related isoforms p1 10a and ⁇ are ubiquitously expressed, while ⁇ and ⁇ are more specifically expressed in the haematopoietic cell system, smooth muscle cells, myocytes and endothelial cells (see e.g. Trends Biochem. Sci. 22 (7),. 267-72 (1997) by Vanhaesebroeck et al). Their expression might also be regulated in an inducible manner depending on the cellular, tissue type and stimuli as well as disease context. Inductibility of protein expression includes synthesis of protein as well as protein stabilization that is in part regulated by association with regulatory subunits. Eight mammalian PI3Ks have been identified so far, including four class I PI3Ks.
  • Class la includes ⁇ 3 ⁇ , ⁇ 3 ⁇ and PI3K5. All of the class la enzymes are heterodimeric complexes comprising a catalytic subunit ( ⁇ 1 10 , ⁇ 1 10 ⁇ or p1 106) associated with an SH2 domain containing p85 adapter subunit. Class la PI3Ks are activated through tyrosine kinase signalling and are involved in cell proliferation and survival. PI3Koc and ⁇ 3 ⁇ have also been implicated in tumorigenesis in a variety of human cancers. Thus, pharmacological inhibitors of PI3Ka and ⁇ 3 ⁇ are useful for treating various types of cancer.
  • ⁇ 3 ⁇ the only member of the Class lb PI3Ks, consists of a catalytic subunit ⁇ 110 ⁇ , which is associated with a p110 regulatory subunit.
  • ⁇ 3 ⁇ is regulated by G protein coupled receptors (GPCRs) via association with ⁇ subunits of heterotrimeric G proteins.
  • GPCRs G protein coupled receptors
  • ⁇ 3 ⁇ is expressed primarily in hematopoietic cells and cardiomyocytes and is involved in inflammation and mast cell function.
  • pharmacological inhibitors of ⁇ 3 ⁇ are useful for treating a variety of inflammatory diseases, allergies and cardiovascular diseases.
  • mTOR The mammalian target of rapamycin (mTOR) also known as FK506 binding protein 12-rapamycin associated protein 1 (FRAP1) is a protein which in humans is encoded by the FRAP1 gene.
  • mTOR is a serine/threonine protein kinase that regulates cell growth, cell proliferation, cell motility, cell survival, protein synthesis, and transcription.
  • the inhibition of mTORs are believed to be useful for treating various diseases/conditions, such as cancer (for example, as described in Easton et al. (2006). "mTOR and cancer therapy”. Oncogene 25 (48): 6436 ⁇ 6).
  • targeted therapies are becoming more important. That is, therapy that has the effect of interfering with specific target molecules that are linked to tumor growth and/or carcinogenesis. Such therapy may be more effective than current treatments (e.g. chemotherapy) and less harmful to normal cells (e.g. because chemotherapy has the potential to kill normal cells as well as cancerous cells).
  • current treatments e.g. chemotherapy
  • targeted therapies may be selective (i.e. it may inhibit a certain targeted molecule more selectively as compared to other molecular targets, e.g. as described hereinafter), may have the benefit of reducing side effects and may also have the benefit that certain specific cancers can be treated (also selectively). The latter may in turn also reduce side effects.
  • n 0, 1 or 2;
  • Ai, A 2 , A 3 and each A 4 independently represents -C(R 4 )R 5 -, -C(O)-, -0-, -S-, -S(O)- or -S(0) 2 -; the dotted lines represent the presence of an optional double bond, which may be present between A, and A 2 , A 2 and A 3 , A 3 and A (if the latter is present, i.e. when n does not represent 0) and/or between two A relie groups (if present, i.e.
  • R 3 represents aryl or heteroaryl (both of which are optionally substituted by one or more substituents selected from E 4 );
  • heteroatom(s) preferably selected from sulfur, oxygen and nitrogen
  • T 1 and T 2 independently represent a single bond, -N(R 10 *)- or -0-;
  • R 10a and R 1 a for example, when attached to the same atom
  • R 10d and R 11d , R 0i and R 1 i , R 10n and R 11n and R 10t and R 11t may be linked together to form (e.g. along with the requisite nitrogen atom to which they may be attached) a 4- to 20- (e.g.
  • each E 1 , E 2 , E 4 , E 5 , E 6 , E 7 , E 8 , E 10 , E 11 and E 12 independently represents, on each occasion when used herein:
  • R 60 , R 61 and R 62 independently represent hydrogen or C 1-6 alkyl optionally substituted by one or more fluoro atoms; or a pharmaceutically acceptable ester, amide, solvate or salt thereof, which compounds, esters, amides, solvates and salts are referred to hereinafter as "the compounds of the invention".
  • salts include acid addition salts and base addition salts.
  • Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of a compound of formula I with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of the invention in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.
  • esters and amides of the compounds of the invention are also included within the scope of the invention.
  • Pharmaceutically acceptable esters and amides of compounds of the invention may be formed from corresponding compounds that have an appropriate group, for example an acid group, converted to the appropriate ester or amide.
  • esters of carboxylic acids of compounds of the invention
  • pharmaceutically acceptable esters include optionally substituted Ci_ 6 alkyl, C 5-10 aryl and/or C 5 . 10 aryl-d-6 alkyl- esters.
  • Pharmaceutically acceptable amides of carboxylic acids of compounds of the invention
  • ⁇ 6 alkyl groups that may be mentioned in the context of such pharmaceutically acceptable esters and amides are not cyclic, e.g. linear and/or branched.
  • Further compounds of the invention that may be mentioned include carbamate, carboxamido or ureido derivatives, e.g. such derivatives of existing amino functional groups.
  • prodrug of a relevant compound of the invention includes any compound that, following oral or parenteral administration, is metabolised in vivo to form that compound in an experimentally-detectable amount, and within a predetermined time (e.g. within a dosing interval of between 6 and 24 hours (i.e. once to four times daily)).
  • parenteral administration includes all forms of administration other than oral administration.
  • Prodrugs of compounds of the invention may be prepared by modifying functional groups present on the compound in such a way that the modifications are cleaved, in vivo when such prodrug is administered to a mammalian subject. The modifications typically are achieved by synthesising the parent compound with a prodrug substituent.
  • Prodrugs include compounds of the invention wherein a hydroxyl, amino, sulfhydryl, carboxy or carbonyl group in a compound of the invention is bonded to any group that may be cleaved in vivo to regenerate the free hydroxyl, amino, sulfhydryl, carboxy or carbonyl group, respectively.
  • prodrugs include, but are not limited to, esters and carbamates of hydroxy functional groups, esters groups of carboxyl functional groups, N-acyl derivatives and N-Mannich bases. General information on prodrugs may be found e.g. in Bundegaard, H. "Design of Prodrugs” p. 1-92, Elesevier, New York-Oxford (1985).
  • Compounds of the invention may contain double bonds and may thus exist as E ⁇ entalle) and Z (zusammen) geometric isomers about each individual double bond. Positional isomers may also be embraced by the compounds of the invention. All such isomers (e.g. if a compound of the invention incorporates a double bond or a fused ring, the cis- and trans- forms, are embraced) and mixtures thereof are included within the scope of the invention (e.g. single positional isomers and mixtures of positional isomers may be included within the scope of the invention).
  • tautomer or tautomeric form
  • proton tautomers also known as prototropic tautomers
  • Valence tautomers include interconversions by reorganisation of some of the bonding electrons.
  • Compounds of the invention may also contain one or more asymmetric carbon atoms and may therefore exhibit optical and/or diastereoisomerism.
  • Diastereoisomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation.
  • the various stereoisomers may be isolated by separation of a racemic or other mixture of the compounds using conventional, e.g. fractional crystallisation or HPLC, techniques.
  • the desired optical isomers may be made by reaction of the appropriate optically active starting materials under conditions which will not cause racemisation or epimerisation (i.e.
  • a 'chiral pool' method by reaction of the appropriate starting material with a 'chiral auxiliary' which can subsequently be removed at a suitable stage, by derivatisation (i.e. a resolution, including a dynamic resolution), for example with a homochiral acid followed by separation of the diastereomeric derivatives by conventional means such as chromatography, or by reaction with an appropriate chiral reagent or chiral catalyst all under conditions known to the skilled person.
  • derivatisation i.e. a resolution, including a dynamic resolution
  • stereoisomers including but not limited to diastereoisomers, enantiomers and atropisomers
  • mixtures thereof e.g. racemic mixtures
  • stereoisomers are included within the scope of the invention.
  • all stereoisomers are contemplated and included as the compounds of the invention.
  • stereochemistry is specified by a solid wedge or dashed line representing a particular configuration, then that stereoisomer is so specified and defined.
  • the compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms.
  • the present invention also embraces isotopicaliy-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature (or the most abundant one found in nature). All isotopes of any particular atom or element as specified herein are contemplated within the scope of the compounds of the invention.
  • Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C , 13 N, 15 0, 17 0, 18 0, 32 P, 33 P, 35 S, 8 F, 36 CI, 23 l, and 25 l.
  • Certain isotopicaliy-labeled compounds of the present invention e.g., those labeled with 3 H and 1 C
  • Tritiated ( 3 H) and carbon-14 ( 1 C) isotopes are useful for their ease of preparation and detectability.
  • isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Scheme 1 and/or in the Examples herein below, by substituting an isotopically labeled reagent for a non- isotopically labeled reagent.
  • C 1-q alkyl groups (where q is the upper limit of the range) defined herein may be straight-chain or, when there is a sufficient number (i.e. a minimum of two or three, as appropriate) of carbon atoms, be branched- chain, and/or cyclic (so forming a C 3 . q -cycloalkyl group).
  • Such cycloalkyl groups may be monocyclic or bicyclic and may further be bridged. Further, when there is a sufficient number (i.e. a minimum of four) of carbon atoms, such groups may also be part cyclic.
  • Such alkyl groups may also be saturated or, when there is a sufficient number (i.e. a minimum of two) of carbon atoms, be unsaturated (forming, for example, a C 2 . q alkenyl or a C 2 . q alkynyl group).
  • C 1-q alkylene (where q is the upper limit of the range) defined herein may be straight-chain or, when there is a sufficient number of carbon atoms, be saturated or unsaturated (so forming, for example, an alkenylene or alkynylene linker group). However, such d. q alkylene groups may not be branched.
  • C 3 .q cycloalkyl groups (where q is the upper limit of the range) that may be specifically mentioned may be monocyclic or bicyclic alkyl groups, which cycloalkyl groups may further be bridged (so forming, for example, fused ring systems such as three fused cycloalkyl groups).
  • Such cycloalkyl groups may be saturated or unsaturated containing one or more double bonds (forming for example a cycloalkenyl group). Substituents may be attached at any point on the cycloalkyl group. Further, where there is a sufficient number (i.e. a minimum of four) such cycloalkyl groups may also be part cyclic.
  • heterocycloalkyl groups that may be mentioned include non-aromatic monocyclic and bicyclic heterocycloalkyl groups in which at least one (e.g. one to four) of the atoms in the ring system is other than carbon (i.e. a heteroatom), and in which the total number of atoms in the ring system is between 3 and 20 (e.g. between three and ten, e.g between 3 and 8, such as 5- to 8-). Such heterocycloalkyl groups may also be bridged.
  • heterocycloalkyl groups may be saturated or unsaturated containing one or more double and/or triple bonds, forming for example a C 2 .
  • q heterocyc!oalkenyl (where q is the upper limit of the range) group.
  • C 2 - q heterocycloalkyl groups that may be mentioned include 7- azabicyclo[2.2.1 ]heptanyl, 6-azabicyclo[3.1.1 ]heptanyl, 6-azabicyclo[3.2.1]- octanyl, 8-azabicyclo-[3.2.1]octanyl, aziridinyl, azetidinyl, dihydropyranyl, dihydropyridyl, dihydropyrrolyl (including 2,5-dihydropyrrolyl), dioxolanyl (including 1 ,3-dioxolanyl), dioxanyl (including 1 ,3-dioxanyl and 1 ,4-dioxanyl), dithianyl (including 1 ,4-dithianyl), dithiolanyl (including 1 ,3-dithiolanyl), imidazolidinyl, imidazolinyl, morpholinyl, 7-ox
  • heterocycloalkyl groups may, where appropriate, be located on any atom in the ring system including a heteroatom.
  • the point of attachment of heterocycloalkyl groups may be via any atom in the ring system including (where appropriate) a heteroatom (such as a nitrogen atom), or an atom on any fused carbocyclic ring that may be present as part of the ring system.
  • Heterocycloalkyl groups may also be in the N- or S- oxidised form.
  • Heterocycloalkyl mentioned herein may be stated to be specifically monocyclic or bicyclic.
  • bicyclic refers to groups in which the second ring of a two-ring system is formed between two adjacent atoms of the first ring.
  • bridged e.g. when employed in the context of cycloalkyl or heterocycloalkyl groups refers to monocyclic or bicyclic groups in which two non-adjacent atoms are linked by either an alkylene or heteroalkylene chain (as appropriate).
  • Aryl groups that may be mentioned include C 6 .2o, such as C 6 .12 (e.g. Ce- ⁇ ) aryl groups. Such groups may be monocyclic, bicyclic or tricyclic and have between 6 and 12 (e.g. 6 and 10) ring carbon atoms, in which at least one ring is aromatic.
  • C 6 . o aryl groups include phenyl, naphthyl and the like, such as 1 ,2,3,4-tetrahydro- naphthyl.
  • the point of attachment of aryl groups may be via any atom of the ring system. For example, when the aryl group is polycyclic the point of attachment may be via atom including an atom of a non-aromatic ring. However, when aryl groups are polycyclic (e.g. bicyclic or tricyclic), they are preferably linked to the rest of the molecule via an aromatic ring.
  • heteroaryl when used herein refers to an aromatic group containing one or more heteroatom(s) (e.g. one to four heteroatoms) preferably selected from N, O and S.
  • Heteroaryl groups include those which have between 5 and 20 members (e.g. between 5 and 10) and may be monocyclic, bicyclic or tricyclic, provided that at least one of the rings is aromatic (so forming, for example, a mono-, bi-, or tricyclic heteroaromatic group).
  • the heteroaryl group is polycyclic the point of attachment may be via atom including an atom of a non-aromatic ring.
  • heteroaryl groups are polycyclic (e.g.
  • bicyclic or tricyclic they are preferably linked to the rest of the molecule via an aromatic ring.
  • Heteroaryl groups that may be mentioned include 3,4-dihydro-1 H-isoquinolinyl, 1 ,3-dihydroisoindolyl, 1 ,3-dihydroisoindolyl (e.g. 3,4- dihydro-1/-/-isoquinolin-2-yl, ,3-dihydroisoindol-2-yl, 1 ,3-dihydroisoindol-2-yl; i.e.
  • heteroaryl groups that are linked via a non-aromatic ring or, preferably, acridinyl, benzimidazolyl, benzodioxanyl, benzodioxepinyl, benzodioxolyl (including 1 ,3- benzodioxolyl), benzofuranyl, benzofurazanyl, benzothiadiazolyl (including 2,1 ,3- benzothiadiazolyl), benzothiazolyl, benzoxadiazolyl (including 2,1 ,3- benzoxadiazolyl), benzoxazinyl (including 3,4-dihydro-2H-1 ,4-benzoxazinyl), benzoxazolyl, benzomorpholinyl, benzoselenadiazolyl (including 2, 1 ,3-benzoselenadiazolyl), benzothienyl, carbazolyl, chromanyl, cinnolinyl, furanyl, imid
  • heteroaryl groups may, where appropriate, be located on any atom in the ring system including a heteroatom.
  • the point of attachment of heteroaryl groups may be via any atom in the ring system including (where appropriate) a heteroatom (such as a nitrogen atom), or an atom on any fused carbocyclic ring that may be present as part of the ring system.
  • the heteroaryl group is monocyclic or bicyclic.
  • the heteroaryl may be consist of a five-, six- or seven-membered monocyclic ring (e.g. a monocyclic heteroaryl ring) fused with another a five-, six- or seven-membered ring (e.g. a monocyclic aryl or heteroaryl ring).
  • Heteroatoms that may be mentioned include phosphorus, silicon, boron and, preferably, oxygen, nitrogen and sulfur.
  • a group e.g. a C1.12 alkyl group
  • substituents e.g. selected from E 6
  • those substituents are independent of one another. That is, such groups may be substituted with the same substituent (e.g. defined by E 6 ) or different substituents (defined by E 6 ).
  • a 1 to A 4 -containing ring we mean the 5-, 6- or 7-membered ring containing the integers A,, A 2 , A 3 , and optionally, one or two A integers.
  • the dotted lines in the Ai to A ⁇ -containing ring represent the presence of an optional double bond.
  • the Ai to A 4 -containing ring may not be aromatic.
  • compounds of the invention that are the subject of this invention include those that are stable. That is, compounds of the invention include those that are sufficiently robust to survive isolation from e.g. a reaction mixture to a useful degree of purity.
  • Particularly preferred compounds of the invention include the following compounds of formula I (in which formulae IA and IB are the most preferred):
  • R 4 ' 5 represents hydrogen or one or more substituent(s) defined by R 4 or R 5 , which may be present on a carbon atom of the A, to A -containing ring (preferably these represent hydrogen and hence the AT to A 4 -containing ring is preferably unsubstituted on the carbon atoms);
  • R 5 is as defined herein, i.e. it may represent hydrogen or a substituent that is present on the nitrogen atom of the A to Vcontaining ring as defined herein;
  • R 3 most preferably represents pyrimidinyl (e.g. 5-pyrimidinyl) optionally substituted by one or more (e.g. two or, preferably, one) substituent(s) selected from E 4 (in which E 4 is as defined herein) and most preferably represents the following fragment:
  • R 3a , R 3b and R 3c represents a substituent and the other two (e.g. R 3a and R 3b ) represent hydrogen;
  • R 3a , R 3 and R 3c e.g. R 3c
  • E 4 preferably represents Q 4
  • Q 4 preferably represents -N(R 20 )R 21 , in which R 20 and R 21 each preferably represent hydrogen, so forming a -NH 2 group
  • R 3 preferably represents amino-pyrimidinyl, for instance 2-amino-5-pyrimidinyl (e.g. 2-NH 2 -5-pyrimidinyl), i.e. :
  • Preferred compounds of the invention that may be mentioned include those in which:
  • R 4 and R 5 independently represent, on each occasion when used herein, hydrogen, halo, -OR 10c , -N(R 10d )R 1d , -N(R 10e )-C(O)-R 0f , -C(O)R 10g , -C(O)OR 10h , -C(O)N(R 0i )R 1 1 i , -N(R 10i )-C(O)OR 10k , -N(R 10m )-C(O)-N(R 10n )R 1 ln ,
  • R 4 and R 5 are linked together as defined herein.
  • Further preferred compounds of the invention include those in which, for instance when n represents 0 or 1 (which is preferably the case):
  • A represents -C(R 4 )(R 5 )- or -C(O)-;
  • a 2 represents -C(R 4 )(R 5 )- or -N(R 6 )-;
  • a 3 represents -C(R )(R 5 )- or -N(R 6 )- (provided that both A 2 and A 3 do not represent -N(R 5 )-);
  • a 4 (if present) represents -C(R ⁇ ! )(R 5 )- or -C(O)-;
  • a 3 and A represent -C(R )(R 5 )-, then A 2 does not represent -C(O)- when ⁇ ⁇ represents -N(R 6 )-;
  • preferred Ai to A 4 -containing rings include those in which:
  • a 2 represents -N(R 6 )-
  • a 3 represents -C(R 4 )(R 5 )- and one of ⁇ ⁇ and A4 represents -C(O)- and the other represents -C(R 4 )(R 5 )-; or, preferably,
  • AT represents -C(R 4 )(R 5 )- or -C(O)-
  • a 2 and A 3 independently represent -N(R 6 )- or -C(R 4 )(R 5 )- and
  • a 4 represents -C(R 4 )(R 5 )- or -C(O)-;
  • a 2 represents -N(R 6 )- or -C(R )(R 5 )-, A-, and A 3 independently represent -C(R 4 )(R 5 )- or -C(O)-.
  • R 3 include, but are not limited to: pyrrole, pyrazole, triazole, tetrazole, thiazole, isothiazole, oxazole, isoxazole, isoindole, 1 ,3-dihydro- indol-2-one, pyridine-2-one, pyridine, pyridine-3-ol, imidazole, 1 H-indazole, 1 H- indole, indolin-2-one, 1-(indolin-1-yl)ethanone, pyrimidine, pyridazine, pyrazine and isatin groups, 1 H-benzo[d][1 ,2,3]triazole, 1 H-pyrazolo [3,4-b]pyridine, 1 H- pyrazolo[3,4-d]pyrimidine, 1 H-benzo[d]imidazole, 1 H-benzo[d]imidazol-2(3H)- one,
  • Preferred compounds of the invention include those in which:
  • R 3 represents aryl (e.g. phenyl), then that group may be unsubstituted but is preferably substituted by at least one (e.g. two or, preferably, one) substituent(s) selected from E";
  • R 3 represents monocyclic heteroaryl (e.g. a 5- or 6-membered heteroaryl group), then that group preferably contains 1 , 2, 3 or 4 nitrogen atoms and, optionally 1 or 2 additional heteroatoms selected from oxygen and sulfur, and which heteroaryl group is optionally substituted by one or more substituents selected from E 4 ;
  • R 3 represents bicyciic heteroaryl (e.g. a 8-, 9- or 10-membered heteroaryl group)
  • that group preferably consists of a 5- or 6-membered ring fused to another 5- or 6-membered ring (in which either one of those rings may contain one or more (e.g. four, or, preferably one to three) heteroatoms), in which the total number of heteroatoms is preferably one to four, and which ring is optionally substituted by one or more (e.g.
  • optional substituents are preferably selected from -OR, -SR, -CH 2 OR, C0 2 R, CF 2 OH, CH(CF 3 )OH, C(CF 3 ) 2 OH, -(CH 2 )wOR, -(CH 2 ) W NR 2 , -C(0)N(R) 2 , -NR 2 , -NRC(0)R, -NRC(0)NHR, -NRC(0)N(R) 2 , -S(0) y N(R) 2 , -OC(0)R, OC(0)N(R) 2 , -NRS(0) y R, -NRC(0)N(R) 2 , CN, halogen and -N0 2 (in which each R is independently selected from H, d-C 6 alkyl, C 3 -C 10 cycloalkyl and a 5- to 12-member
  • a first substituent as defined above and, optionally a further substituent; or a further two substituents) preferably selected from halo, d-12 alkyl, CN, N0 2 , OR d , SR d , NR d 2 , C(0)R d , SOR d , S0 2 R d , S0 2 N(R) 2 , NC(0)R d and C0 2 R d (wherein each R d is independently H or d-C 6 alkyl);
  • R 3 represents substituted aryl (e.g. phenyl)
  • the substituent may be situated at the 2-, 3-, 4-, 5- or 6- position of the phenyl ring (typically it is situated at position 3 or 4); particularly preferred are phenyl groups substituted by -OR d (in which R d is independently H or d-C 6 alkyl, e.g. methyl), e.g.
  • the -OR d group, or -OH group is typically situated at the 3- or 4- position of the phenyl ring, so forming a 3-hydroxyphenyl or 4-hydroxyphenyl group or an isostere thereof, 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 compounds of the invention; isosteres of 3-hydroxyphenyl and 4- hydroxyphenyl groups are encompassed within the definition of R 3 ;
  • R 3 when R 3 represents heteroaryl, it is unsubstituted or substituted (when substituted, it may be substituted by one or more substitutents selected from those listed in respect of substituents on R 3 , when R 3 is a phenyl group; typically, the substituents are selected from OH and NH 2 , or alkylated derivatives thereof).
  • E 1 , E 2 , E 4 , E 5 , E 6 , E 7 , E 8 , E 10 , E 11 or E 12 groups may be linked together, but are preferably not linked together;
  • aryl e.g. phenyl; preferably unsubstituted, but which may be substituted by one to three J 5 groups
  • Ci. 6 e.g. C ⁇ alkyl optionally
  • e alkyl optionally substituted by one or more fluoro atoms;
  • each R 50 , R 51 , R 52 and R 53 substituent independently represents, on each occasion when used herein, hydrogen or d. 6 (e.g. 0,. 3 ) alkyl optionally substituted by one or more substituents selected from fluoro;
  • R 60 , R 61 and R 62 independently represent hydrogen or C 1 (e.g. Ci. 2 ) alkyl optionally substituted by one or more fluoro atoms.
  • Preferred optional substituents on R 3 and the , to A4-containing ring include:
  • halo e.g. fluoro, chloro or bromo
  • alkyl which alkyl group may be cyclic, part-cyclic, unsaturated or, preferably, linear or branched (e.g. alkyl (such as ethyl, ?-propyl, isopropyl, /-butyl or, preferably, n-butyl or methyl), all of which are optionally substituted with one or more halo (e.g.
  • fluoro groups (so forming, for example, fluoromethyl, difluoromethyl or, preferably, trifluoromethyl) or substituted with an aryl, heteroaryl or heterocycloalkyi group (which themselves may be substituted with one or more -OR z , -CiOR 22 , -C(0)OR z3 , -N(R z4 )R zS , -S(0) 2 R z6 , -S(0) 2 N(R z7 )R z8 ;
  • aryl e.g. phenyl
  • substitutent is on an alkyl group, thereby forming e.g. a benzyl group
  • each R z1 to R z 2 independently represents, on each occasion when used herein, H or alkyl (e.g. ethyl, n-propyl, f-butyl or, preferably, n-butyl, methyl, isopropyl or cyclopropylmethyl (i.e. a part cyclic alkyl group)) optionally substituted by one or more halo (e.g. fluoro) groups (so forming e.g. a trifluoromethyl group).
  • any two R z groups e.g. R z4 and R z5 ), when attached to the same nitrogen heteroatom may also be linked together to form a ring such as one hereinbefore defined in respect of corresponding linkage of R 10a and R 11a groups.
  • Preferred compounds of the invention include those in which:
  • each R 50 , R 51 , R 52 and R 53 independently represents hydrogen or (e.g. Ci. 4 ) alkyl optionally substituted by one or more fluoro atoms;
  • each R 60 , R 61 and R 62 independently represents hydrogen or C 1-2 alkyl (e.g. methyl).
  • More preferred compounds of the invention include those in which:
  • each R 10a , R 1a , R 10c , R 10d , R l 1d , R 10e , R 10f , R 109 , R 10h , R 10i , R 1 i , R 0i , R 10k , R 0m , R ion ( R nn R iop R io q) ⁇ ⁇ ⁇ r Rl0ti ⁇ ⁇ « R iou and independentl represents hydrogen, (e.g. C 1-3 ) alkyl (optionally substituted by one or more substituents selected from 0 and E 10 , but which alkyl group is more preferably unsubstituted) or aryl (e.g. phenyl; which aryl group is optionally substituted by one or more E 1 substituents); or
  • each E 1 , E 2 , E 4 , E 5 , E 6 , E 7 , E 8 , E 10 , E 11 and E 2 independently represents a substituent selected from Q 4 , or (e.g.) E 4 may represent C -4 alkyl optionally substituted by one or more Q 5 substituents;
  • R 2Z represents C 1-3 alkyl or, preferably, hydrogen
  • each J 1 , J 2 , J 3 , J 4 , J 5 and J 6 independently represent a substituent selected from Q 7 , or J 1 to J 6 (e.g. J 4 ) represents (e.g. preferably unsubstituted) C 1-6 alkyl (e.g. C 3 .5 cycloalkyl);
  • Preferred ⁇ to / ⁇ -containing rings of the compounds of the invention include those of the following formulae: Particularly preferred rings include:
  • carbon atoms may be unsubstituted or substituted by a substituent defined by R 4 or R 5 , and R 6 is as hereinbefore defined.
  • Particularly preferred rings include:
  • R 3 groups of the compounds of the compounds of the invention include optionally substituted phenyl and pyrimidinyl (e.g. 5-pyrimidinyl), azaindolyl (e.g. azaindol-5-yl), indolyl (e.g. 5-indolyl or 4-indolyl) and pyridyl (e.g. 3-pyridyl).
  • Particularly preferred R 3 groups of compounds of the invention include optionally substituted phenyl and pyrimidinyl (e.g. 5-pyrimidinyl).
  • Preferred compounds of the invention include those in which:
  • R 3 represents aryl (e.g. phenyl) or heteroaryl (e.g. a 5- or 6-membered monocyclic heteroaryl group or a 9- or 10-membered bicyclic heteroaryl group; which groups may contain one to four, e.g 3 or, preferably, 1 or 2, heteroatoms preferably selected from nitrogen, oxygen and sulfur) both of which are optionally substituted by one or more (e.g. two, or, preferably, one) substituent(s) selected from E 4 (e.g. -CF 3 , -OH, -OCH 3 and/or -N(R 20 )R 21 (e.g. -NH 2 or -N(H)-CH 2 -cyclopropyl));
  • E 4 e.g. -CF 3 , -OH, -OCH 3 and/or -N(R 20 )R 21 (e.g. -NH 2 or -N(H)-CH 2 -cyclopropyl)
  • each R 4 and R 5 independently represent -C(O)N(R 0i )R 1 i (e.g. in which one of R 10 ' and R 11 ' is hydrogen and the other is as herein defined), or each R 4 and R 5 preferably (and independently) represent hydrogen, 0 1-6 alkyl (optionally substituted as defined herein; but preferably unsubstituted), -OR 10c or -C(O)OR 10h ;
  • R 4 and R 5 may be linked, but are more preferably not linked together;
  • R 4 or R 5 substituents there is two or, preferably one or none R 4 or R 5 substituents (i.e. that are not hydrogen) present in the A, to A 4 -containing ring (i.e. all the R 4 ' 5 substituents present, except two or preferably one, represent hydrogen);
  • each R 6 independently represents hydrogen, -C(O)R 10r , -C(O)OR 10s , -C(O)N(R 10l )R 11t , -S(O) 2 R 10u or C 1-6 (e.g. C ⁇ , such as methyl or butyl (e.g. s-butyl) or C 5 . 6 cycloalkyl, e.g. cyclohexyl) alkyl optionally substituted by one or more (e.g. two or, preferably, one) E 7 substituents;
  • C 1-6 e.g. C ⁇ , such as methyl or butyl (e.g. s-butyl) or C 5 . 6 cycloalkyl, e.g. cyclohexyl) alkyl optionally substituted by one or more (e.g. two or, preferably, one) E 7 substituents;
  • R 10c represents hydrogen
  • R 10s represents C 1-3 alkyl (e.g. ethyl);
  • R 101 represents hydrogen
  • en tiy represent heteroaryl or, preferably, aryl (e.g. phenyl; which aryl/heteroaryl group is/are optionally substituted by one or more E 11 substituent, so forming e.g. a fluorophenyl group) or d. 3 alkyl;
  • E 4 represents Q 4 (e.g. -OR 20 and/or -N(R 20 )R 21 ) or d. 6 (e.g. C v3> such as methyl) alkyl optionally substituted by one or more Q 5 substituents (e.g. fluoro, so forming for example a trifluoromethyl group);
  • E 7 and E 11 independently represent Ci- 6 (e.g. C 3-6 ) alkyl or Q 4 ;
  • heterocycloalkyl e.g. a 4- to 6-membered ring, containing preferably one heteroatom selected from nitrogen and oxygen
  • aryl e.g. phenyl; optionally substituted with two or, preferably, one substituent selected from J 3
  • heteroaryl e.g. a 5- or 6-membered mono
  • Q 5 represents -OR 20 , -N(R 20 )R 21 or, preferably, halo (e.g. fluoro);
  • R 20 and R 21 independently represent hydrogen, C 1-3 alkyl (e.g. methyl or ethyl), which latter group is optionally substituted by one or more (e.g. one) substituent(s) selected from J 4 ;
  • R 20 and R 21 when there is a -N(R 20 )R 21 moiety present, then one of R 20 and R 21 represents hydrogen, and the other represents hydrogen, alkyl (e.g. methyl or ethyl), which latter group is optionally substituted by one or more (e.g. one) substituent(s) selected from J 4 ;
  • J 3 represents Q 7 ;
  • J 4 represents C-,. 6 alkyl, such as C 3 . 6 alkyl (especially C 3 . 6 cycloalkyl, such as cyclopropyl);
  • Q 7 represents halo (e.g. fluoro) or -S(0) 2 R 5 °;
  • R 50 represents alkyl (e.g. methyl).
  • Preferred compounds of the invention include those in which:
  • R 3 represents aryl (e.g. phenyl) or heteroaryl (e.g. a 5- or 6-membered monocyclic heteroaryl group; which may contain one to four, e.g. 3 or, preferably, 1 or 2, heteroatoms preferably selected from nitrogen, oxygen and sulfur) both of which are optionally substituted by one or more (e.g. two, or, preferably, one) substituent(s) selected from E 4 (e.g. -CF 3 , preferably, -OH and/or -N(R 0 )R 21 (e.g. -NH 2 ));
  • E 4 e.g. -CF 3 , preferably, -OH and/or -N(R 0 )R 21 (e.g. -NH 2 )
  • each R 4 and R 5 independently represent hydrogen or 0,. 6 alkyl (optionally substituted as defined herein; but preferably unsubstituted); R 4 and R 5 may be linked, but are more preferably not linked together;
  • each R 6 independently represents hydrogen, -C(O)R 0r , -C(O)OR 10s , -C(O)N(R 10, )R 11t , -S(O) 2 R 0u or C 1-6 (e.g. C 1-4 ) alkyl (e.g. ethyl or methyl) optionally substituted by one or more (e.g. two or, preferably, one) E 7 substituents;
  • R 10r represents aryl (e.g. phenyl; which aryl group is optionally substituted by one or more E 11 substituent, so forming e.g. a fluorophenyl group) or CL 3 alkyl (e.g. methyl);
  • R 10s represents d -3 alkyl (e.g. isopropyl, methyl or, preferably, ethyl);
  • R 10t represents hydrogen
  • R 11t represents aryl (e.g. phenyl; which aryl group is optionally substituted by one or more E 11 substituent, so forming e.g. a fluorophenyl group) or C 3 alkyl (e.g. ethyl);
  • R 10u represents aryl (e.g. phenyl; which aryl group is optionally substituted by one or more E 11 substituent, so forming e.g. a fluorophenyl group) or CL 3 (e.g. C 1 .2) alkyl (e.g. ethyl or, preferablyl, methyl);
  • E 7 represents Q 4 ;
  • Q 4 preferably represents aryl (e.g. phenyl) optionally substituted by one or more substituents selected from J 3 (so forming e.g. a fluorophenyl group);
  • E 11 represents Q 4 ;
  • Q 4 represents halo (e.g. fluoro);
  • J 3 represents Q 7 , in which Q 7 preferably represents halo (e.g. fluoro);
  • E 4 represents alkyl (e.g. methyl; which alkyl group is optionally substituted by one or more substituents selected from Q 5 , in which Q 5 is preferably fluoro, so forming e.g. a -CF 3 group) or, more preferably, E 4 represents Q 4 ;
  • Q 4 represents alkyl (e.g. methyl; which alkyl group is optionally substituted by one or more substituents selected from J 2 , in which J 2 is preferably fluoro, so forming e.g. a -CF 3 group) or, more preferably, Q 4 represents halo, -OR 20 ,
  • Q 5 represents halo (e.g. fluoro);
  • J 2 represents halo (e.g. fluoro);
  • Y 0; R 20 and R 21 independently represent hydrogen or C 1 .3 alkyl (e.g. methyl or ethyl); Q 7 represents halo (e.g. fluoro).
  • Particularly preferred compounds of the invention include those in which:
  • R 3 represents hydroxyphenyl (e.g. 3-hydroxyphenyl) or pyrimidinyl (e.g. 5- pyrimidinyl, such as 2-amino-4-trifluoromethyl-5-pyrimidinyl (e.g. 2-NH 2 ,4-CF 3 - pyrimidin-5-yl) or, preferably, 2-amino-5-pyrimidinyl (i.e. 2-[-N(R 0 )(R 21 )]-pyrimidin- 5-yl such as 2-NH 2 -pyrimidin-5-yl));
  • pyrimidinyl e.g. 5- pyrimidinyl, such as 2-amino-4-trifluoromethyl-5-pyrimidinyl (e.g. 2-NH 2 ,4-CF 3 - pyrimidin-5-yl) or, preferably, 2-amino-5-pyrimidinyl (i.e. 2-[-N(R 0 )(R 21 )]-pyrimidin- 5-
  • A represents -C(R 4 )R 5 -;
  • a 2 and A 3 represents -N(R 6 )- and the other (preferably A 3 ) represents -C(R 4 )R 5 -;
  • n 0 or 1 ;
  • a 4 represents (if present) -C(R 4 )R 5 -;
  • the dotted lines do not represent the presence of an optional double bond (i.e. the ⁇ to A 4 -containing ring does not contain a double bond, other than that double bond that is integral to the requisite imidazopyrazine of formula I);
  • B 1 , B 1a , B 2 , B 2a , B 3 , B 3a , B 4 and B a independently represent hydrogen
  • each R 4 and R 5 independently represent hydrogen or C 1-3 alkyl (e.g. methyl); there is two or, preferably one or none R 4 or R 5 moieties present that do not represent hydrogen (e.g. there are two or, preferably, one that represent C v3 alkyl);
  • each R 6 independently represents -S(0) 2 -CH 2 CH 3 , -C(0)OCH 3 , -C(0)0-C(H)(CH 3 ) 2 , preferably, -C(0)-N(H)-[4-fluorophenyl], -S(0) 2 CH 3 , hydrogen or, more preferably, -C(0)OCH 2 CH 3 , -C(0)N(H)CH 2 CH 3 , -S(0) 2 -[4-fluorophenyl], -C(0)-[4-fluorophenyl], -C(0)CH 3 , ethyl or -CH 2 -[4- fluorophenyl].
  • Particularly preferred compounds of the invention include those of the examples described hereinafter.
  • L 1 represents a suitable leaving group, such as iodo, bromo, chloro or a sulfonate group (e.g. -OS(0) 2 CF 3 , -OS(0) 2 CH 3 or -OS(0) 2 PhMe), and A 1 , A 2 , A 3 , A 4 , n, the dotted lines, B 1 , B 1a , B 2 , B 2a , B 3 , B 3a , B 4 , B 4a and R 2 are as hereinbefore defined, with a compound of formula III,
  • L 2 represents a suitable group such as -B(OH) 2 , -B(OR w ) 2 or -Sn(R *) 3 , in which each R wx independently represents a d-e alkyl group, or, in the case of -B(OR wx ) 2 , the respective R"* groups may be linked together to form a 4- to 6- membered cyclic group (such as a 4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl group), thereby forming e.g.
  • a pinacolato boronate ester group (or L 2 may represent iodo, bromo or chloro, provided that L 1 and L 2 are mutually compatible) and R 3 is as hereinbefore defined.
  • the reaction may be performed, for example in the presence of a suitable catalyst system, e.g. a metal (or a salt or complex thereof) such as Pd, Cul, Pd/C, PdCI 2 , Pd(OAc) 2 , Pd(Ph 3 P) 2 CI 2 , Pd(Ph 3 P) 4 (i.e.
  • a suitable catalyst system e.g. a metal (or a salt or complex thereof) such as Pd, Cul, Pd/C, PdCI 2 , Pd(OAc) 2 , Pd(Ph 3 P) 2 CI 2 , Pd(Ph 3 P) 4 (i.e.
  • Pd 2 (dba) 3 and/or NiCI 2 palladium
  • Pd 2 (dba) 3 and/or NiCI 2 palladium
  • a ligand such as PdCI 2 (dppf).DCM, f-Bu 3 P, (C 6 Hn) 3 P, Ph 3 P, AsPh 3 , P(o-Tol) 3 , 1 ,2-bis(diphenylphosphino)ethane, 2,2'-bis(di-ferf-butyl- phosphino)-1 , 1 '-biphenyl, 2,2'-bis(diphenylphosphino)-1 , 1 '-bi-naphthyl, 1 ,1'- bis(diphenyl-phosphino-ferrocene), 1 ,3-bis(diphenylphosphino)propane, xantphos, or a mixture thereof
  • L 3 represents a suitable leaving group, such as one hereinbefore defined in respect of L 1 , and A 1 , A 2 , A 3 , A 4 , n, the dotted lines and R 3 are as hereinbefore defined, with a compound of formula V,
  • L 4 may represent hydrogen (so forming an amine group), and L , B ⁇ B 1a , B 2 , B 2a , B 3 , B 3a , B 4 and B 4a are as hereinbefore defined, and the reaction may be performed in the presence of an appropriate metal catalyst (or a salt or complex thereof) such as Cu, Cu(OAc) 2 , Cul (or Cul/diamine complex), copper tris(triphenylphosphine)bromide, Pd(OAc) 2 , tris(dibenzylideneacetone)- dipalladium(O) (Pd 2 (dba) 3 ) or NiCI 2 and an optional additive such as Ph 3 P, 2,2'- bis(diphenylphosphino)-1 ,1'-binaphthyl, xantphos, Nal or an appropriate crown ether such as 18-crown-6-benzene, in the presence of an appropriate base such as NaH, Et 3 N, pyridine,
  • the compound of formula IV (e.g. in which L 3 is chloro) may be prepared in situ, for example from a compound corresponding to a compound of formula IV, but in which L 3 represents -Od. 3 alkyl (e.g. methoxy) by reaction in the presence of e.g. a chlorinating agent (such as POCI 3 );
  • a 1 , A 2 , A 3 , A 4 , n, the dotted lines and R 3 are as hereinbefore defined, with a compound of formula V in which L 4 represents hydrogen (so forming optionally substituted morpholine), which reaction may proceed by the reaction initially with a reagent that may convert the oxo moiety into a leaving group (and hence form a compound of formula IV in situ), for instance para-toluenesulfonyl chloride (e.g.
  • (A x ) and (A y ) denotes the optional presence of the relevant AT to A 4 groups that are/may be present in the compound of formula I, and FG 1 and FG 2 independently represent mutually compatible functional groups, which may undergo an intramolecular reaction to form the requisite Ai to A4-containing ring of formula I (and L 1 R 3 represents R 3 or L 1 , and R 3 , L 1 , B 1 , B 1a , B 2 , B 2a , B 3 , B 3a , B 4 and B 4a are as hereinbefore defined; when L R 3 represents L 1 , then this step is followed by reaction with a compound of formula III as hereinbefore defined).
  • a ⁇ groups in the compound of formula I formed may be present either at the positions represented by (A x ) or (A y ) or may be an integral part of FG 1 and/or FG 2 .
  • mutually compatible functional groups FG 1 and FG 2
  • FG 1 and FG 2 we mean that such groups may be manipulated so as to promote an intramolecular reaction, for example, FG 1 may be -NH 2 and FG 2 may be -C(0)OH (or a derivative thereof; e.g. an ester), which functional groups may undergo an amide coupling reaction to form a -N(H)C(0)- linkage (and therefore a ring).
  • (A x ) is absent, the -N(H)-C(0)- linkage so formed represents the Ai and A 2 moieties (i.e. these emanate from an integral part of FG 1 and FG 2 ), and (A y ) represents A 3 and A 4 , which together are -CH2-CH2-.
  • FG 1 and FG 2 may independently represent leaving groups, such as those hereinbefore defined in respect of L 1 .
  • the compound of formula VII may be reacted with a nucleophile (such as one with more than one nucleophilic site e.g.
  • R 111 is as hereinbefore defined, for example, under reaction conditions known to those skilled in the art, such as those described herein, e.g. in the presence of a suitable base such as an amine base (e.g. diisopropylamine, diisopropylethylamine, or the like) and optionally in the presence of a suitable solvent (e.g. acetonitrile or the like);
  • a suitable base such as an amine base (e.g. diisopropylamine, diisopropylethylamine, or the like) and optionally in the presence of a suitable solvent (e.g. acetonitrile or the like);
  • G 1 -L 1b XI wherein G 1 represents either -C(O)R 10r or -S(O) 2 R 10u , and L (attached to the -C(O)- or -S(0) 2 moieties) represents a suitable leaving group such as iodo, bromo or, preferably, chloro, under reaction conditons known to those skilled in the art, for example at around room temperature or above in the presence of a suitable base (e.g. pyridine, triethylamine, dimethylaminopyridine, diisopropylamine, sodium hydroxide, or mixtures thereof), an appropriate solvent (e.g.
  • a suitable base e.g. pyridine, triethylamine, dimethylaminopyridine, diisopropylamine, sodium hydroxide, or mixtures thereof
  • an appropriate solvent e.g.
  • (x) for compounds of formula I in which n represents 1 or 2, in which a (or the) A4 moiety adjacent to the requisite bicycle represents -C(O)-, A, represents -C(R )(R 5 )- and A 2 represents -N(R 6 )- (and preferably n represents 1 , Ai and A 3 represent -C(R 4 )(R 5 )- and A 2 represents -N(R 6 )- in which R 6 is preferably hydrogen) may be prepared by reaction of a compound of formula XIA,
  • (A 4 ) denotes the optional presence of a further A4 group
  • L R 3 , B 1 , B 1a , B 2 , B 2a , B 3 , B 3a , B 4 and B 4a , A 3 , A 4 and R 6 are as hereinbefore defined, with a compound of formula XIB,
  • R -C(0)-R 5 XIB wherein R 4 and R 5 are as hereinbefore defined, for instance under Pictet- Spengler reaction conditions (e.g. as described in Bioorg. Med. Chem. 2008 (16), 542-559 (e.g. in the presence of a source of H + ), optionally followed by, if necessary (i.e. for reaction with compounds in which L 1 R 3 represents L 1 ), reaction with a compound of formula III as hereinbefore defined.
  • a 1 , A 2 , A 3 , A 4 , n and the dotted lines are as hereinbefore defined, with the appropriate sulfonyl chloride (e.g. para-toluene sulfonyl chloride) (e.g. in the presence of an appropriate amine, such as an organic amine base e.g. triethylamine, in an appropriate solvent, e.g. dichloromethane, optionally in the presence of catalytic DMAP), which is followed by the addition of a compound of formula V in which L 4 represents hydrogen (e.g. optionally substituted morpholine).
  • an appropriate amine such as an organic amine base e.g. triethylamine
  • an appropriate solvent e.g. dichloromethane
  • catalytic DMAP catalytic DMAP
  • R** represents hydrogen or is preferably other than hydrogen (so forming an ester) for instance an optionally substituted C n .i 2 alkyl group (e.g. methyl), and A 1 , A 2 , A 3 , A 4 , n and the dotted lines are as hereinbefore defined, with a compound of formula XIV, R 3 -CsN XIV wherein R 3 is as hereinbefore defined, i.e. optionally substituted aryl or heteroaryl (and -CN represents a substituent on a carbon atom of that aryl or heteroaryl ring), for example in the presence of a suitable solvent system (e.g. dioxane), acid (e.g.
  • a suitable solvent system e.g. dioxane)
  • acid e.g.
  • 4M HCI under pressure (e.g. reaction in a pressurised sealed tube).
  • the sealed tube may be left in an ultrasonic bath for a periodof time (at elevated temperature, e.g. at about 130°C), and the reaction mixture may then be evaporated, the residue taken up in dry toluene, treated with base (e.g. triethylamine or the like), and heated at reflux for a period of time, after which a further different solvent may be added (e.g. diethyl ether) and the desired product may precipitate out.
  • base e.g. triethylamine or the like
  • L 7 represents a suitable leaving group (such as one hereinbefore defined by L 1 , e.g. chloro) and (At), L 1 R 3 , B 1 , B 1a , B 2 , B 2a , B 3 , B 3a , B 4 and B 4a , A 3 and A4 are as hereinbefore defined, under standard amination conditions (e.g. in the presence of an amine H 2 NR 6 , and optionally a reagent that promotes the substitution).
  • a suitable leaving group such as one hereinbefore defined by L 1 , e.g. chloro
  • a 3 and A4 are as hereinbefore defined, under standard amination conditions (e.g. in the presence of an amine H 2 NR 6 , and optionally a reagent that promotes the substitution).
  • Compounds of formula XII may be prepared by intramolecular reaction of a compound of formula XV, wherein R xx (preferably methyl), A 1 , A 2 , A 3 , A 4 , n and the dotted lines are as hereinbefore defined, under standard conditions, for instance in the presence of basic conditions (e.g. in the presence of KOH, in an appropriate solvent such as methanol), which reaction mixture may be heated at reflux followed by quench by the addition of HCI.
  • R xx preferably methyl
  • a 1 , A 2 , A 3 , A 4 , n and the dotted lines are as hereinbefore defined, under standard conditions, for instance in the presence of basic conditions (e.g. in the presence of KOH, in an appropriate solvent such as methanol), which reaction mixture may be heated at reflux followed by quench by the addition of HCI.
  • Compounds of formula XIII (in particular those in which ⁇ and A (if present) represents -C(R 4 )R 5 -, one of A 2 and A 3 (preferably A 3 ) represents -C(R 4 )R 5 - and the other (e.g. A 2 ) represents -C(R 4 )R 5 - or -N(R 6 )-; preferably A 3 represents -C(R 4 )R 5 - and A 2 represents -N(R 6 )-) may be prepared by reaction of a compound of formula XVI,
  • a stereoisomer e.g. tautomer, such as the keto tautomer
  • another suitable derivative thereof e.g. an alkylated derivative, where there is e.g. an alkoxy group (e.g. -OCH 3 ) present in place of the hydroxy group
  • a 1 , A 2 , A 3 , A 4 , n and the dotted lines are as hereinbefore defined (and preferably A, and A 4 (if present) represents -C(R 4 )R 5 -, one of A 2 and A 3 (preferably A 3 ) represents -C(R 4 )R 5 - and the other (e.g.
  • a 2 represents -C(R 4 )R 5 - or -N(R 6 )-; but, may also represent -C(O)-), with a compound of formula XVII, wherein R xx is as hereinbefore defined, for example the compound of formula XVI may first be subjected to conditions to convert the hydroxy moiety to a suitable leaving group, for instance reaction conditions include the presence of a base (e.g. an organic amine base, such as triethylamine or the like), a suitable solvent (such as dichloromethane) and a reagent that is suitable for the conversion (e.g.
  • a base e.g. an organic amine base, such as triethylamine or the like
  • a suitable solvent such as dichloromethane
  • a reagent that is suitable for the conversion
  • a sulfonyl choride to convert the -OH to a sulfonate, for instance mesyl chloride to convert to -0-S(0) 2 -CH 3
  • reaction with the compound of formula XVII for instance in the presence of a suitable base (e.g. an alkoxide, such as sodium methoxide).
  • a suitable base e.g. an alkoxide, such as sodium methoxide.
  • Other compounds of formula XVI that may be mentioned include those in which n represents 1 , A-i , A 2 and , each represent -C(R 4 )R 5 - (e.g. in which R 4 and R 5 represent hydrogen) and A 3 represents -N(R 6 )-, in which R 6 may represent -C(0)-0-alkyl (e.g.
  • n represents 1 , and preferably A, represents -C(O)-, and A 2 , A 3 and A 4 independently represent -C(R 4 )(R 5 )- (which compounds are preferably prepared for instance by reaction in accordance with the procedures described in Akhrem et al.
  • Still further compounds of formula XVI that may be mentioned include those in which n represents 2, A ⁇ A 2 and A 3 represent -C(R )R 5 -, the A4 adjacent A 3 represents -N(R 6 )- and the second A represents -C(O)-, which compounds may be prepared in accordance with the techniques described in e.g. R. G. Glushkov and T. V. Stezhko, Chemistry of Heterocyclic Compounds 1978; 14(9): 1013-1016 (Reaction of 2,3-dioxo-4-(/V,/V- dimethylaminomethylene)hexahydroazepine with hydroxylamine).
  • Compounds of formula XIVA may be prepared by reaction of a compound of XVIIA, wherein L 1 R 3 , B 1 , B 1a , B 2 , B 2a , B 3 , B 3a , B 4 and B a are as hereinbefore defined, with a compound of formula XVIIB,
  • Compounds of formula XV may be prepared from corresponding compounds of formula XIII as hereinbefore defined, in the presence of a reagent to convert the amino group to a urea, e.g. chlorosulfonyl isocyanate or the like.
  • a reagent to convert the amino group to a urea e.g. chlorosulfonyl isocyanate or the like.
  • oxidations for example of a moiety containing an alcohol group (e.g. -CH 2 OH) to an aldehyde (e.g. -C(O)H), for example in the presence of a suitable oxidising agent, e.g. n0 2 or the like;
  • a suitable oxidising agent e.g. n0 2 or the like;
  • reductive amination of an aldehyde and an amine under appropriate reaction conditions, for example in "one-pot” procedure in the presence of an appropriate reducing agent, such as a chemoselective reducing agent such as sodium cyanoborohydride or, preferably, sodium triacetoxyborohydride, or the like.
  • an appropriate reducing agent such as a chemoselective reducing agent such as sodium cyanoborohydride or, preferably, sodium triacetoxyborohydride, or the like.
  • such reactions may be performed in two steps, for example a condensation step (in the presence of e.g. a dehydrating agent such as trimethyl orthoformate or MgS0 4 or molecular sieves, etc) followed by a reduction step (e.g. by reaction in the presence of a reducing agent such as a chemoselective one mentioned above or NaBH 4 , AIH , or the like), for instance the conversion of -NH 2 to -N(H)-isopropyl by condensation in the presence of acetone (H 3 C-C(0)-CH 3 ) followed by reduction in the presence of a reducing agent such as sodium cyanaoborohydride (i.e. overall a reductive amination);
  • a condensation step in the presence of e.g. a dehydrating agent such as trimethyl orthoformate or MgS0 4 or molecular sieves, etc
  • a reduction step e.g. by reaction in the presence of a reducing agent such as a che
  • amide coupling reactions i.e. the formation of an amide from a carboxylic acid (or ester thereof), for example when R 2 represents -C(0)OH (or an ester thereof), it may be converted to a -C(O)N(R 10b )R l 1 group (in which R 10b1 and R 11b1 are as hereinbefore defined, and may be linked together, e.g. as defined above), and which reaction may (e.g. when R 2 represents -C(O)OH) be performed in the presence of a suitable coupling reagent (e.g.
  • R 2 represents an ester (e.g. -C(0)OCH 3 or -C(0)OCH 2 CH 3 ), in the presence of e.g.
  • the -C(0)OH group may first be activated to the corresponding acyl halide (e.g -C(0)CI, by treatment with oxalyl chloride, thionyl chloride, phosphorous pentachloride, phosphorous oxychloride, or the like), and, in all cases, the relevant compound is reacted with a compound of formula HN(R 10a )R 11a (in which R 10a and R 1a are as hereinbefore defined), under standard conditions known to those skilled in the art (e.g. optionally in the presence of a suitable solvent, suitable base and/or in an inert atmosphere);
  • acyl halide e.g -C(0)CI, by treatment with oxalyl chloride, thionyl chloride, phosphorous pentachloride, phosphorous oxychloride, or the like
  • nucleophilic substitution reactions where any nucleophile replaces a leaving group, e.g. methylsulfonylpiperazine may replace a chloro leaving group;
  • alkyl-halide e.g. ethylbromide
  • alkyl-C(0)-halide e.g. H 3 C-C(0)CI
  • an anhydride e.g. H 3 C-C(0)-0-C(0)-CH 3 , i.e. "-0-C(0)-CH 3 " is the leaving group
  • dimethylformamide i.e. -N(CH 3 ) 2 is the leaving group
  • a sulfonyl halide e.g. H 3 C-S(0) 2 CI
  • a urea functional group by reaction of an amine (e.g. a secondary amine, such as a -NH moiety that is a part of a heterocyclic group) with an alkyl isocyanate (e.g. ethyl isocyanate) to form a -N-C(0)-N(H)-alkyl (e.g. -N-C(0)-N(H)-CH 2 CH 3 moiety), which transformation may be performed in the presence of a suitable solvent (e.g. acetonitrile) and base (e.g. N,N- diisopropylethylamine);
  • a suitable solvent e.g. acetonitrile
  • base e.g. N,N- diisopropylethylamine
  • a -N-(-C(0)0-ethyl) moiety to a -N(H) moiety, which nitrogen atom may be an integral part of a heterocycloalkyl ring system
  • a base e.g. lithium hydroxide hydrate/monohydrate
  • a suitable solvent e.g. a mixture of methanol/isopropanol
  • an electrophile that provides a source of iodide ions includes iodine, diiodoethane, diiodotetrachloroethane or, preferably, A/-iodosuccinimide, a source of bromide ions includes A -bromosuccinimide and bromine, and a source of chloride ions includes V-chlorosuccinimide, chlorine and iodine monochloride, for instance in the presence of a suitable solvent, such as CHCI 3 or an alcohol (e.g. methanol), optionally in the presence of a suitable base, such as a weak inorganic base, e.g. sodium bicarbonate.
  • the reaction maybe performed by heating at a convenient temperature, either by conventional heating under reflux or under microwave irradiation;
  • the substituents R 3 , B , B 1a , B 2 , B 2a , B 3 , B 3a , B 4 , B 4a , A 1 t A 2 , A 3 and (A ⁇ in final compounds of the invention or relevant intermediates may be modified one or more times, after or during the processes described above by way of methods that are well known to those skilled in the art. Examples of such methods include substitutions, reductions, oxidations, alkylations, acylations, hydrolyses, esterifications, etherifications, halogenations or nitrations. Such reactions may result in the formation of a symmetric or asymmetric final compound of the invention or intermediate.
  • the precursor groups can be changed to a different such group, or to the groups defined in formula I, at any time during the reaction sequence.
  • substituents in the compounds of the invention e.g. represented by R 3 , B 1 , B 1a , B 2 , B 2a , B 3 , B 3a , B 4 , B 4a , A 1 t A 2 , A 3 and (A 4 ) n
  • substituents in the compounds of the invention e.g. represented by R 3 , B 1 , B 1a , B 2 , B 2a , B 3 , B 3a , B 4 , B 4a , A 1 t A 2 , A 3 and (A 4 ) n
  • these groups can be further derivatized to other fragments described (e.g. by those integers mentioned above) in compounds of the invention, following synthetic protocols very well know to the person skilled in the art and/or according to the experimental part described in the patent.
  • transformation steps include: the reduction of a nitro or azido group to an amino group; the hydrolysis of a nitrile group to a carboxylic acid group; and standard nucleophilic aromatic substitution reactions, for example in which an iodo-, preferably, fluoro- or bromo-phenyl group is converted into a cyanophenyl group by employing a source of cyanide ions (e.g. by reaction with a compound which is a source of cyano anions, e.g. sodium, copper (I), zinc or potassium cyanide, optionally in the presence of a palladium catalyst) as a reagent (alternatively, in this case, palladium catalysed cyanation reaction conditions may also be employed).
  • a source of cyanide ions e.g. by reaction with a compound which is a source of cyano anions, e.g. sodium, copper (I), zinc or potassium cyanide, optionally in the presence of a palladium catalyst
  • transformations that may be mentioned include: the conversion of a halo group (preferably iodo or bromo) to a 1 -alkynyl group (e.g. by reaction with a 1 - alkyne), which latter reaction may be performed in the presence of a suitable coupling catalyst (e.g. a palladium and/or a copper based catalyst) and a suitable base (e.g.
  • a suitable coupling catalyst e.g. a palladium and/or a copper based catalyst
  • a suitable base e.g.
  • a tri-(Ci.6 alkyl)amine such as triethylamine, tributylamine or ethyldiisopropylamine
  • introduction of amino groups and hydroxy groups in accordance with standard conditions using reagents known to those skilled in the art; the conversion of an amino group to a halo, azido or a cyano group, for example via diazotisation (e.g. generated in situ by reaction with NaN0 2 and a strong acid, such as HCI or H 2 S0 4 , at low temperature such as at 0°C or below, e.g. at about -5°C) followed by reaction with the appropriate nucleophile e.g.
  • diazotisation e.g. generated in situ by reaction with NaN0 2 and a strong acid, such as HCI or H 2 S0 4 , at low temperature such as at 0°C or below, e.g. at about -5°C
  • a source of the relevant anions for example by reaction in the presence of a halogen gas (e.g. bromine, iodine or chlorine), or a reagent that is a source of azido or cyanide anions, such as NaN 3 or NaCN; the conversion of -C(0)OH to a -NH 2 group, under Schmidt reaction conditions, or variants thereof, for example in the presence of HN 3 (which may be formed in by contacting NaN 3 with a strong acid such as H 2 S0 4 ), or, for variants, by reaction with diphenyl phosphoryl azide ((PhO) 2 P(0)N 3 ) in the presence of an alcohol, such as tert-butanol, which may result in the formation of a carbamate intermediate; the conversion of -C(0)NH 2 to -NH 2 , for example under Hofmann rearrangement reaction conditions, for example in the presence of NaOBr (which may be formed by contacting NaOH and Br 2 ) which may result in the formation of a
  • Compounds of the invention bearing a carboxyester functional group may be converted into a variety of derivatives according to methods well known in the art to convert carboxyester groups into carboxamides, N-substituted carboxamides, ⁇ , ⁇ -disubstituted carboxamides, carboxylic acids, and the like.
  • the operative conditions are those widely known in the art and may comprise, for instance in the conversion of a carboxyester group into a carboxamide group, the reaction with ammonia or ammonium hydroxide in the presence of a suitable solvent such as a lower alcohol, dimethylformamide or a mixture thereof; preferably the reaction is carried out with ammonium hydroxide in a methanol/dimethyl- formamide mixture, at a temperature ranging from about 50°C to about 100°C.
  • Analogous operative conditions apply in the preparation of N-substituted or N,N- disubstituted carboxamides wherein a suitable primary or secondary amine is used in place of ammonia or ammonium hydroxide.
  • carboxyester groups may be converted into carboxylic acid derivatives through basic or acidic hydrolysis conditions, widely known in the art.
  • amino derivatives of compounds of the invention may easily be converted into the corresponding carbamate, carboxamido or ureido derivatives.
  • Compounds of the invention may be isolated from their reaction mixtures using conventional techniques (e.g. recrystallisations).
  • Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz), 9-fluorenylmethyleneoxycarbonyl (Fmoc) and 2,4,4-trimethylpentan-2-yl (which may be deprotected by reaction in the presence of an acid, e.g. HCI in water/alcohol (e.g. MeOH)) or the like.
  • an acid e.g. HCI in water/alcohol (e.g. MeOH)
  • protection and deprotection of functional groups may take place before or after a reaction in the above-mentioned schemes.
  • Protecting groups may be removed in accordance with techniques that are well known to those skilled in the art and as described hereinafter.
  • protected compounds/intermediates described herein may be converted chemically to unprotected compounds using standard deprotection techniques.
  • the type of chemistry involved will dictate the need, and type, of protecting groups as well as the sequence for accomplishing the synthesis.
  • Compounds of the invention may inhibit protein or lipid kinases, such as a PI3 kinase (especially a class I PI3K), for example as may be shown in the tests described below (for example, the test for PI3Ka inhibition described below) and/or in tests known to the skilled person.
  • the compounds of the invention may also inhibit mTOR.
  • the compounds of the invention may be useful in the treatment of those disorders in an individual in which the inhibition of such protein or lipid kinases (e.g. PI3 , particularly class I PI3K, and/or mTOR) is desired and/or required (for instance compounds of the invention may inhibit PI3K, particularly class I PI3K and, optionally, may also inhibit mTOR).
  • inhibitor may refer to any measurable reduction and/or prevention of catalytic kinase (e.g. PI3K, particularly class I PI3K, and/or mTOR) activity.
  • the reduction and/or prevention of kinase activity may be measured by comparing the kinase activity in a sample containing a compound of the invention and an equivalent sample of kinase (e.g. PI3K, particularly class I PI3K, and/or mTOR) in the absence of a compound of the invention, as would be apparent to those skilled in the art.
  • the measurable change may be objective (e.g.
  • Compounds of the invention may be found to exhibit 50% inhibition of a protein or lipid kinase (e.g. PI3K, such as class I PI3K, and/or mTOR) at a concentration of 100 ⁇ or below (for example at a concentration of below 50 ⁇ , or even below 10 ⁇ , such as below 1 ⁇ ), when tested in an assay (or other test), for example as described hereinafter, or otherwise another suitable assay or test known to the skilled person.
  • PI3K protein or lipid kinase
  • a protein or lipid kinase e.g. PI3K, such as class I PI3K, and/or mTOR
  • PI3K protein or lipid kinase
  • mTOR a protein or lipid kinase
  • compounds of the invention are expected to be useful in the treatment of a disease/disorder arising from abnormal cell growth, function or behaviour associated with the protein or lipid kinase (e.g. PI3 , such as class I PI3K, and/or mTOR).
  • Such conditions/disorders include cancer, immune disorders, cardiovascular diseases, viral infections, inflammation, metabolism/endocrine function disorders and neurological disorders.
  • the disorders/conditions that the compounds of the invention may be useful in treating hence includes cancer (such as lymphomas, solid tumours or a cancer as described hereinafter), obstructive airways diseases, allergic diseases, inflammatory diseases (such as asthma, allergy and Chrohn's disease), immunosuppression (such as transplantation rejection and autoimmune diseases), disorders commonly connected with organ transplantation, AIDS- related diseases and other associated diseases.
  • Other associated diseases that may be mentioned (particularly due to the key role of kinases in the regulation of cellular proliferation) include other cell proliferative disorders and/or non- malignant diseases, such as benign prostate hyperplasia, familial adenomatosis, polyposis, neurofibromatosis, psoriasis, bone disorders, atherosclerosis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis glomerulonephritis and post-surgical stenosis and restenosis.
  • non- malignant diseases such as benign prostate hyperplasia, familial adenomatosis, polyposis, neurofibromatosis, psoriasis, bone disorders, atherosclerosis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis glomerulonephritis and post-surgical stenosis and restenosis.
  • cardiovascular disease cardiovascular disease, stroke, diabetes, hepatomegaly, Alzheimer's disease, cystic fibrosis, hormone- related diseases, immunodeficiency disorders, destructive bone disorders, infectious diseases, conditions associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukaemia, liver disease, pathologic immune conditions involving T cell activation and CNS disorders.
  • cardiovascular disease stroke, diabetes, hepatomegaly, Alzheimer's disease, cystic fibrosis, hormone- related diseases, immunodeficiency disorders, destructive bone disorders, infectious diseases, conditions associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukaemia, liver disease, pathologic immune conditions involving T cell activation and CNS disorders.
  • the compounds of the invention may be useful in the treatment of cancer.
  • the compounds of the invention may therefore be useful in the treatment of a variety of cancer including, but not limited to: carcinoma such as cancer of the bladder, breast, colon, kidney, liver, lung (including non-small cell cancer and small cell lung cancer), esophagus, gall- bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, skin, squamous cell carcinoma, testis, genitourinary tract, larynx, glioblastoma, neuroblastoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, non-small cell lung carcinoma, small cell lung carcinoma, lung adenocarcinoma, bone, adenoma, adenocarcinoma, follicular carcinoma, undifferentiated carcinoma, papilliary carcinoma, seminona, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells
  • carcinoma
  • protein or lipid kinases may also be implicated in the multiplication of viruses and parasites. They may also play a major role in the pathogenesis and development of neurodegenerative disorders. Hence, compounds of the invention may also be useful in the treatment of viral conditions, parasitic conditions, as well as neurodegenerative disorders.
  • a method of treatment of a disease which is associated with the inhibition of protein or lipid kinase (e.g. PI3K, such as class I PI3K, and/or mTOR) is desired and/or required (for example, a method of treatment of a disease/disorder arising from abnormal cell growth, function or behaviour associated with protein or lipid kinases, e.g. PI3K, such as class I PI3K, and/or mTOR), which method comprises administration of a therapeutically effective amount of a compound of the invention, as hereinbefore defined, to a patient suffering from, or susceptible to, such a condition.
  • a disease e.g. cancer or another disease as mentioned herein
  • PI3K protein or lipid kinase
  • PI3K protein or lipid kinase
  • PI3K protein or lipid kinase
  • PI3K protein or lipid kinase
  • PI3K protein or lipid kinas
  • “Patients” include mammalian (including human) patients. Hence, the method of treatment discussed above may include the treatment of a human or animal body.
  • the term "effective amount” refers to an amount of a compound, which confers a therapeutic effect on the treated patient. The effect may be objective (e.g. measurable by some test or marker) or subjective (e.g. the subject gives an indication of or feels an effect).
  • Compounds of the invention may be administered orally, intravenously, subcutaneously, buccally, rectally, dermally, nasally, tracheally, bronchially, sublingually, by any other parenteral route or via inhalation, in a pharmaceutically acceptable dosage form.
  • Compounds of the invention may be administered alone, but are preferably administered by way of known pharmaceutical formulations, including tablets, capsules or elixirs for oral administration, suppositories for rectal administration, sterile solutions or suspensions for parenteral or intramuscular administration, and the like.
  • the type of pharmaceutical formulation may be selected with due regard to the intended route of administration and standard pharmaceutical practice.
  • Such pharmaceutically acceptable carriers may be chemically inert to the active compounds and may have no detrimental side effects or toxicity under the conditions of use.
  • Such formulations may be prepared in accordance with standard and/or accepted pharmaceutical practice. Otherwise, the preparation of suitable formulations may be achieved non-inventively by the skilled person using routine techniques and/or in accordance with standard and/or accepted pharmaceutical practice.
  • a pharmaceutical formulation including a compound of the invention, as hereinbefore defined, in admixture with a pharmaceutically acceptable adjuvant, diluent and/or carrier.
  • a pharmaceutically acceptable adjuvant i.e. active ingredient
  • pharmaceutical formulations include those in which the active ingredient is present in at least 1 % (or at least 10%, at least 30% or at least 50%) by weight. That is, the ratio of active ingredient to the other components (i.e. the addition of adjuvant, diluent and carrier) of the pharmaceutical composition is at least 1 :99 (or at least 10:90, at least 30:70 or at least 50:50) by weight.
  • the amount of compound of the invention in the formulation will depend on the severity of the condition, and on the patient, to be treated, as well as the compound(s) which is/are employed, but may be determined non-inventively by the skilled person.
  • the invention further provides a process for the preparation of a pharmaceutical formulation, as hereinbefore defined, which process comprises bringing into association a compound of the invention, as hereinbefore defined, or a pharmaceutically acceptable ester, amide, solvate or salt thereof with a pharmaceutically-acceptable adjuvant, diluent or carrier.
  • Compounds of the invention may also be combined with other therapeutic agents that are inhibitors of protein or lipid kinases (e.g. PI3K (such as class I PI3K), mTOR, a PI family kinase (e.g. PIM-1 , PI -2 or PIM-3), EGFR and/or MEK) and/or useful in the treatment of a cancer and/or a proliferative disease.
  • PI3K protein or lipid kinases
  • each of components (A) and (B) is formulated in admixture with a pharmaceutically-acceptable adjuvant, diluent or carrier.
  • Such combination products provide for the administration of a compound of the invention in conjunction with the other therapeutic agent, and may thus be presented either as separate formulations, wherein at least one of those formulations comprises a compound of the invention, and at least one comprises the other therapeutic agent, or may be presented (i.e. formulated) as a combined preparation (i.e. presented as a single formulation including a compound of the invention and the other therapeutic agent).
  • a pharmaceutical formulation including a compound of the invention, as hereinbefore defined, another therapeutic agent that is useful in the treatment of cancer and/or a proliferative disease, and a pharmaceutically-acceptable adjuvant, diluent or carrier; and
  • a pharmaceutical formulation including another therapeutic agent that is useful in the treatment of cancer and/or a proliferative disease in admixture with a pharmaceutically-acceptable adjuvant, diluent or carrier, which components (a) and (b) are each provided in a form that is suitable for administration in conjunction with the other.
  • the invention further provides a process for the preparation of a combination product as hereinbefore defined, which process comprises bringing into association a compound of the invention, as hereinbefore defined, or a pharmaceutically acceptable ester, amide, solvate or salt thereof with the other therapeutic agent that is useful in the treatment of cancer and/or a proliferative disease, and at least one pharmaceutically-acceptable adjuvant, diluent or carrier.
  • kits of parts By “bringing into association”, we mean that the two components are rendered suitable for administration in conjunction with each other.
  • the two components of the kit of parts may be:
  • compounds of the invention may be administered at varying therapeutically effective doses to a patient in need thereof.
  • the dose administered to a mammal, particularly a human, in the context of the present invention should be sufficient to effect a therapeutic response in the mammal over a reasonable timeframe.
  • the selection of the exact dose and composition and the most appropriate delivery regimen will also be influenced by inter alia the pharmacological properties of the formulation, the nature and severity of the condition being treated, and the physical condition and mental acuity of the recipient, as well as the potency of the specific compound, the age, condition, body weight, sex and response of the patient to be treated, and the stage/severity of the disease.
  • Administration may be continuous or intermittent (e.g. by bolus injection).
  • the dosage may also be determined by the timing and frequency of administration. In the case of oral or parenteral administration the dosage can vary from about 0.01 mg to about 1000 mg per day of a compound of the invention.
  • the medical practitioner or other skilled person, will be able to determine routinely the actual dosage, which will be most suitable for an individual patient.
  • the above-mentioned dosages are exemplary of the average case; there can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
  • Compounds of the invention may have the advantage that they are effective inhibitors of protein or lipid kinases (e.g. PI3K, such as class I PI3K, and/or mTOR).
  • PI3K protein or lipid kinases
  • compounds of the invention may have the advantage that they are both PI3K (e.g. class I PI3K, such as PI3Ka) inhibitors and mTOR inhibitors, i.e. they may exhibit dual kinase inhibition.
  • Compounds of the invention may also have the advantage that they may be more efficacious than, be less toxic than, be longer acting than, be more potent than, produce fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile (e.g. higher oral bioavailability and/or lower clearance) than, and/or have other useful pharmacological, physical, or chemical properties over, compounds known in the prior art, whether for use in the above- stated indications or otherwise.
  • pharmacokinetic profile e.g. higher oral bioavailability and/or lower clearance
  • Compounds of the invention may be beneficial as they are medicaments with targeted therapy, i.e. which target a particular molecular entity by inferring or inhibiting it (e.g. in this case by inhibiting one or more protein or lipid kinases as hereinbefore described). Compounds of the invention may therefore also have the benefit that they have a new effect (for instance as compared to known compounds in the prior art), for instance, the new effect may be a particular mode of action or another effect resultant of the targeted therapy.
  • Targeted therapies may be beneficial as they may have the desired effect (e.g. reduce cancer, by reducing tumor growth or carcinogenisis) but may also have the advantage of reducing side effects (e.g. by preventing the killing of normal cells, as may occur using e.g. chemotherapy).
  • compounds of the invention may selectively target particular protein or lipid kinases (e.g. the ones described herein) compared to other known protein or lipid kinases (as may be shown experimentally hereinafter). Accordingly, compounds of the invention may have the advantage that certain, specific, cancers may be treated selectively, which selective treatment may also have the effect of reducing side effects.
  • Compounds of the invention may have the advantage that they may exhibit multiple kinase inhibitory activity. In this respect, advantageously, compounds of the invention may be considered as multi-targeted kinase inhibitors. Compounds of the invention that exhibit single selectivity for a kinase may have the additional benefit that they exhibit less side effects, whereas compounds of the invention that exhibit multiple kinase selectivity may have the additional benefit that they exhibit better potency and/or efficacy.
  • the compounds of the invention may have the benefit that they inhibit multiple targets (or even multiple pathways).
  • compounds of the invention may be considered to have an improved kinase inhibition cross-reactivity profile, e.g. by being selective against multiple kinases of therapeutic interest, for instance compared to compounds known in the prior art.
  • Compounds of the invention may therefore additionally act on other key kinases, thereby allowing single-agent administration (or, potentially, combination products with reduced dosages) and providing the associated benefits, e.g. reducing the risk of drug-drug interactions, etc.
  • PI3 kinase activity of compounds of the invention is possible by a number of direct and indirect detection methods.
  • Certain exemplary compounds described herein were prepared, characterized, and assayed for their PI3Ka and mTOR enzymatic activities using the methods described herein. The activities are expressed in IC 50 values that range between 1 to 100 nM (**) and 100 nM to 10 ⁇ (*), as shown in the Examples below (Table 1). The compounds may also be tested in cell-based assays.
  • the kinase activity was measured by using the commercial ADP HunterTM Plus assay available from DiscoveR x (#33-016), which is a homogeneous assay to measure the accumulation of ADP, a universal product of kinase activity.
  • the enzyme, PI3K (p110a/p85o was purchased from Carna Biosciences (#07CBS- 0402A).
  • the assay was done following the manufacturer recommendations with slight modifications: Mainly the kinase buffer was replace by 50 mM HEPES, pH 7.5, 3 mM MgCI 2 , 100 mM NaCI, 1 mM EGTA, 0.04% CHAPS, 2 mM TCEP and 0.01 mg/ml BGG.
  • the PI3K was assayed in a titration experiment to determine the optimal protein concentration for the inhibition assay.
  • serial 1 :5 dilutions of the compounds were added to the enzyme at a fixed concentration (2.5 g/ml).
  • the enzyme was preincubated with the inhibitor and 30 ⁇ PIP 2 substrate (P9763, Sigma) for 5 min and then ATP was added to a final 50 ⁇ concentration. Reaction was carried out for 1 hour at 25°C. Reagent A and B were sequentially added to the wells and plates were incubated for 30 min at 37 °C.
  • Cell culture The cell lines are obtained from the American Type Culture Collection (ATCC). U20S (human osteosarcoma) is cultured in Dulbecco's modified Eagle's medium (DME ). PC3 (human prostate carcinoma), MCF7 (human breast cardinoma), HCT116 (human colon carcinoma), 768-0 (human neuroblastoma), U251 (human glyoblastoma) are grown in RPMI. All media are supplemented with 10% fetal bovine serum (FBS) (Sigma) and antibiotics- antimycotics. Cells are maintained in a humidified incubator at 37°C with 5% C0 2 and passaged when confluent using trypsin/EDTA.
  • DME Dulbecco's modified Eagle's medium
  • PC3 human prostate carcinoma
  • MCF7 human breast cardinoma
  • HCT116 human colon carcinoma
  • 768-0 human neuroblastoma
  • U251 human glyoblastoma
  • FBS fetal bovine serum
  • U2foxRELOC and U2nesRELOC assay The U2nesRELOC assay and the U2foxRELOC assay have been described. Briefly, cells are seeded at a density of 1.0*10 5 cells/ml into black-wall clear-bottom 96-well microplates (BD Biosciences). After incubation at 37°C with 5% C0 2 for 12 hours, 2 ⁇ of each test compound are transferred from the mother plates to the assay plates. Cells are incubated in the presence of the compounds for one hour. Then cells are fixed and the nucleus stained with DAPI (Invitrogen). Finally the plates are washed with X PBS twice and stored at 4°C before analysis.
  • DAPI Invitrogen
  • Image acquirement and processing Assay plates are read on the BD PathwayTM 855 Bioimager equipped with a 488/10 nm EGFP excitation filter, a 380/10 nm DAPI excitation filter, a 515LP nm EGFP emission filter and a 435LP nm DAPI emission filter. Images are acquired in the DAPI and GFP channels of each well using 10x dry objective. The plates are exposed 0.066 ms (Gain 31) to acquire DAPI images and 0.55 ms (Gain 30) for GFP images.
  • the BD Pathway Bioimager outputs its data in standard text files. Data are imported into the data analysis software BD Image Data Explorer.
  • the nuclear/cytoplasmic (Nuc/Cyt) ratios of fluorescence intensity are determined by dividing the fluorescence intensity of the nucleus by the cytoplasmic.
  • a threshold ratio of greater than 1.8 is employed to define nuclear accumulation of fluorescent signal for each cell. Based on this procedure we calculate the percentage of cells per well displaying nuclear translocation or inhibition of nuclear export. Compounds that induce a nuclear accumulation of the fluorescent signal greater than 60% of that obtained from wells treated with 4nM LMB are considered as hits.
  • AKT phosphorylation Inhibition Subconfluent cells are incubated under different conditions and are washed twice with TBS prior to lysis. Lysis buffer is added containing 50 mM Tris HCI, 150 mM NaCI, 1% NP-40, 2mM Na 3 V0 4 , 100 mM NaF, 20 mM Na relieP 2 0 7 and protease inhibitor cocktail (Roche Molecular Biochemicals). The proteins are resolved on 10% SDS-PAGE and are transferred to nitrocellulose membrane (Schleicher & Schuell, Dassel, Germany).
  • the membranes are incubated overnight at 4°C with antibodies specific for Akt, phospho-Ser-473-Akt (Cell Signaling Technology) and ct-tubulin (Sigma), they are washed and then incubated with IRDye800 conjugated anti- mouse and Alexa Fluor 680 goat anti-rabbit IgG secondary antibodies. The bands are visualized using an Odyssey infrared imaging system (Li-Cor Biosciences).
  • the compounds are tested on 96-well trays.
  • Cells growing in a flask are harvested just before they became confluent, counted using a haemocytometer and are diluted down with media adjusting the concentration to the required number of cells per 0.2 ml (volume for each well).
  • Cells are then seeded in 96- well trays at a density between 1000 and 4000 cells/well, depending of the cell size. Cells are left to plate down and grow for 24 hours before adding the drugs.
  • Drugs are weighed out and diluted with DMSO to get them into solution to a concentration of 10mM. From here a "mother plate" with serial dilutions is prepared at 200X the final concentration in the culture.
  • the final concentration of DMSO in the tissue culture media should not exceed 0.5%.
  • the appropriate volume of the compound solution (usually 2 microlitres) is added automatically (Beckman FX 96 tip) to media to make it up to the final concentration for each drug.
  • the medium is removed from the cells and replaced with 0.2 ml of medium dosed with drug.
  • Each concentration is assayed in triplicate.
  • Two sets of control wells are left on each plate, containing either medium without drug or medium with the same concentration of DMSO.
  • a third control set is obtained with the cells untreated just before adding the drugs (seeding control, number of cells starting the culture). Cells are exposed to the drugs for 72 hours and then processed for MTT colorimetric read-out.
  • the enzymatic mTOR activity was measured using a LanthaScreenTM kinase activity assay (Invitrogen).
  • the enzyme was purchased from Invitrogen (PV4754), as well as the GFP-labeled substrate (4EBP1-GFP; PV4759) and the Tb-anti- p4EBP1(pThr46) antibody (PV4757).
  • the assay was performed in 50 mM HEPES buffer, pH 7.5, containing 1.5 mM MnCI 2 , 10 mM MgCI 2 , 1 mM EGTA, 2.5 mM DTT and 0.01% Tween-20.
  • the concentration of the assay components were the following: 0.24 nM mTOR kinase, 400 nM 4EBP1-GFP, 10 mM ATP and serial dilutions of the compound (inhibitor) to be evaluated. After 1 h incubation at room temperature, 20 mM EDTA was used to stop the reaction and terbium- labeled antibody (4 nM) added to detect phosphorylated product. The antibody associates with the phosphorylated product resulting in an increased TR-FRET value.
  • the TR-FRET value (a dimensionless number) was calculated as the ratio of the acceptor signal (GFP, emission at 520 nm) to the donor signal (terbium, emission at 495 nm). Values were plotted against the inhibitor concentration and fitted to a sigmoid dose-response curve using GraphPad software PI3K cellular activity (Elisa assay)
  • Osteosarcoma U20S cells were plated in 96 Poly-D-Lysine coating tissue culture 10 plates (18.000 cells/well). After the treatment with serial dilutions of the compound during 3h, the cells were fixed directly in the wells with 4% paraformaldehyde.
  • DCM dichloromethane
  • CHCI 3 means chloroform
  • MeOH means methanol
  • EtOH means ethanol
  • EtOAc means ethyl acetate
  • THF means tetrahydrofuran
  • ACN means acetonitrile
  • DMAP means 4,4- dimethylaminopyridine
  • DMF means dimethylformamide
  • DME means dimethoxyethane
  • DMSO means dimethylsulfoxide
  • Et 2 0 means diethyl ether
  • Hex means hexane
  • EtOAc means ethyl acetate
  • BA/BE means boronic acid/ester
  • Pd(PPh 3 ) 4 means tetrakis(triphenylphosphine)palladium
  • Pd(Ph 3 P) 2 CI 2 means dichlorobis(triphenylphosphine)palladium(ll
  • Reversed phase HPLC was carried out on a Gemini-NX C18 (100 x 2.0 mm; 5um), Solvent A: water with 0.1% formic acid; Solvent B: acetonitrile with 0.1% formic acid. Gradient: 5% of B to 100% of B within 8 min at 50 °C, DAD.
  • Reversed phase HPLC was carried out on a Gemini-NX C18 (100 x 2.0 mm; 5um), Solvent A: water with 0.1 % formic acid; Solvent B: acetonitrile with 0.1 % formic acid. Gradient: 50% of B to 100% of B within 8 min at 50 °C, DAD.
  • Reversed phase HPLC was carried out on a Gemini-NX C18 (100 x 2.0 mm; 5um), Solvent A: water with 0.1 % formic acid; Solvent B: acetonitrile with 0.1 % formic acid. Gradient: 5% of B to 40% of B within 8 min at 50 °C, DAD.
  • Reversed phase HPLC was carried out on a Gemini C18 column (50 x 2 mm, 3 urn); Solvent A: water with 0.1 % formic acid; Solvent B: acetonitrile with 0.1 % formic acid. Gradient: 10-95 % of B within 4 min at a flow rate of 0.5 mlJmin followed by 2 min of 100 % of B at 0.8 mlJmin, controlled temperature at 50 °C, DAD.
  • Reversed phase HPLC was carried out on a Gemini C18 column (50 x 2 mm, 3 urn); Solvent A: water with 10mM ammonium bicarbonate; Solvent B: acetonitrile. Gradient: 20-100 % of B within 3 min at a flow rate of 0.5 mL/min followed by 2 min of 100 % of B at 0.8 mL/min, controlled temperature at 40 °C, DAD.
  • Reversed phase HPLC was carried out on a Gemini-NX C 8 (100 x 2.0 mm; 5um), Solvent A: water with 0.1% formic acid; Solvent B: acetonitrile with 0.1 % formic acid. Gradient: 0% of B to 30% of B within 8 min at 50 °C, DAD. "Found mass” refers to the most abundant isotope detected in the HPLC-MS.
  • ylamine (0.035 g, 0.08 mmol) was suspended in acetonitrile (1.5 mL) and treated with ethyl isocyanate (0.008 mL, 0.095 mmol) and N,N-diisopropylethylamine (0.035 mL, 0.198 mmol). The reaction mixture was stirred at RT overnight. More ethyl isocyanate (0.02 mL) was added and the reaction mixture was stirred at RT overnight. The reaction mixture was diluted with EtOAc and washed with water. The organic layer was dried over Na 2 S0 4 , filtered and evaporated.
  • TsCI (0.050 g, 0.24 mmol) and DMAP (cat.) were added to a previously sonicated suspension of 5-(3-methoxy-phenyl)-7-oxo-1 ,3,6,7-tetrahydro-8-thia-2,4,6-triaza- cyclopenta[a]indene-2-carboxylic acid ethyl ester (0.030 g, 0.08 mmol) and ⁇ (0.06 mL, 0.24 mmol) in dry acetonitrile (1 mL). The mixture was stirred at RT for 4h. Morpholine (0.03 mL, 0.32 mmol) was added, and the reaction mixture was stirred at 45°C overnight.
  • Example 14 and its corresponding intermediates have been prepared following the synthetic route as described for 5-(2-Amino-pyrimidin-5-yl)-7-morpholin-4-yl- 1,3-dihydro-8-thia-2,4,6-triaza-cyclopenta[a]indene-2-carboxylic acid ethyl ester (Example 13).
  • reaction mixture was heated under microwave irradiation at 130°C for 30 min. On cooling, the mixture was purified by column chromatography (DCM/MeOH 98:2 to 9:1) and by prep-HPLC to afford the title compound ( 2 mg, 15%).
  • Biological activity in PI3-K and/or mTOR for certain examples is represented in Table 1 by semi-quantative results: between 100 nM and 10 ⁇ (0.1-10 ⁇ ) ( * ) and between 1 and 100 nM (0.001-0.1 ⁇ ) (**).
  • selected exemplary compounds displayed the following specific IC 50 values (in ⁇ ): Example 1 (PI3Kcc 0.530; mTOR 0.112), Example 5 (PI3Ka 0.061 ; mTOR 0.200), Example 12 (PI3Ka 0.132; mTOR 0.081).
  • Biological activity for certain examples is also represented by quantitative results in the table below.
  • Cellular activity is also represented in the table below, which measures the cellular activity of certain compounds of the invention/examples by the inhibition of phosphorylation of AKT using ELISA assay (as described hereinbefore).

Abstract

There is provided compounds of formula I, wherein A1, A2, A3, A4, n, the dotted lines, B1, B1a, B2, B2a, B3, B3a, B4, B4a and R3 have meanings given in the description, and pharmaceutically-acceptable esters, amides, solvates or salts thereof, which compounds are useful in the treatment of diseases in which inhibition of a protein or lipid kinase (e.g. a PI3-K and/or mTOR) is desired and/or required, and particularly in the treatment of cancer or a proliferative disease.

Description

TRICYCLIC COMPOUNDS FOR USE AS KINASE INHIBITORS
Field of the Invention
This invention relates to novel pharmaceutically-useful compounds, which compounds are useful as inhibitors of protein or lipid kinases (such as inhibitors of the phosphoinositide 3ΌΗ kinase (PI3 kinase) family, particularly the PI3K class I sub-type. The compounds may also be useful as inhibitors of the mammalian target of rapamycin (mTOR)). The compounds are of potential utility in the treatment of diseases such as cancer. The invention also relates to the use of such compounds as medicaments, to the use of such compounds for in vitro, in situ and in vivo diagnosis or treatment of mammalian cells (or associated pathological conditions), to pharmaceutical compositions containing them, and to synthetic routes for their production.
Background of the Invention
The malfunctioning of protein kinases (PKs) is the hallmark of numerous diseases. A large share of the oncogenes and proto-oncogenes involved in human cancers code for PKs. The enhanced activities of PKs are also implicated in many non-malignant diseases, such as benign prostate hyperplasia, familial adenomatosis, polyposis, neuro-fibromatosis, psoriasis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis glomerulonephritis and post-surgical stenosis and restenosis. PKs are also implicated in inflammatory conditions and in the multiplication of viruses and parasites. PKs may also play a major role in the pathogenesis and development of neurodegenerative disorders. For a general reference to PKs malfunctioning or disregulation see, for instance, Current Opinion in Chemical Biology 1999, 3, 459 - 465.
Phosphatidylinositol 3-kinases (PI3Ks) are a family of lipid and serine/threonine kinases that catalyze the phosphorylation of the membrane lipid phosphatidylinositol (PI) on the 3'-OH of the inositol ring to produce l phosphoinositol-3-phosphate (PIP), phosphoinositol-3,4-diphosphate (PIP2) and phosphoinositol-3,4,5-triphosphate (PIP3), which act as recruitment sites for various intracellular signalling proteins, which in turn form signalling complexes to relay extracellular signals to the cytoplasmic face of the plasma membrane. These 3'-phosphoinositide subtypes function as second messengers in intracellular signal transduction pathways (see e.g. Trends Biochem. Sci 22 87,267-72 (1997) by Vanhaesebroeck et al.; Chem. Rev. 101 (8), 2365-80 (2001) by Leslie et al (2001 ); Annu. Rev. Cell. Dev. Boil. 17, 615-75 (2001) by Katso et al; and Cell. Mol. Life Sci. 59 (5), 761-79 (2002) by Toker et al).
Multiple PI3K isoforms categorized by their catalytic subunits, their regulation by corresponding regulatory subunits, expression patterns and signalling specific funtions (ρ110α, β, δ, γ) perform this enzymatic reaction (Exp. Cell. Res. 25 (1 ),. 239-54 (1999) by Vanhaesebroeck and Katso et al., 2001 , above).
The closely related isoforms p1 10a and β are ubiquitously expressed, while δ and γ are more specifically expressed in the haematopoietic cell system, smooth muscle cells, myocytes and endothelial cells (see e.g. Trends Biochem. Sci. 22 (7),. 267-72 (1997) by Vanhaesebroeck et al). Their expression might also be regulated in an inducible manner depending on the cellular, tissue type and stimuli as well as disease context. Inductibility of protein expression includes synthesis of protein as well as protein stabilization that is in part regulated by association with regulatory subunits. Eight mammalian PI3Ks have been identified so far, including four class I PI3Ks. Class la includes ΡΙ3Κα, ΡΙ3Κβ and PI3K5. All of the class la enzymes are heterodimeric complexes comprising a catalytic subunit (ρ1 10 , ρ1 10β or p1 106) associated with an SH2 domain containing p85 adapter subunit. Class la PI3Ks are activated through tyrosine kinase signalling and are involved in cell proliferation and survival. PI3Koc and ΡΙ3Κβ have also been implicated in tumorigenesis in a variety of human cancers. Thus, pharmacological inhibitors of PI3Ka and ΡΙ3Κβ are useful for treating various types of cancer.
ΡΙ3Κγ, the only member of the Class lb PI3Ks, consists of a catalytic subunit ρ110γ, which is associated with a p110 regulatory subunit. ΡΙ3Κγ is regulated by G protein coupled receptors (GPCRs) via association with βγ subunits of heterotrimeric G proteins. ΡΙ3Κγ is expressed primarily in hematopoietic cells and cardiomyocytes and is involved in inflammation and mast cell function. Thus, pharmacological inhibitors of ΡΙ3Κγ are useful for treating a variety of inflammatory diseases, allergies and cardiovascular diseases.
These observations show that deregulation of phosphoinositol-3-kinase and the upstream and downstream components of this signalling pathway is one of the most common deregulations associated with human cancers and proliferative diseases (see e.g. Parsons et al., Nature 436:792 (2005); Hennessey et al., Nature Rev. Drug Discovery 4: 988-1004 (2005).
The mammalian target of rapamycin (mTOR) also known as FK506 binding protein 12-rapamycin associated protein 1 (FRAP1) is a protein which in humans is encoded by the FRAP1 gene. mTOR is a serine/threonine protein kinase that regulates cell growth, cell proliferation, cell motility, cell survival, protein synthesis, and transcription. The inhibition of mTORs are believed to be useful for treating various diseases/conditions, such as cancer (for example, as described in Easton et al. (2006). "mTOR and cancer therapy". Oncogene 25 (48): 6436^6).
For the treatment of cancer, targeted therapies are becoming more important. That is, therapy that has the effect of interfering with specific target molecules that are linked to tumor growth and/or carcinogenesis. Such therapy may be more effective than current treatments (e.g. chemotherapy) and less harmful to normal cells (e.g. because chemotherapy has the potential to kill normal cells as well as cancerous cells). This, and also the fact that targeted therapies may be selective (i.e. it may inhibit a certain targeted molecule more selectively as compared to other molecular targets, e.g. as described hereinafter), may have the benefit of reducing side effects and may also have the benefit that certain specific cancers can be treated (also selectively). The latter may in turn also reduce side effects.
Hence, it is a clear goal of current oncologists to develop targeted therapies (e.g. ones that are selective). In this respect, it should be pointed out that several different molecular targets may exist that are linked to certain diseases (e.g. cancer). However, one simply cannot predict if a therapy (e.g. a small molecule as a therapeutic) that interferes with or inhibits one target molecule could inhibit a different molecular target (be it one that will ultimately have the effect of treating the same disease or a different one).
The listing or discussion of an apparently prior-published document in this specification should not necessarily be taken as an acknowledgement that the document is part of the state of the art or is common general knowledge.
International patent applications WO 2009/040552 and WO 2009/060197 disclose imidazolodiathiazoles and imidazopyridazines for use as kinase inhibitors and/or for use in the treatment of e.g. cancer. These applications do not disclose or suggest tricyclic compounds.
Disclosure of the Invention
According to the invention, there is now provided a compound of formula I,
Figure imgf000006_0001
n represents 0, 1 or 2;
Ai, A2, A3 and each A4 (if present) independently represents -C(R4)R5-, -C(O)-, -0-, -S-, -S(O)- or -S(0)2-; the dotted lines represent the presence of an optional double bond, which may be present between A, and A2, A2 and A3, A3 and A (if the latter is present, i.e. when n does not represent 0) and/or between two A„ groups (if present, i.e. when n represents 2), provided that the At to A^containing ring is not aromatic; each B\ B1a, B2, B2a, B3, B3a, B4 and B4a independently represent hydrogen or a substituent selected from halo, -C(=Y)-R10a, -C(=Y)-OR10a, -C(=Y)N(R 0a)R11a, -S(O)2N(R10a)R11 a, CV12 alkyl, heterocycloalkyl (which latter two groups are optionally substituted by one or more substituents selected from =0 and E1), aryl and/or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from E2); or any two B , B a, B2, B2a, B3, B3a, B4 and B a substituents that are attached to the same carbon atom (i.e. B1 and B1a; B2 and B2a; B3 and B3a; and/or B4 and B4a) may together form a =0 group; or, any two B1, B1a, B2, B2a, B3, B3a, B4 and B4a substituents may be linked together to form a further 3- to 12- membered (e.g. 3- to 6-membered) ring, optionally containing (in addition to the atom(s) of the morpholine ring) one or more (e.g. two or, preferably, one) heteroatom(s) (preferably selected from sulfur, oxygen and nitrogen), which ring optionally contains one or more (e.g. one to three) double bonds, and which ring is itself optionally substituted by one or more substituents selected from halo, =0 and C,.3 alkyl optionally substituted by one or more fluoro atoms;
R3 represents aryl or heteroaryl (both of which are optionally substituted by one or more substituents selected from E4);
R4 and R5 independently represent (if present), on each occasion when used herein, hydrogen, halo, -OR10c, -N(R10d)R 1d, -N(R10e)-C(O)-R10', -C(0)R1°9, -C(O)OR10h, -C(O)N(R10i)R11i, -N(R10i)-C(O)OR10k, -N(R10m)-C(O)-N(Rl0n)R1 1 n, -N[-C(O)-T1-R10p]-C(O)-T2-R 0q, Ci-12 alkyl, heterocycloalkyl (which latter two groups are optionally substituted by one or more substituents selected from E5 and =0), aryl or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from E6 and =0); or R4 and R5 may be linked together to form a 3- to 6-membered ring, optionally containing one or more (e.g. two or, preferably, one) heteroatom(s) (preferably selected from sulfur, oxygen and nitrogen), which ring optionally contains one or more (e.g. one or two) double bonds, and which ring is itself optionally substituted by one or more substituents selected from halo, =0 and Ci-3 alkyl optionally substituted by one or more fluoro atoms;
T1 and T2 independently represent a single bond, -N(R10*)- or -0-;
R6 represents (if present) hydrogen, -C(O)-R10r, -C(O)-OR10s, -C(O)-N(R10,)R11t, -S(O)2R10u, Ο ΐ2 alkyl, heterocycloalkyl (which latter two groups are optionally substituted by one or more substituents selected from E7 and =0), aryl or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from E8 and =0); each R10a, R1 a, R 0c, R10d, R11d, R10e, R10', R 0g, R 0h, R10i, R11i, R1t>i, R10k, R10m, R10n, R11n, R10p, R10q, R10f, R10s, R10', R11t, R10u and R10x independently represent, on each occasion when used herein, hydrogen, d.12 alkyl, heterocycloalkyl (which latter two groups are optionally substituted by one or more substituents selected from =0, =S, =N(R20) and E10), aryl or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from E11); or any relevant pair of R10a and R1 a (for example, when attached to the same atom) and/or any pair of R10d and R11d, R 0i and R1 i, R10n and R11n and R10t and R11t may be linked together to form (e.g. along with the requisite nitrogen atom to which they may be attached) a 4- to 20- (e.g. 4- to 12-) membered ring, optionally containing one or more heteroatoms (for example, in addition to those that may already be present, e.g. (a) heteroatom(s) selected from oxygen, nitrogen and sulfur), optionally containing one or more unsaturations (preferably, double bonds), and which ring is optionally substituted by one or more substituents selected from =0, =S, =N(R20) and E12; each E1, E2, E4, E5, E6, E7, E8, E10, E11 and E12 independently represents, on each occasion when used herein:
(i) Q4;
(ii) C1-12 alkyl optionally substituted by one or more substituents selected from =0 and Q5; or any two E1, E2, E4, E5, E6, E7, E8, E10, E11 or E12 groups, for example on CM2 alkyl groups, e.g. when they are attached to the same or adjacent carbon atoms, or, on aryl groups e.g. when two groups (e.g. two E4 groups) are attached to adjacent carbon atoms of the aryl group, may be linked together to form a 3- to 12-membered ring, optionally containing one or more (e.g. one to three) unsaturations (preferably, double bonds), and which ring is optionally substituted by one or more substituents selected from =0 and J1; each Q4 and Q5 independently represent, on each occasion when used herein: halo, -CN, -N02, -N(R 0)R21, -OR20, -C(=Y)-R20, -C(=Y)-OR20, -C(=Y)N(R20)R21, -OC(=Y)-R20, -OC(=Y)-OR20, -OC(=Y)N(R20)R21, -OS(0)2OR20, -OP(=Y)(OR20)(OR21), -OP(OR20)(OR21), -N(R2 )C(=Y)R21, -N(R2 )C(=Y)OR21, -N(R22)C(=Y)N(R20)R21, -NR22S(0)2R20, -NR22S(O)2N(R20)R21, -S(O)2N(R20)R21, -SC(=Y)R20, -S(0)2R20, -SR20, -S(0)R20, C1-6 alkyl, heterocycloalkyl (which latter two groups are optionally substituted by one or more substituents selected from =0 and J2), aryl or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from J3); each Y independently represents, on each occasion when used herein, =0, =S, =NR23 or =N-CN; each R20, R21, R22 and R23 independently represent, on each occasion when used herein, hydrogen, C1-6 alkyl, heterocycloalkyl (which latter two groups are optionally substituted by one or more substituents selected from J4 and =0), aryl or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from J5); or any relevant pair of R20, R2 and R22, may (for example, when attached to the same atom, adjacent atom (i.e. 1 ,2-relationship) or to atoms that are two atom atoms apart, i.e. in a 1 ,3-relationship) be linked together to form (e.g. along with the requisite nitrogen atom to which they may be attached) a 4- to 20- (e.g. 4- to 12-) membered ring, optionally containing one or more heteroatoms (for example, in addition to those that may already be present, e.g. (a) heteroatom(s) selected from oxygen, nitrogen and sulfur), optionally containing one or more unsaturations (preferably, double bonds), and which ring is optionally substituted by one or more substituents selected from J6 and =0; each J1, J2, J3, J4, J5 and J6 independently represents, on each occasion when used herein:
(i) Q7;
(ii) Ci-6 alkyl or heterocycloalkyl, both of which are optionally substituted by one or more substituents selected from =0 and Q8; each Q7 and Q8 independently represents, on each occasion when used herein: halo, -N(R50)R51, -OR50, -C(=Ya)-R50, -C(=Ya)-OR50, -C(=Ya)N(R50)R51, -N(R52)C(=Ya)R51 , -NR52S(0)2R50, -S(O)2N(R50)R51 , -N(R5 )-C(=Ya)-N(R50)R51 , -S(0)2R50, -SR50, -S(0)R50 or d.6 alkyl optionally substituted by one or more fluoro atoms; each Ya independently represents, on each occasion when used herein, =0, =S, =NR53 or =N-CN; each R50, R5 , R52 and R53 independently represents, on each occasion when used herein, hydrogen or 01-6 alkyl optionally substituted by one or more substituents selected from fluoro, -OR60 and -N(R61)R62; or
any relevant pair of R50, R51 and R52 may (for example when attached to the same or adjacent atoms) be linked together to form, a 3- to 8-membered ring, optionally containing one or more heteroatoms (for example, in addition to those that may already be present, heteroatoms selected from oxygen, nitrogen and sulfur), optionally containing one or more unsaturations (preferably, double bonds), and which ring is optionally substituted by one or more substituents selected from =0 and C,.3 alkyl; R60, R61 and R62 independently represent hydrogen or C1-6 alkyl optionally substituted by one or more fluoro atoms; or a pharmaceutically acceptable ester, amide, solvate or salt thereof, which compounds, esters, amides, solvates and salts are referred to hereinafter as "the compounds of the invention".
Pharmaceutically-acceptable salts include acid addition salts and base addition salts. Such salts may be formed by conventional means, for example by reaction of a free acid or a free base form of a compound of formula I with one or more equivalents of an appropriate acid or base, optionally in a solvent, or in a medium in which the salt is insoluble, followed by removal of said solvent, or said medium, using standard techniques (e.g. in vacuo, by freeze-drying or by filtration). Salts may also be prepared by exchanging a counter-ion of a compound of the invention in the form of a salt with another counter-ion, for example using a suitable ion exchange resin.
By "pharmaceutically acceptable ester, amide, solvate or salt thereof, we include salts of pharmaceutically acceptable esters or amides, and solvates of pharmaceutically acceptable esters, amides or salts. For instance, pharmaceutically acceptable esters and amides such as those defined herein may be mentioned, as well as pharmaceutically acceptable solvates or salts. Pharmaceutically acceptable esters and amides of the compounds of the invention are also included within the scope of the invention. Pharmaceutically acceptable esters and amides of compounds of the invention may be formed from corresponding compounds that have an appropriate group, for example an acid group, converted to the appropriate ester or amide. For example, pharmaceutically acceptable esters (of carboxylic acids of compounds of the invention) that may be mentioned include optionally substituted Ci_6 alkyl, C5-10 aryl and/or C5.10 aryl-d-6 alkyl- esters. Pharmaceutically acceptable amides (of carboxylic acids of compounds of the invention) that may be mentioned include those of the formula -C(0)N(Rz1)Rz2, in which Rz1 and Rz2 independently represent optionally substituted C^e alkyl, C5-io aryl, or C5-io aryl-C,^ alkylene-. Preferably, Ο 6 alkyl groups that may be mentioned in the context of such pharmaceutically acceptable esters and amides are not cyclic, e.g. linear and/or branched. Further compounds of the invention that may be mentioned include carbamate, carboxamido or ureido derivatives, e.g. such derivatives of existing amino functional groups.
For the purposes of this invention, therefore, prodrugs of compounds of the invention are also included within the scope of the invention.
The term "prodrug" of a relevant compound of the invention includes any compound that, following oral or parenteral administration, is metabolised in vivo to form that compound in an experimentally-detectable amount, and within a predetermined time (e.g. within a dosing interval of between 6 and 24 hours (i.e. once to four times daily)). For the avoidance of doubt, the term "parenteral" administration includes all forms of administration other than oral administration.
Prodrugs of compounds of the invention may be prepared by modifying functional groups present on the compound in such a way that the modifications are cleaved, in vivo when such prodrug is administered to a mammalian subject. The modifications typically are achieved by synthesising the parent compound with a prodrug substituent. Prodrugs include compounds of the invention wherein a hydroxyl, amino, sulfhydryl, carboxy or carbonyl group in a compound of the invention is bonded to any group that may be cleaved in vivo to regenerate the free hydroxyl, amino, sulfhydryl, carboxy or carbonyl group, respectively.
Examples of prodrugs include, but are not limited to, esters and carbamates of hydroxy functional groups, esters groups of carboxyl functional groups, N-acyl derivatives and N-Mannich bases. General information on prodrugs may be found e.g. in Bundegaard, H. "Design of Prodrugs" p. 1-92, Elesevier, New York-Oxford (1985).
Compounds of the invention may contain double bonds and may thus exist as E {entgegen) and Z (zusammen) geometric isomers about each individual double bond. Positional isomers may also be embraced by the compounds of the invention. All such isomers (e.g. if a compound of the invention incorporates a double bond or a fused ring, the cis- and trans- forms, are embraced) and mixtures thereof are included within the scope of the invention (e.g. single positional isomers and mixtures of positional isomers may be included within the scope of the invention).
Compounds of the invention may also exhibit tautomerism. All tautomeric forms (or tautomers) and mixtures thereof are included within the scope of the invention. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies which are interconvertible via a low energy barrier. For example, proton tautomers (also known as prototropic tautomers) include interconversions via migration of a proton, such as keto-enol and imine-enamine isomensations. Valence tautomers include interconversions by reorganisation of some of the bonding electrons.
Compounds of the invention may also contain one or more asymmetric carbon atoms and may therefore exhibit optical and/or diastereoisomerism. Diastereoisomers may be separated using conventional techniques, e.g. chromatography or fractional crystallisation. The various stereoisomers may be isolated by separation of a racemic or other mixture of the compounds using conventional, e.g. fractional crystallisation or HPLC, techniques. Alternatively the desired optical isomers may be made by reaction of the appropriate optically active starting materials under conditions which will not cause racemisation or epimerisation (i.e. a 'chiral pool' method), by reaction of the appropriate starting material with a 'chiral auxiliary' which can subsequently be removed at a suitable stage, by derivatisation (i.e. a resolution, including a dynamic resolution), for example with a homochiral acid followed by separation of the diastereomeric derivatives by conventional means such as chromatography, or by reaction with an appropriate chiral reagent or chiral catalyst all under conditions known to the skilled person.
All stereoisomers (including but not limited to diastereoisomers, enantiomers and atropisomers) and mixtures thereof (e.g. racemic mixtures) are included within the scope of the invention. In the structures shown herein, where the stereochemistry of any particular chiral atom is not specified, then all stereoisomers are contemplated and included as the compounds of the invention. Where stereochemistry is specified by a solid wedge or dashed line representing a particular configuration, then that stereoisomer is so specified and defined.
The compounds of the present invention may exist in unsolvated as well as solvated forms with pharmaceutically acceptable solvents such as water, ethanol, and the like, and it is intended that the invention embrace both solvated and unsolvated forms.
The present invention also embraces isotopicaliy-labeled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature (or the most abundant one found in nature). All isotopes of any particular atom or element as specified herein are contemplated within the scope of the compounds of the invention. Exemplary isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, chlorine and iodine, such as 2H, 3H, 11C, 13C, 14C , 13N, 150, 170, 180, 32P, 33P, 35S, 8F, 36CI, 23l, and 25l. Certain isotopicaliy-labeled compounds of the present invention (e.g., those labeled with 3H and 1 C) are useful in compound and for substrate tissue distribution assays. Tritiated (3H) and carbon-14 (1 C) isotopes are useful for their ease of preparation and detectability. Further, substitution with heavier isotopes such as deuterium (i.e., 2H may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances. Positron emitting isotopes such as 150, 13N, 11C and 18F are useful for positron emission tomography (PET) studies to examine substrate receptor occupancy. Isotopically labeled compounds of the present invention can generally be prepared by following procedures analogous to those disclosed in the Scheme 1 and/or in the Examples herein below, by substituting an isotopically labeled reagent for a non- isotopically labeled reagent. Unless otherwise specified, C1-q alkyl groups (where q is the upper limit of the range) defined herein may be straight-chain or, when there is a sufficient number (i.e. a minimum of two or three, as appropriate) of carbon atoms, be branched- chain, and/or cyclic (so forming a C3.q-cycloalkyl group). Such cycloalkyl groups may be monocyclic or bicyclic and may further be bridged. Further, when there is a sufficient number (i.e. a minimum of four) of carbon atoms, such groups may also be part cyclic. Such alkyl groups may also be saturated or, when there is a sufficient number (i.e. a minimum of two) of carbon atoms, be unsaturated (forming, for example, a C2.q alkenyl or a C2.q alkynyl group).
Unless otherwise stated, the term C1-q alkylene (where q is the upper limit of the range) defined herein may be straight-chain or, when there is a sufficient number of carbon atoms, be saturated or unsaturated (so forming, for example, an alkenylene or alkynylene linker group). However, such d.q alkylene groups may not be branched.
C3.q cycloalkyl groups (where q is the upper limit of the range) that may be specifically mentioned may be monocyclic or bicyclic alkyl groups, which cycloalkyl groups may further be bridged (so forming, for example, fused ring systems such as three fused cycloalkyl groups). Such cycloalkyl groups may be saturated or unsaturated containing one or more double bonds (forming for example a cycloalkenyl group). Substituents may be attached at any point on the cycloalkyl group. Further, where there is a sufficient number (i.e. a minimum of four) such cycloalkyl groups may also be part cyclic.
The term "halo", when used herein, preferably includes fluoro, chloro, bromo and iodo. Heterocycloalkyl groups that may be mentioned include non-aromatic monocyclic and bicyclic heterocycloalkyl groups in which at least one (e.g. one to four) of the atoms in the ring system is other than carbon (i.e. a heteroatom), and in which the total number of atoms in the ring system is between 3 and 20 (e.g. between three and ten, e.g between 3 and 8, such as 5- to 8-). Such heterocycloalkyl groups may also be bridged. Further, such heterocycloalkyl groups may be saturated or unsaturated containing one or more double and/or triple bonds, forming for example a C2.q heterocyc!oalkenyl (where q is the upper limit of the range) group. C2-q heterocycloalkyl groups that may be mentioned include 7- azabicyclo[2.2.1 ]heptanyl, 6-azabicyclo[3.1.1 ]heptanyl, 6-azabicyclo[3.2.1]- octanyl, 8-azabicyclo-[3.2.1]octanyl, aziridinyl, azetidinyl, dihydropyranyl, dihydropyridyl, dihydropyrrolyl (including 2,5-dihydropyrrolyl), dioxolanyl (including 1 ,3-dioxolanyl), dioxanyl (including 1 ,3-dioxanyl and 1 ,4-dioxanyl), dithianyl (including 1 ,4-dithianyl), dithiolanyl (including 1 ,3-dithiolanyl), imidazolidinyl, imidazolinyl, morpholinyl, 7-oxabicyclo[2.2.1]heptanyl, 6- oxabicyclo-[3.2.1]octanyl, oxetanyl, oxiranyl, piperazinyl, piperidinyl, pyranyl, pyrazolidinyl, pyrrolidinonyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, sulfolanyl, 3- sulfolenyl, tetrahydropyranyl, tetrahydrofuranyl, tetrahydropyridyl (such as 1 ,2,3,4-tetrahydropyridyl and 1 ,2,3,6-tetrahydropyridyl), thietanyl, thiiranyl, thiolanyl, thiomorpholinyl, trithianyl (including 1 ,3,5-trithianyl), tropanyl and the like. Substituents on heterocycloalkyl groups may, where appropriate, be located on any atom in the ring system including a heteroatom. The point of attachment of heterocycloalkyl groups may be via any atom in the ring system including (where appropriate) a heteroatom (such as a nitrogen atom), or an atom on any fused carbocyclic ring that may be present as part of the ring system. Heterocycloalkyl groups may also be in the N- or S- oxidised form. Heterocycloalkyl mentioned herein may be stated to be specifically monocyclic or bicyclic.
For the avoidance of doubt, the term "bicyclic" (e.g. when employed in the context of heterocycloalkyl groups) refers to groups in which the second ring of a two-ring system is formed between two adjacent atoms of the first ring. The term "bridged" (e.g. when employed in the context of cycloalkyl or heterocycloalkyl groups) refers to monocyclic or bicyclic groups in which two non-adjacent atoms are linked by either an alkylene or heteroalkylene chain (as appropriate).
Aryl groups that may be mentioned include C6.2o, such as C6.12 (e.g. Ce-ιο) aryl groups. Such groups may be monocyclic, bicyclic or tricyclic and have between 6 and 12 (e.g. 6 and 10) ring carbon atoms, in which at least one ring is aromatic. C6. o aryl groups include phenyl, naphthyl and the like, such as 1 ,2,3,4-tetrahydro- naphthyl. The point of attachment of aryl groups may be via any atom of the ring system. For example, when the aryl group is polycyclic the point of attachment may be via atom including an atom of a non-aromatic ring. However, when aryl groups are polycyclic (e.g. bicyclic or tricyclic), they are preferably linked to the rest of the molecule via an aromatic ring.
Unless otherwise specified, the term "heteroaryl" when used herein refers to an aromatic group containing one or more heteroatom(s) (e.g. one to four heteroatoms) preferably selected from N, O and S. Heteroaryl groups include those which have between 5 and 20 members (e.g. between 5 and 10) and may be monocyclic, bicyclic or tricyclic, provided that at least one of the rings is aromatic (so forming, for example, a mono-, bi-, or tricyclic heteroaromatic group). When the heteroaryl group is polycyclic the point of attachment may be via atom including an atom of a non-aromatic ring. However, when heteroaryl groups are polycyclic (e.g. bicyclic or tricyclic), they are preferably linked to the rest of the molecule via an aromatic ring. Heteroaryl groups that may be mentioned include 3,4-dihydro-1 H-isoquinolinyl, 1 ,3-dihydroisoindolyl, 1 ,3-dihydroisoindolyl (e.g. 3,4- dihydro-1/-/-isoquinolin-2-yl, ,3-dihydroisoindol-2-yl, 1 ,3-dihydroisoindol-2-yl; i.e. heteroaryl groups that are linked via a non-aromatic ring), or, preferably, acridinyl, benzimidazolyl, benzodioxanyl, benzodioxepinyl, benzodioxolyl (including 1 ,3- benzodioxolyl), benzofuranyl, benzofurazanyl, benzothiadiazolyl (including 2,1 ,3- benzothiadiazolyl), benzothiazolyl, benzoxadiazolyl (including 2,1 ,3- benzoxadiazolyl), benzoxazinyl (including 3,4-dihydro-2H-1 ,4-benzoxazinyl), benzoxazolyl, benzomorpholinyl, benzoselenadiazolyl (including 2, 1 ,3-benzoselenadiazolyl), benzothienyl, carbazolyl, chromanyl, cinnolinyl, furanyl, imidazolyl, imidazo[1 ,2-a]pyridyl, indazolyl, indolinyl, indolyl, isobenzofuranyl, isochromanyl, isoindolinyl, isoindolyl, isoquinolinyl, isothiaziolyl, isothiochromanyl, isoxazolyl, naphthyridinyl (including 1 ,6-naphthyridinyl or, preferably, 1 ,5-naphthyridinyl and 1 ,8-naphthyridinyl), oxadiazolyl (including 1 ,2,3-oxadiazolyl, 1 ,2,4-oxadiazolyl and 1 ,3,4-oxadiazolyl), oxazolyl, phenazinyl, phenothiazinyl, phthalazinyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quinolizinyl, quinoxalinyl, tetrahydroisoquinolinyl (including 1 ,2,3,4-tetrahydroisoquinolinyl and 5,6,7,8-tetrahydroisoquinolinyl), tetrahydroquinolinyl (including 1 ,2,3,4- tetrahydroquinolinyl and 5,6,7,8-tetrahydroquinolinyl), tetrazolyl, thiadiazolyl (including 1 ,2,3-thiadiazolyl, 1 ,2,4-thiadiazolyl and 1 ,3,4-thiadiazolyl), thiazolyl, thiochromanyl, thiophenetyl, thienyl, triazolyl (including 1 ,2,3-triazolyl, 1 ,2,4-triazolyl and 1 ,3,4-triazolyl) and the like. Substituents on heteroaryl groups may, where appropriate, be located on any atom in the ring system including a heteroatom. The point of attachment of heteroaryl groups may be via any atom in the ring system including (where appropriate) a heteroatom (such as a nitrogen atom), or an atom on any fused carbocyclic ring that may be present as part of the ring system. Heteroaryl groups may also be in the N- or S- oxidised form. Heteroaryl groups mentioned herein may be stated to be specifically monocyclic or bicyclic. When heteroaryl groups are polycyclic in which there is a non- aromatic ring present, then that non-aromatic ring may be substituted by one or more =0 group.
It may be specifically stated that the heteroaryl group is monocyclic or bicyclic. In the case where it is specified that the heteroaryl is bicyclic, then it may be consist of a five-, six- or seven-membered monocyclic ring (e.g. a monocyclic heteroaryl ring) fused with another a five-, six- or seven-membered ring (e.g. a monocyclic aryl or heteroaryl ring).
Heteroatoms that may be mentioned include phosphorus, silicon, boron and, preferably, oxygen, nitrogen and sulfur.
For the avoidance of doubt, where it is stated herein that a group (e.g. a C1.12 alkyl group) may be substituted by one or more substituents (e.g. selected from E6), then those substituents (e.g. defined by E6) are independent of one another. That is, such groups may be substituted with the same substituent (e.g. defined by E6) or different substituents (defined by E6).
For the avoidance of doubt, in cases in which the identity of two or more substituents in a compound of the invention may be the same, the actual identities of the respective substituents are not in any way interdependent. For example, in the situation in which there is more than one e.g. B1 to B4 or E1 to E12 (such as E6) substituent present, then those B1 to B4 or E1 to E12 (e.g. E6) substituents may be the same or different. Further, in the case where there are e.g. B to B4 or E to E12 (such as E6) substituents present, in which one represents -C(O)R 0a (or e.g. -OR20, as appropriate) and the other represents -C(O)2R10a (or e.g. -C(0)2R20, as appropriate), then those R10a or R20 groups are not to be regarded as being interdependent. Also, when e.g. there are two -OR20 substituents present, then those -OR20 groups may be the same or different (i.e. each R20 group may be the same or different).
For the avoidance of doubt, when a term such as "£' to E12" is employed herein, this will be understood by the skilled person to mean E1, E2, E3 (if present), E4, E5, E6, E7, E8, E9 (if present), E10, E11 and E12, inclusively. The term "B1 to B4" as employed herein will be understood to mean B1, B1a, B2, B2a, B3, B3a, B4 and B4a, inclusively.
For the avoidance of doubt, compounds of formula I that are included within the scope of the invention include those of the following formulae:
Figure imgf000019_0001
wherein the integers are as defined herein. For the avoidance of doubt the two A4 substituents are independent of one another. Further, the morpholinyl group of the above formulae may be substituted as defined herein and the Ai to A4 containing ring may contain one or two double bonds, provided that the ring is not aromatic. The skilled person will appreciate that by "A1 to A4-containing ring", we mean the 5-, 6- or 7-membered ring containing the integers A,, A2, A3, and optionally, one or two A integers. As stated herein, the dotted lines in the Ai to A^-containing ring represent the presence of an optional double bond. However, the Ai to A4-containing ring may not be aromatic. Hence, when the A, to A4-containing ring is 5- or 6-membered (i.e. when n represents 0 or 1 ), then a maximum of one double bond may be present (in addition to the requisite double bond that is common to the Ai to A4- containing ring and the thienyl moiety of the compound of formula I). When the A, to A^-containing ring is 7-membered, then up to two double bonds may be present (in addition to the requisite double bond already present). The skilled person will also appreciate that the rules of valency should be adhered to. Hence, where for example any one of A, to A4 represents e.g. -O- or -C(O)-, then a double bond may not be adjacent that A, to A4 group. Similarly, a double bond may not be adjacent another double bond, etc.
All individual features (e.g. preferred features) mentioned herein may be taken in isolation or in combination with any other feature (including preferred feature) mentioned herein (hence, preferred features may be taken in conjunction with other preferred features, or independently of them).
The skilled person will appreciate that compounds of the invention that are the subject of this invention include those that are stable. That is, compounds of the invention include those that are sufficiently robust to survive isolation from e.g. a reaction mixture to a useful degree of purity.
Particularly preferred compounds of the invention that may be mentioned include the following compounds of formula I (in which formulae IA and IB are the most preferred):
Figure imgf000020_0001
IA IB IC
which:
1 to B4 all preferably represent hydrogen, and hence there is preferably unsubstituted morpholinyl group present;
R4'5 represents hydrogen or one or more substituent(s) defined by R4 or R5, which may be present on a carbon atom of the A, to A -containing ring (preferably these represent hydrogen and hence the AT to A4-containing ring is preferably unsubstituted on the carbon atoms); R5 is as defined herein, i.e. it may represent hydrogen or a substituent that is present on the nitrogen atom of the A to Vcontaining ring as defined herein; R3 most preferably represents pyrimidinyl (e.g. 5-pyrimidinyl) optionally substituted by one or more (e.g. two or, preferably, one) substituent(s) selected from E4 (in which E4 is as defined herein) and most preferably represents the following fragment:
Figure imgf000021_0001
in which the squiggly line represents the point of attachment to the core tricyclic structure of formula I, and R3a, R3b and R3c each independently represent hydrogen or a substituent as defined by E4 herein;
one of R3a, R3b and R3c (preferably R3c) represents a substituent and the other two (e.g. R3a and R3b) represent hydrogen;
when one of R3a, R3 and R3c (e.g. R3c) represents E4, then E4 preferably represents Q4 (and Q4 preferably represents -N(R20)R21, in which R20 and R21 each preferably represent hydrogen, so forming a -NH2 group);
R3 preferably represents amino-pyrimidinyl, for instance 2-amino-5-pyrimidinyl (e.g. 2-NH2-5-pyrimidinyl), i.e. :
Figure imgf000021_0002
Preferred compounds of the invention that may be mentioned include those in which:
R4 and R5 independently represent, on each occasion when used herein, hydrogen, halo, -OR10c, -N(R10d)R 1d, -N(R10e)-C(O)-R 0f, -C(O)R10g, -C(O)OR10h, -C(O)N(R 0i)R1 1 i, -N(R10i)-C(O)OR10k, -N(R10m)-C(O)-N(R10n)R1 ln,
C .i2 alkyl, heterocycloalkyl (which latter two groups are optionally substituted by one or more substituents selected from E5 and =0), aryl or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from E6 and =0); or
R4 and R5 are linked together as defined herein. Further preferred compounds of the invention that may be mentioned include those in which, for instance when n represents 0 or 1 (which is preferably the case):
A, represents -C(R4)(R5)- or -C(O)-;
A2 represents -C(R4)(R5)- or -N(R6)-;
A3 represents -C(R )(R5)- or -N(R6)- (provided that both A2 and A3 do not represent -N(R5)-);
A4 (if present) represents -C(R<!)(R5)- or -C(O)-;
it is preferred that, when AT represents -C(O)-, A2 and A4 represent -C(R4)(R5)-, then A3 does not represent -N(R6)-;
it is preferred that, when A3 and A represent -C(R )(R5)-, then A2 does not represent -C(O)- when ΑΊ represents -N(R6)-;
preferred Ai to A4-containing rings include those in which:
(a) when n represents 1 , then:
(i) A2 represents -N(R6)-, A3 represents -C(R4)(R5)- and one of ΑΊ and A4 represents -C(O)- and the other represents -C(R4)(R5)-; or, preferably,
(ii) AT represents -C(R4)(R5)- or -C(O)-, A2 and A3 independently represent -N(R6)- or -C(R4)(R5)- and A4 represents -C(R4)(R5)- or -C(O)-;
(b) when n represents 0, then A2 represents -N(R6)- or -C(R )(R5)-, A-, and A3 independently represent -C(R4)(R5)- or -C(O)-.
Exemplary embodiments of R3 include, but are not limited to: pyrrole, pyrazole, triazole, tetrazole, thiazole, isothiazole, oxazole, isoxazole, isoindole, 1 ,3-dihydro- indol-2-one, pyridine-2-one, pyridine, pyridine-3-ol, imidazole, 1 H-indazole, 1 H- indole, indolin-2-one, 1-(indolin-1-yl)ethanone, pyrimidine, pyridazine, pyrazine and isatin groups, 1 H-benzo[d][1 ,2,3]triazole, 1 H-pyrazolo [3,4-b]pyridine, 1 H- pyrazolo[3,4-d]pyrimidine, 1 H-benzo[d]imidazole, 1 H-benzo[d]imidazol-2(3H)- one, 1 H-pyrazolo[3,4-c]pyridine, 1 H-pyrazolo[4,3-d]pyrimidine, 5H-pyrrolo[3,2- d]pyrimidine, 2-amino-1 H-purin-6(9H)-one, quinoline, quinazoline, quinoxaline, isoquinoline, isoquinolin-1 (2H)-one, 3,4-dihydroisoquinolin-1 (2H)-one, 3,4- dihydroquinolin-2(1 H)-one, quinazolin-2(1H)-one, quinoxalin-2(1 H)-one, 1 ,8- napthyridine, pyrido[3,4-d]pyrimidine, and pyrido[3,2-b]pyrazine, 1 ,3-dihydro benzimidazolone, benzimidazole, benzothiazole and benzothiadiazole, groups. These groups may be unsubstituted or substituted.
Preferred compounds of the invention include those in which:
when R3 represents aryl (e.g. phenyl), then that group may be unsubstituted but is preferably substituted by at least one (e.g. two or, preferably, one) substituent(s) selected from E";
when R3 represents monocyclic heteroaryl (e.g. a 5- or 6-membered heteroaryl group), then that group preferably contains 1 , 2, 3 or 4 nitrogen atoms and, optionally 1 or 2 additional heteroatoms selected from oxygen and sulfur, and which heteroaryl group is optionally substituted by one or more substituents selected from E4;
when R3 represents bicyciic heteroaryl (e.g. a 8-, 9- or 10-membered heteroaryl group), then that group preferably consists of a 5- or 6-membered ring fused to another 5- or 6-membered ring (in which either one of those rings may contain one or more (e.g. four, or, preferably one to three) heteroatoms), in which the total number of heteroatoms is preferably one to four, and which ring is optionally substituted by one or more (e.g. two or, preferably, one) substituent(s) selected from E4 (and, if there is a non-aromatic ring present in the bicyciic heteroaryl group, then such a group may also be substituted by one or more (e.g. one) =0 groups);
optional substituents (e.g. the first optional substituent) on the R3 group (e.g. when it represents aryl, such as phenyl) are preferably selected from -OR, -SR, -CH2OR, C02R, CF2OH, CH(CF3)OH, C(CF3)2OH, -(CH2)wOR, -(CH2)WNR2, -C(0)N(R)2, -NR2, -NRC(0)R, -NRC(0)NHR, -NRC(0)N(R)2, -S(0)yN(R)2, -OC(0)R, OC(0)N(R)2, -NRS(0)yR, -NRC(0)N(R)2, CN, halogen and -N02 (in which each R is independently selected from H, d-C6 alkyl, C3-C10 cycloalkyl and a 5- to 12-membered aryl or heteroaryl group, the groups being unsubstituted or substituted (for example by one or more substituents as defined herein, e.g. substituents on E4 moieties, e.g. =0, J2, J3, J4 and/or J5), w is 0, 1 or 2 and y is 1 or 2); when R3 represents aryl (e.g. phenyl), then that group is substituted by one or two substituents (e.g. by a first substituent as defined above, and, optionally a further substituent; or a further two substituents) preferably selected from halo, d-12 alkyl, CN, N02, ORd, SRd, NRd 2, C(0)Rd, SORd, S02Rd, S02N(R) 2, NC(0)Rd and C02Rd (wherein each Rd is independently H or d-C6 alkyl);
when R3 represents substituted aryl (e.g. phenyl), the the substituent may be situated at the 2-, 3-, 4-, 5- or 6- position of the phenyl ring (typically it is situated at position 3 or 4); particularly preferred are phenyl groups substituted by -ORd (in which Rd is independently H or d-C6 alkyl, e.g. methyl), e.g. -OH; in this embodiment the -ORd group, or -OH group, is typically situated at the 3- or 4- position of the phenyl ring, so forming a 3-hydroxyphenyl or 4-hydroxyphenyl group or an isostere thereof, 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 compounds of the invention; isosteres of 3-hydroxyphenyl and 4- hydroxyphenyl groups are encompassed within the definition of R3;
when R3 represents heteroaryl, it is unsubstituted or substituted (when substituted, it may be substituted by one or more substitutents selected from those listed in respect of substituents on R3, when R3 is a phenyl group; typically, the substituents are selected from OH and NH2, or alkylated derivatives thereof).
Further preferred compounds of the invention include those in which:
each R10a, R11a, R 0c, R10d, R1 d, R 0e, R10f, R109, R10h, R10i, Rl 1i, R10', R10k, R10m,
R10n, R 1n, R10p, R10q, R10r, R 0s, R10', R11t, R10u and R10x independently represent, on each occasion when used herein, hydrogen, d-12 (e.g. d-e) alkyl (which latter group is optionally substituted by one or more substituents selected from =0 and E10) or aryl (optionally substituted by one or more E 1 substituents); or
any relevant pair of R10a and R11a and/or any pair of R10d and R11d, R10i and R 1i, R10n and R11n and R 0t and R111 may, when attached to the same nitrogen atom, be linked together to form (along with the requisite nitrogen atom to which they are attached) a 3- to 12- (e.g. 4- to 12-) membered ring, optionally containing one or more (e.g. one to three) double bonds, and which ring is optionally substituted by one or more substituents selected from E12 and =0;
T1 and T2 independently represent a single bond; each of E\ E2, E4, E5, E6, E7, E8, E10, E11 and E12 independently represents, on each occasion when used herein, Q4 or C1.6 alkyl (e.g. C1-3) alkyl optionally substituted by one or more substituents selected from =0 and Q5;
each Q4 and Q5 independently represent halo, -CN, -N02, -N(R20)R21 , -OR20, -C(=Y)-R20, -C(=Y)-OR20, -C(=Y)N(R20)R21, -N(R22)C(=Y)R21, -N(R22)C(=Y)OR21 , -N(R 2)C(=Y)N(R20)R21 , -NR22S(0)2R20, -NR2 S(O)2N(R20)R21, -S(O)2N(R20)R21 , -S(0)2R20, -SR20, -S(0)R20 or C,.6 alkyl optionally substituted by one or more fluoro atoms (and each Q5 more preferably represents halo, such as fluoro);
any two E1, E2, E4, E5, E6, E7, E8, E10, E11 or E12 groups may be linked together, but are preferably not linked together;
each R20, R21, R22 and R23 independently represent, on each occasion when used herein, aryl (e.g. phenyl; preferably unsubstituted, but which may be substituted by one to three J5 groups) or, more preferably, hydrogen or Ci.6 (e.g. C^) alkyl optionally substituted by one or more substituents selected from =0 and J4; or any pair of R20 and R2 , may, when attached to the same nitrogen atom, be linked together to form a 4- to 8-membered (e.g. 5- or 6-membered) ring, optionally containing one further heteroatom selected from nitrogen and oxygen, optionally containing one double bond, and which ring is optionally substituted by one or more substituents selected from J6 and =0;
each J1 , J2, J3, J4, J5 and J6 independently represent Ci.6 alkyl (e.g. acyclic C1.3 alkyl or, e.g. in the case of J4, C3.5 cycloalkyl) optionally substituted by one or more substituents selected from =0 and Q8, or, more preferably, such groups independently represent a substituent selected from Q7;
each Q7 and Q8 independently represents a substituent selected from halo (e.g. fluoro), -N(R50)R51, -OR50, -C(=Ya)-R50, -C(=Ya)-OR50, -C(=Ya)N(R50)R51 , -NR5 S(0)2R50, -S(O)2-N(R50)R51, -N(R5 )-C(=Ya)-N(R50)R51 , -S(0)2R50 or C,.e alkyl optionally substituted by one or more fluoro atoms;
each R50, R51, R52 and R53 substituent independently represents, on each occasion when used herein, hydrogen or d.6 (e.g. 0,.3) alkyl optionally substituted by one or more substituents selected from fluoro;
when any relevant pair of R50, R51 and R52 are linked together, then those pairs that are attached to the same nitrogen atom may be linked together (i.e. any pair of R50 and R51), and the ring so formed is preferably a 5- or 6-membered ring, optionally containing one further nitrogen or oxygen heteroatom, and which ring is optionally substituted by one or more substituents selected from =0 and C 3 alkyl (e.g. methyl);
R60, R61 and R62 independently represent hydrogen or C1 (e.g. Ci.2) alkyl optionally substituted by one or more fluoro atoms.
Preferred optional substituents on R3 and the , to A4-containing ring (and, when they represent a substituent other than hydrogen on R2, R3 and R4 groups) include:
=0 (e.g. in the case of alkyl, cycloalkyl or heterocycloalkyi groups);
-CN;
halo (e.g. fluoro, chloro or bromo);
alkyl, which alkyl group may be cyclic, part-cyclic, unsaturated or, preferably, linear or branched (e.g. alkyl (such as ethyl, ?-propyl, isopropyl, /-butyl or, preferably, n-butyl or methyl), all of which are optionally substituted with one or more halo (e.g. fluoro) groups (so forming, for example, fluoromethyl, difluoromethyl or, preferably, trifluoromethyl) or substituted with an aryl, heteroaryl or heterocycloalkyi group (which themselves may be substituted with one or more -ORz , -CiOR22, -C(0)ORz3, -N(Rz4)RzS, -S(0)2Rz6, -S(0)2N(Rz7)Rz8;
-N(Rz9)-C(0)-Rz1°, -C(0)-N(Rz1 l)Rz'2, -N(Rz9)-S(0)2Rz10 and/or -N(RZ9)-C(0)- N(Rz °) substituents;
aryl (e.g. phenyl), if appropriate (e.g. when the substitutent is on an alkyl group, thereby forming e.g. a benzyl group);
-ORz1;
-C(0)Rz2;
-C(0)ORz3;
-N(Rz4)R25;
-S(0)2Rz6;
-S(0)2N(Rz7)RzB;
-N(Rz9)-C(0)-Rz °;
-C(0)-N(Rz )Rz12;
-N(Rz9)-C(0)-N(Rz1°);
-N(Rz9)-S(0)2Rz °;
wherein each Rz1 to Rz 2 independently represents, on each occasion when used herein, H or alkyl (e.g. ethyl, n-propyl, f-butyl or, preferably, n-butyl, methyl, isopropyl or cyclopropylmethyl (i.e. a part cyclic alkyl group)) optionally substituted by one or more halo (e.g. fluoro) groups (so forming e.g. a trifluoromethyl group). Further, any two Rz groups (e.g. Rz4 and Rz5), when attached to the same nitrogen heteroatom may also be linked together to form a ring such as one hereinbefore defined in respect of corresponding linkage of R10a and R11a groups.
Preferred compounds of the invention include those in which:
B1, B1a, B2, B2a, B3, B3a, B4 and B4a independently represent hydrogen, C1-6 (e.g. d.3) alkyl optionally substituted by one or more substituents selected from =0 and E1, any two of these together form a =0 substituent on the morpholinyl ring, or, any two B1, B a, B2, B2a, B3, B3a, B4 and B a substituents when linked together, may form a linkage, for example between a B2 or B2a substituent and a B3 or B3a substituent for a further ring, e.g. a five membered ring such as the one depicted below:
Figure imgf000027_0001
each E\ E2, E4, E5, E6, E7, E8, E10, E11 and E12 independently represents d.12 alkyl optionally substituted by one or more substituents selected from =0 and Q5, or, preferably (each E1 to E12 independently represent) Q4;
each R20, R2 , R22 and R23 (e.g. each R20 and R21) independently represents heteroaryl, preferably, aryl (e.g. phenyl) (which latter two groups are optionally substituted by one or more substituents selected from J5), or, more preferably, hydrogen or C1-B (e.g. C1-4) alkyl optionally substituted by one or more substituents selected from =0 and J4; or
any relevant pair of R20, R21 and R22 (e.g. R20 and R21) may (e.g. when both are attached to the same nitrogen atom) be linked together to form a 3- to 8- (e.g. 4- to 8-) membered ring, optionally containing a further heteroatom, and optionally substituted by one or more substituents selected from =0 and J6;
each J1, J2, J3, J4, J5 and J6 independently represent alkyl (e.g. C1-3 acyclic alkyl or C3.5 cycloalkyl) optionally substituted by one or more substituents selected from Q8, or, J1 to J6 more preferably represent a substituent selected from Q7; each Q7 and Q8 independently represent halo, -N(R50)R51, -OR50, -C(=Ya)-OR50, .C(=Ya)-RS0, -S(O)2RS0 or C1 alkyl optionally substituted by one or more fluoro atoms;
Ya and Y each independently represent =NR23 or =NR53 (as appropriate) or, preferably, =0;
each R50, R51, R52 and R53 independently represents hydrogen or (e.g. Ci.4) alkyl optionally substituted by one or more fluoro atoms;
each R60, R61 and R62 independently represents hydrogen or C1-2 alkyl (e.g. methyl).
More preferred compounds of the invention include those in which:
each R10a, R 1a, R10c, R10d, Rl 1d, R10e, R10f, R109, R10h, R10i, R1 i, R 0i, R10k, R 0m, Rion( Rnn Riop Rioq) ριοΓι r Rl0ti ρι« Riou and independentl represents hydrogen, (e.g. C1-3) alkyl (optionally substituted by one or more substituents selected from =0 and E10, but which alkyl group is more preferably unsubstituted) or aryl (e.g. phenyl; which aryl group is optionally substituted by one or more E1 substituents); or
any relevant pair of R10a and R 1a and/or any pair of R10d and R11d, R 0i and R1 ', R 0n and R11n and R10t and R111 may be linked together to form a 5- or, preferably, a 6-membered ring, optionally containing a further heteroatom (preferably selected from nitrogen and oxygen), which ring is preferably saturated (so forming, for example, a piperazinyl or morpholinyl group), and optionally substituted by one or more substituents selected from =0 and E12 (which E12 substituent may be situated on a nitrogen heteroatom; and/or E12 is preferably halo (e.g. fluoro) or C1-3 alkyl optionally substituted by one or more fluoro atoms); each E1, E2, E4, E5, E6, E7, E8, E10, E11 and E 2 independently represents a substituent selected from Q4, or (e.g.) E4 may represent C -4 alkyl optionally substituted by one or more Q5 substituents;
Q4 and Q5 independently represent halo (e.g. fluoro), -OR20, -N(R20)R21, -C(=Y)OR20, -C(=Y)N(R20)R21, -NR22S(0)2R2°, heterocycloalkyl, aryl, heteroaryl (which latter three groups are optionally substituted with one or more substitutents selected from J2 or J3, as appropriate) and/or Ci.6 alkyl (e.g. C -3 alkyl) optionally substituted by one or more fluoro atoms;
each Y represents, on each occasion when used herein, =NR23, preferably, =S, or more preferably =0; each R20, R21, R22 and R23 (e.g. each R20 and R21) independently represents hydrogen or C (e.g. C1.3) alkyl (e.g. tert-butyl, ethyl, methyl or a part cyclic group such as cyclopropylmethyl) optionally substituted (but preferably unsubstituted) by one or more (e.g. one) J4 substituent(s); or
any relevant pair of R20, R21 and R22 (e.g. R20 and R21) may (e.g. when both are attached to the same nitrogen atom) be linked together to form a 5- or, preferably, a 6-membered ring, optionally containing a further heteroatom (preferably selected from nitrogen and oxygen), which ring is preferably saturated (so forming, for example, a piperazinyl or morpholinyl group), and optionally substituted by one or more substituents selected from =0 and J6 (which J6 substituent may be situated on a nitrogen heteroatom);
R2Z represents C1-3 alkyl or, preferably, hydrogen;
each J1, J2, J3, J4, J5 and J6 independently represent a substituent selected from Q7, or J1 to J6 (e.g. J4) represents (e.g. preferably unsubstituted) C1-6 alkyl (e.g. C3.5 cycloalkyl);
each Q7 and Q8 independently represent halo (e.g. fluoro), -C(=Ya)-OR50, -C(=Ya)-R50, -S(0)2R50 or d.3 alkyl optionally substituted by one or more fluoro atoms;
each Ya independently represents =NR53, preferably, =S or, more preferably, =0; each R50 independently represents C1.4 alkyl (e.g. ferf-butyl or methyl).
Preferred ^ to /^-containing rings of the compounds of the invention include those of the following formulae: Particularly preferred rings include:
Figure imgf000029_0001
wherein, the carbon atoms may be unsubstituted or substituted by a substituent defined by R4 or R5, and R6 is as hereinbefore defined. Particularly preferred rings include:
Figure imgf000029_0002
and especially preferred is/are (the) 6-membered ring(s). Preferred R3 groups of the compounds of the compounds of the invention include optionally substituted phenyl and pyrimidinyl (e.g. 5-pyrimidinyl), azaindolyl (e.g. azaindol-5-yl), indolyl (e.g. 5-indolyl or 4-indolyl) and pyridyl (e.g. 3-pyridyl). Particularly preferred R3 groups of compounds of the invention include optionally substituted phenyl and pyrimidinyl (e.g. 5-pyrimidinyl).
Preferred compounds of the invention include those in which:
R3 represents aryl (e.g. phenyl) or heteroaryl (e.g. a 5- or 6-membered monocyclic heteroaryl group or a 9- or 10-membered bicyclic heteroaryl group; which groups may contain one to four, e.g 3 or, preferably, 1 or 2, heteroatoms preferably selected from nitrogen, oxygen and sulfur) both of which are optionally substituted by one or more (e.g. two, or, preferably, one) substituent(s) selected from E4 (e.g. -CF3, -OH, -OCH3 and/or -N(R20)R21 (e.g. -NH2 or -N(H)-CH2-cyclopropyl));
each R4 and R5 independently represent -C(O)N(R 0i)R1 i (e.g. in which one of R10' and R11' is hydrogen and the other is as herein defined), or each R4 and R5 preferably (and independently) represent hydrogen, 01-6 alkyl (optionally substituted as defined herein; but preferably unsubstituted), -OR10c or -C(O)OR10h;
R4 and R5 may be linked, but are more preferably not linked together;
there is two or, preferably one or none R4 or R5 substituents (i.e. that are not hydrogen) present in the A, to A4-containing ring (i.e. all the R4'5 substituents present, except two or preferably one, represent hydrogen);
each R6 (when/if present) independently represents hydrogen, -C(O)R10r, -C(O)OR10s, -C(O)N(R10l)R11t, -S(O)2R10u or C1-6 (e.g. C^, such as methyl or butyl (e.g. s-butyl) or C5.6 cycloalkyl, e.g. cyclohexyl) alkyl optionally substituted by one or more (e.g. two or, preferably, one) E7 substituents;
R10c represents hydrogen;
R10s represents C1-3 alkyl (e.g. ethyl);
R101 represents hydrogen;
R ior R iou and Rnt jnc|epenc|entiy represent heteroaryl or, preferably, aryl (e.g. phenyl; which aryl/heteroaryl group is/are optionally substituted by one or more E11 substituent, so forming e.g. a fluorophenyl group) or d.3 alkyl; E4 represents Q4 (e.g. -OR20 and/or -N(R20)R21) or d.6 (e.g. Cv3> such as methyl) alkyl optionally substituted by one or more Q5 substituents (e.g. fluoro, so forming for example a trifluoromethyl group);
E7 and E11 independently represent Ci-6 (e.g. C3-6) alkyl or Q4;
Q4 represents halo, -OR20, -N(R20)R21 , -C(=Y)OR20, heterocycloalkyl (e.g. a 4- to 6-membered ring, containing preferably one heteroatom selected from nitrogen and oxygen), aryl (e.g. phenyl; optionally substituted with two or, preferably, one substituent selected from J3) or heteroaryl (e.g. a 5- or 6-membered monocyclic heteroaryl group preferably containing one or two heteroatoms preferably selected from nitrogen, oxygen and sulfur; which group may be substituted, but is preferably unsubstituted);
Q5 represents -OR20, -N(R20)R21 or, preferably, halo (e.g. fluoro);
Y represents =0;
R20 and R21 independently represent hydrogen, C1-3 alkyl (e.g. methyl or ethyl), which latter group is optionally substituted by one or more (e.g. one) substituent(s) selected from J4;
when there is a -N(R20)R21 moiety present, then one of R20 and R21 represents hydrogen, and the other represents hydrogen, alkyl (e.g. methyl or ethyl), which latter group is optionally substituted by one or more (e.g. one) substituent(s) selected from J4;
J3 represents Q7;
J4 represents C-,.6 alkyl, such as C3.6 alkyl (especially C3.6 cycloalkyl, such as cyclopropyl);
Q7 represents halo (e.g. fluoro) or -S(0)2R5°;
R50 represents alkyl (e.g. methyl).
Preferred compounds of the invention include those in which:
R3 represents aryl (e.g. phenyl) or heteroaryl (e.g. a 5- or 6-membered monocyclic heteroaryl group; which may contain one to four, e.g. 3 or, preferably, 1 or 2, heteroatoms preferably selected from nitrogen, oxygen and sulfur) both of which are optionally substituted by one or more (e.g. two, or, preferably, one) substituent(s) selected from E4 (e.g. -CF3, preferably, -OH and/or -N(R 0)R21 (e.g. -NH2));
each R4 and R5 independently represent hydrogen or 0,.6 alkyl (optionally substituted as defined herein; but preferably unsubstituted); R4 and R5 may be linked, but are more preferably not linked together;
each R6 (when/if present) independently represents hydrogen, -C(O)R 0r, -C(O)OR10s, -C(O)N(R10,)R11t, -S(O)2R 0u or C1-6 (e.g. C1-4) alkyl (e.g. ethyl or methyl) optionally substituted by one or more (e.g. two or, preferably, one) E7 substituents;
R10r represents aryl (e.g. phenyl; which aryl group is optionally substituted by one or more E11 substituent, so forming e.g. a fluorophenyl group) or CL3 alkyl (e.g. methyl);
R10s represents d-3 alkyl (e.g. isopropyl, methyl or, preferably, ethyl);
R10t represents hydrogen;
R11t represents aryl (e.g. phenyl; which aryl group is optionally substituted by one or more E11 substituent, so forming e.g. a fluorophenyl group) or C 3 alkyl (e.g. ethyl);
R10u represents aryl (e.g. phenyl; which aryl group is optionally substituted by one or more E11 substituent, so forming e.g. a fluorophenyl group) or CL3 (e.g. C1.2) alkyl (e.g. ethyl or, preferablyl, methyl);
E7 represents Q4;
when E7 represents Q4, then Q4 preferably represents aryl (e.g. phenyl) optionally substituted by one or more substituents selected from J3 (so forming e.g. a fluorophenyl group);
E11 represents Q4;
when E11 represents Q4, then Q4 represents halo (e.g. fluoro);
J3 represents Q7, in which Q7 preferably represents halo (e.g. fluoro);
E4 represents alkyl (e.g. methyl; which alkyl group is optionally substituted by one or more substituents selected from Q5, in which Q5 is preferably fluoro, so forming e.g. a -CF3 group) or, more preferably, E4 represents Q4;
when E4 represents Q4, then Q4 represents halo, -OR20 or -N(R20)R21;
Q4 represents alkyl (e.g. methyl; which alkyl group is optionally substituted by one or more substituents selected from J2, in which J2 is preferably fluoro, so forming e.g. a -CF3 group) or, more preferably, Q4 represents halo, -OR20,
-N(R20)R21 or aryl (optionally substituted by one or more substituents selected from J3);
Q5 represents halo (e.g. fluoro);
J2 represents halo (e.g. fluoro);
Y represents =0; R20 and R21 independently represent hydrogen or C1.3 alkyl (e.g. methyl or ethyl); Q7 represents halo (e.g. fluoro).
Particularly preferred compounds of the invention include those in which:
R3 represents hydroxyphenyl (e.g. 3-hydroxyphenyl) or pyrimidinyl (e.g. 5- pyrimidinyl, such as 2-amino-4-trifluoromethyl-5-pyrimidinyl (e.g. 2-NH2,4-CF3- pyrimidin-5-yl) or, preferably, 2-amino-5-pyrimidinyl (i.e. 2-[-N(R 0)(R21)]-pyrimidin- 5-yl such as 2-NH2-pyrimidin-5-yl));
A, represents -C(R4)R5-;
one of A2 and A3 (preferably A2) represents -N(R6)- and the other (preferably A3) represents -C(R4)R5-;
n represents 0 or 1 ;
A4 represents (if present) -C(R4)R5-;
only a maximum of two of Ai, A2, A3 and, if present, A4, represents -C(O)- (which, if there are two -C(O)- moieties, are preferably not adjacent to one another);
the dotted lines do not represent the presence of an optional double bond (i.e. the ΑΊ to A4-containing ring does not contain a double bond, other than that double bond that is integral to the requisite imidazopyrazine of formula I);
B1 , B1a, B2, B2a, B3, B3a, B4 and B a independently represent hydrogen;
each R4 and R5 independently represent hydrogen or C1-3 alkyl (e.g. methyl); there is two or, preferably one or none R4 or R5 moieties present that do not represent hydrogen (e.g. there are two or, preferably, one that represent Cv3 alkyl);
each R6 (when/if present) independently represents -S(0)2-CH2CH3, -C(0)OCH3, -C(0)0-C(H)(CH3)2, preferably, -C(0)-N(H)-[4-fluorophenyl], -S(0)2CH3, hydrogen or, more preferably, -C(0)OCH2CH3, -C(0)N(H)CH2CH3, -S(0)2-[4-fluorophenyl], -C(0)-[4-fluorophenyl], -C(0)CH3, ethyl or -CH2-[4- fluorophenyl]. Particularly preferred compounds of the invention include those of the examples described hereinafter.
Compounds of the invention may be made in accordance with techniques that are well known to those skilled in the art, for example as described hereinafter. According to a further aspect of the invention there is provided a process preparation of a compound of formula I which process comprises:
(i) reaction of a compound of formula II,
Figure imgf000034_0001
wherein L1 represents a suitable leaving group, such as iodo, bromo, chloro or a sulfonate group (e.g. -OS(0)2CF3, -OS(0)2CH3 or -OS(0)2PhMe), and A1, A2, A3, A4, n, the dotted lines, B1, B1a, B2, B2a, B3, B3a, B4, B4a and R2 are as hereinbefore defined, with a compound of formula III,
R3-L2 III wherein L2 represents a suitable group such as -B(OH)2, -B(ORw )2 or -Sn(R *)3, in which each Rwx independently represents a d-e alkyl group, or, in the case of -B(ORwx)2, the respective R"* groups may be linked together to form a 4- to 6- membered cyclic group (such as a 4,4,5, 5-tetramethyl-1 ,3,2-dioxaborolan-2-yl group), thereby forming e.g. a pinacolato boronate ester group, (or L2 may represent iodo, bromo or chloro, provided that L1 and L2 are mutually compatible) and R3 is as hereinbefore defined. The reaction may be performed, for example in the presence of a suitable catalyst system, e.g. a metal (or a salt or complex thereof) such as Pd, Cul, Pd/C, PdCI2, Pd(OAc)2, Pd(Ph3P)2CI2, Pd(Ph3P)4 (i.e. palladium tetrakistriphenylphosphine), Pd2(dba)3 and/or NiCI2 (preferred catalysts include palladium) and a ligand such as PdCI2(dppf).DCM, f-Bu3P, (C6Hn)3P, Ph3P, AsPh3, P(o-Tol)3, 1 ,2-bis(diphenylphosphino)ethane, 2,2'-bis(di-ferf-butyl- phosphino)-1 , 1 '-biphenyl, 2,2'-bis(diphenylphosphino)-1 , 1 '-bi-naphthyl, 1 ,1'- bis(diphenyl-phosphino-ferrocene), 1 ,3-bis(diphenylphosphino)propane, xantphos, or a mixture thereof (preferred ligands include PdCI2(dppf).DCM), together with a suitable base such as, Na2C03, K3P04, Cs2C03> NaOH, KOH, K2C03, CsF, Et3N, ( -Pr)2NEt, f-BuONa or f-BuOK (or mixtures thereof; preferred bases include Na2C03 and K2C03) in a suitable solvent such as dioxane, toluene, ethanol, dimethylformamide, dimethoxyethane, ethylene glycol dimethyl ether, water, dimethylsulfoxide, acetonitrile, dimethylacetamide, /V-methylpyrrolidinone, tetrahydrofuran or mixtures thereof (preferred solvents include dimethylformamide and dimethoxyethane). The reaction may be carried out for example at room temperature or above (e.g. at a high temperature such as at about the reflux temperature of the solvent system). Alternative reaction conditions include microwave irradiation conditions, for example at elevated temperature of about 130°C;
(ii) reaction of a compound of formula IV,
Figure imgf000035_0001
wherein L3 represents a suitable leaving group, such as one hereinbefore defined in respect of L1, and A1, A2, A3, A4, n, the dotted lines and R3 are as hereinbefore defined, with a compound of formula V,
Figure imgf000035_0002
wherein L4 may represent hydrogen (so forming an amine group), and L , B\ B1a, B2, B2a, B3, B3a, B4 and B4a are as hereinbefore defined, and the reaction may be performed in the presence of an appropriate metal catalyst (or a salt or complex thereof) such as Cu, Cu(OAc)2, Cul (or Cul/diamine complex), copper tris(triphenylphosphine)bromide, Pd(OAc)2, tris(dibenzylideneacetone)- dipalladium(O) (Pd2(dba)3) or NiCI2 and an optional additive such as Ph3P, 2,2'- bis(diphenylphosphino)-1 ,1'-binaphthyl, xantphos, Nal or an appropriate crown ether such as 18-crown-6-benzene, in the presence of an appropriate base such as NaH, Et3N, pyridine, A/.A/'-dimethylethylenediamine, Na2C03, K2C03, K3P04, Cs2C03, r-BuONa or f-BuOK (or a mixture thereof, optionally in the presence of 4A molecular sieves), in a suitable solvent (e.g. dichloromethane, dioxane, toluene, ethanol, isopropanol, dimethylformamide, ethylene glycol, ethylene glycol dimethyl ether, water, dimethylsulfoxide, acetonitrile, dimethylacetamide, /V-methylpyrrolidinone, tetrahydrofuran or a mixture thereof). This reaction may be performed at elevated temperature or under microwave irradiation reaction conditions, for example as described in process step (i). The compound of formula IV (e.g. in which L3 is chloro) may be prepared in situ, for example from a compound corresponding to a compound of formula IV, but in which L3 represents -Od.3 alkyl (e.g. methoxy) by reaction in the presence of e.g. a chlorinating agent (such as POCI3);
(iii) reaction of a compound of formula VI,
Figure imgf000036_0001
wherein A1, A2, A3, A4, n, the dotted lines and R3 are as hereinbefore defined, with a compound of formula V in which L4 represents hydrogen (so forming optionally substituted morpholine), which reaction may proceed by the reaction initially with a reagent that may convert the oxo moiety into a leaving group (and hence form a compound of formula IV in situ), for instance para-toluenesulfonyl chloride (e.g. in the presence of base (such as triethylamine or the like), a catalytic amount of DMAP, and the compound of formula VI in a suitable solvent such as dry acetonitrile or the like), followed by the addition of the compound of formula V in which L4 represents hydrogen; (iv) for certain compounds of formula I, reaction of a compound of formula VII,
Figure imgf000037_0001
wherein (Ax) and (Ay) denotes the optional presence of the relevant AT to A4 groups that are/may be present in the compound of formula I, and FG1 and FG2 independently represent mutually compatible functional groups, which may undergo an intramolecular reaction to form the requisite Ai to A4-containing ring of formula I (and L1R3 represents R3 or L1, and R3, L1, B1, B1a, B2, B2a, B3, B3a, B4 and B4a are as hereinbefore defined; when L R3 represents L1 , then this step is followed by reaction with a compound of formula III as hereinbefore defined). The skilled person will appreciate that the to A^ groups in the compound of formula I formed, may be present either at the positions represented by (Ax) or (Ay) or may be an integral part of FG1 and/or FG2. By mutually compatible functional groups (FG1 and FG2), we mean that such groups may be manipulated so as to promote an intramolecular reaction, for example, FG1 may be -NH2 and FG2 may be -C(0)OH (or a derivative thereof; e.g. an ester), which functional groups may undergo an amide coupling reaction to form a -N(H)C(0)- linkage (and therefore a ring). In this instance, (Ax) is absent, the -N(H)-C(0)- linkage so formed represents the Ai and A2 moieties (i.e. these emanate from an integral part of FG1 and FG2), and (Ay) represents A3 and A4, which together are -CH2-CH2-. Alternatively, FG1 and FG2 may independently represent leaving groups, such as those hereinbefore defined in respect of L1. In this instance, the compound of formula VII may be reacted with a nucleophile (such as one with more than one nucleophilic site e.g. ammonia), which may displace each of the leaving groups to form the requisite AT to A„-containing ring of formula I (followed by, if necessary, reaction with a compound of formula III as hereinbefore defined). Transformation of the functional group(s) (FG1 and/or FG2) following methodology very well known for any person skill in the art, would allow to obtain an intramolecular cyclisation to obtain a tricycle of formula I; (v) compounds of formula I in which there is a -N(R6)- moiety present, in which R6 represents d.12 alkyl optionally substituted as hereinbefore defined (i.e. by one or more substituent(s) selected from E7 and =0), may be prepared by reaction of a corresponding compound of formula I in which R6 represents hydrogen, with either: a compound of formula VIII,
R6a-C(0)H VIII wherein R6a represents d.n alkyl optionally substituted by one or more substituent(s) selected from E7 and =0 (but preferably not substituted with a =0 substituent), under reductive amination reaction conditions, for example in a "one- pot" procedure in the presence of an appropriate reducing agent, such as sodium cyanoborohydride or sodium triacetoxyborohydride, or alternatively in two distinct steps by a condensation reaction to form e.g. an enamine, followed by reduction under reaction conditions such as in the presence of NaBH4 or the like; or a compound of formula IX,
R6b-L c IX in which L1c represents a suitable leaving group such as one hereinbefore defined in respect of L1 (e.g. halo, such as chloro or bromo), and R6b represents C,.12 alkyl optionally substituted by one or more substituents selected from =0 and E7 (the skilled person will appreciate that certain other compounds of formula I may also be prepared by reaction by the foregoing reaction with a compound of formula IX, e.g. compounds in which R6 represents a certain heterocycloalkyi group attached to a relevant leaving group, although the substitution may be hindered or a SN1 substitution may need to be promoted);
(vi) compounds of formula I in which there is a -N(R6)- moiety present, in which R6 represents -C(0)N(H)R111, may be prepared by reaction of a corresponding compound of formula I in which R6 represents hydrogen, with a compound of formula X,
R11,-N=C=0 X wherein R111 is as hereinbefore defined, for example, under reaction conditions known to those skilled in the art, such as those described herein, e.g. in the presence of a suitable base such as an amine base (e.g. diisopropylamine, diisopropylethylamine, or the like) and optionally in the presence of a suitable solvent (e.g. acetonitrile or the like);
(vii) compounds of formula I in which there is a -N(R6)- moiety present, in which R6 represents -C(O)R10r or -S(O)2R10u, may be prepared by reaction of a corresponding compound of formula I in which R6 represents hydrogen, with a compound of formula XI,
G1-L1b XI wherein G1 represents either -C(O)R10r or -S(O)2R10u, and L (attached to the -C(O)- or -S(0)2 moieties) represents a suitable leaving group such as iodo, bromo or, preferably, chloro, under reaction conditons known to those skilled in the art, for example at around room temperature or above in the presence of a suitable base (e.g. pyridine, triethylamine, dimethylaminopyridine, diisopropylamine, sodium hydroxide, or mixtures thereof), an appropriate solvent (e.g. pyridine, dichloromethane, chloroform, tetrahydrofuran, dimethylformamide, triethylamine, dimethylsulfoxide, water or mixtures thereof) and, in the case of biphasic reaction conditions, optionally in the presence of a phase transfer catalyst; (viii) for compounds of formula I in which there is a -N(R6)- moiety present, in which R6 represents hydrogen, deprotection of a corresponding compound of formula I in which R6 represents a carboxylic acid (or ester group) (e.g. the conversion of a -N-(-C(0)-0-ethyl) moiety to a -N(H) moiety, which nitrogen atom is an integral part of a heterocycloalkyl ring system), for instance by reaction in the presence of a base (e.g. lithium hydroxide hydrate/monohydrate) in a suitable solvent (e.g. a mixture of methanol/isopropanol);
(ix) for compounds of formula I in which there is a hydroxy substituent present (e.g. a E4 substituent, which represents -OR20, and R20 represents hydrogen methyl), transformation of a corresponding compound in which there is a methoxy group present (i.e. in which R20 represents methyl), by reaction in the presence of an appropriate reagent, such as boron fluoride-dimethyl sulfide complex or BBr3 (e.g. in the presence of a suitable solvent such as dichloromethane);
(x) for compounds of formula I in which n represents 1 or 2, in which a (or the) A4 moiety adjacent to the requisite bicycle represents -C(O)-, A, represents -C(R )(R5)- and A2 represents -N(R6)- (and preferably n represents 1 , Ai and A3 represent -C(R4)(R5)- and A2 represents -N(R6)- in which R6 is preferably hydrogen) may be prepared by reaction of a compound of formula XIA,
Figure imgf000040_0001
wherein (A4) denotes the optional presence of a further A4 group, and L R3, B1, B1a, B2, B2a, B3, B3a, B4 and B4a, A3, A4 and R6 are as hereinbefore defined, with a compound of formula XIB,
R -C(0)-R5 XIB wherein R4 and R5 are as hereinbefore defined, for instance under Pictet- Spengler reaction conditions (e.g. as described in Bioorg. Med. Chem. 2008 (16), 542-559 (e.g. in the presence of a source of H+), optionally followed by, if necessary (i.e. for reaction with compounds in which L1R3 represents L1), reaction with a compound of formula III as hereinbefore defined.
Compounds of formula II in which L1 represents a sulfonate group (e.g. -OS(0)2-Phlv1e) may be prepared by reaction of a compound of formula XII,
Figure imgf000041_0001
wherein A1, A2, A3, A4, n and the dotted lines are as hereinbefore defined, with the appropriate sulfonyl chloride (e.g. para-toluene sulfonyl chloride) (e.g. in the presence of an appropriate amine, such as an organic amine base e.g. triethylamine, in an appropriate solvent, e.g. dichloromethane, optionally in the presence of catalytic DMAP), which is followed by the addition of a compound of formula V in which L4 represents hydrogen (e.g. optionally substituted morpholine).
Compounds of formula VI may be prepared by reaction of a compound of formula XIII,
Figure imgf000041_0002
wherein R** represents hydrogen or is preferably other than hydrogen (so forming an ester) for instance an optionally substituted Cn.i2 alkyl group (e.g. methyl), and A1, A2, A3, A4, n and the dotted lines are as hereinbefore defined, with a compound of formula XIV, R3-CsN XIV wherein R3 is as hereinbefore defined, i.e. optionally substituted aryl or heteroaryl (and -CN represents a substituent on a carbon atom of that aryl or heteroaryl ring), for example in the presence of a suitable solvent system (e.g. dioxane), acid (e.g. 4M HCI) under pressure (e.g. reaction in a pressurised sealed tube). The sealed tube may be left in an ultrasonic bath for a periodof time (at elevated temperature, e.g. at about 130°C), and the reaction mixture may then be evaporated, the residue taken up in dry toluene, treated with base (e.g. triethylamine or the like), and heated at reflux for a period of time, after which a further different solvent may be added (e.g. diethyl ether) and the desired product may precipitate out.
Compounds of formula XIA may be prepared by amination of a corresponding compound of formula XIVA,
Figure imgf000042_0001
wherein L7 represents a suitable leaving group (such as one hereinbefore defined by L1, e.g. chloro) and (At), L1R3, B1, B1a, B2, B2a, B3, B3a, B4 and B4a, A3 and A4 are as hereinbefore defined, under standard amination conditions (e.g. in the presence of an amine H2NR6, and optionally a reagent that promotes the substitution). Compounds of formula XII may be prepared by intramolecular reaction of a compound of formula XV,
Figure imgf000043_0001
wherein Rxx (preferably methyl), A1 , A2, A3, A4, n and the dotted lines are as hereinbefore defined, under standard conditions, for instance in the presence of basic conditions (e.g. in the presence of KOH, in an appropriate solvent such as methanol), which reaction mixture may be heated at reflux followed by quench by the addition of HCI.
Compounds of formula XIII (in particular those in which ΑΊ and A (if present) represents -C(R4)R5-, one of A2 and A3 (preferably A3) represents -C(R4)R5- and the other (e.g. A2) represents -C(R4)R5- or -N(R6)-; preferably A3 represents -C(R4)R5- and A2 represents -N(R6)-) may be prepared by reaction of a compound of formula XVI,
Figure imgf000043_0002
or a stereoisomer (e.g. tautomer, such as the keto tautomer) thereof (or another suitable derivative thereof; e.g. an alkylated derivative, where there is e.g. an alkoxy group (e.g. -OCH3) present in place of the hydroxy group) wherein A1 , A2, A3, A4, n and the dotted lines are as hereinbefore defined (and preferably A, and A4 (if present) represents -C(R4)R5-, one of A2 and A3 (preferably A3) represents -C(R4)R5- and the other (e.g. A2) represents -C(R4)R5- or -N(R6)-; but, may also represent -C(O)-), with a compound of formula XVII,
Figure imgf000043_0003
wherein Rxx is as hereinbefore defined, for example the compound of formula XVI may first be subjected to conditions to convert the hydroxy moiety to a suitable leaving group, for instance reaction conditions include the presence of a base (e.g. an organic amine base, such as triethylamine or the like), a suitable solvent (such as dichloromethane) and a reagent that is suitable for the conversion (e.g. a sulfonyl choride to convert the -OH to a sulfonate, for instance mesyl chloride to convert to -0-S(0)2-CH3), followed by reaction with the compound of formula XVII, for instance in the presence of a suitable base (e.g. an alkoxide, such as sodium methoxide). Other compounds of formula XVI that may be mentioned include those in which n represents 1 , A-i , A2 and , each represent -C(R4)R5- (e.g. in which R4 and R5 represent hydrogen) and A3 represents -N(R6)-, in which R6 may represent -C(0)-0-alkyl (e.g. -C(0)-0-fe/†-butyl, i.e. a nitrogen-protecting group), which compound of formula XVI may itself be prepared in accordance with standard procedures e.g. such as those described in the prior art, e.g. WO 02/12242. More compounds of formula XVI that may be mentioned include those in which n represents 1 , and preferably A, represents -C(O)-, and A2, A3 and A4 independently represent -C(R4)(R5)- (which compounds are preferably prepared for instance by reaction in accordance with the procedures described in Akhrem et al. Bulletin of Academy of Sciences of USSR, Division of Chemical Science (English translation) 1973, vol 22, p 836 or Achrem et al (SU371209, 1973; ref. Zh. Khim., 1974, vol. 7, #N245P). Further compounds of formula XVI that may be mentioned include those in which n represents 0, Ai and A3 represent -C(O)-, and A2 represents -N(R6)- (in which R6 is preferably hydrogen), which compound may be prepared in accordance with standard procedures, such as those described in J. Am. Chem. Soc, 1958, vol. 80, p. 1385, 1387. Yet further compounds of formula XVI that may be mentioned include those in which n represents 2, A^ A2 and A3 represent -C(R )R5-, the A4 adjacent A3 represents -N(R6)- and the second A represents -C(O)-, which compounds may be prepared in accordance with the techniques described in e.g. R. G. Glushkov and T. V. Stezhko, Chemistry of Heterocyclic Compounds 1978; 14(9): 1013-1016 (Reaction of 2,3-dioxo-4-(/V,/V- dimethylaminomethylene)hexahydroazepine with hydroxylamine).
Compounds of formula XIVA may be prepared by reaction of a compound of XVIIA,
Figure imgf000045_0001
wherein L1R3, B1, B1a, B2, B2a, B3, B3a, B4 and B a are as hereinbefore defined, with a compound of formula XVIIB,
L8-C(0)-(A4)-A3-L7 XVIIB or the like, wherein L represents e.g. a halo, such as a chloro group (or the like), and (A4), A3 and L7 are as hereinbefore defined, under standard Friedel-Crafts reaction conditions, e.g. in the presence of a Lewis acid to promote the leaving of the L8 group (and therefore the reactivity of the compound of formula XVIIB).
Compounds of formula XV may be prepared from corresponding compounds of formula XIII as hereinbefore defined, in the presence of a reagent to convert the amino group to a urea, e.g. chlorosulfonyl isocyanate or the like.
Other specific transformation steps (including those that may be employed in order to form compounds of formula I and any intermediate compounds to compounds of formula I) that may be mentioned include:
(i) reductions, for example of a carboxylic acid (or ester) to either an aldehyde or an alcohol, using appropriate reducing conditions (e.g. -C(0)OH (or an ester thereof), may be converted to a -C(0)H or -CH2-OH group, using DIBAL and L1AIH4, respectively (or similar chemoselective reducing agents));
(ii) reductions of an aldehyde (-C(O)H) group to an alcohol group (-CH2OH), using appropriate reduction conditions such as those mentioned at point (i) above;
(iii) oxidations, for example of a moiety containing an alcohol group (e.g. -CH2OH) to an aldehyde (e.g. -C(O)H), for example in the presence of a suitable oxidising agent, e.g. n02 or the like; (iv) reductive amination of an aldehyde and an amine, under appropriate reaction conditions, for example in "one-pot" procedure in the presence of an appropriate reducing agent, such as a chemoselective reducing agent such as sodium cyanoborohydride or, preferably, sodium triacetoxyborohydride, or the like. Alternatively, such reactions may be performed in two steps, for example a condensation step (in the presence of e.g. a dehydrating agent such as trimethyl orthoformate or MgS04 or molecular sieves, etc) followed by a reduction step (e.g. by reaction in the presence of a reducing agent such as a chemoselective one mentioned above or NaBH4, AIH , or the like), for instance the conversion of -NH2 to -N(H)-isopropyl by condensation in the presence of acetone (H3C-C(0)-CH3) followed by reduction in the presence of a reducing agent such as sodium cyanaoborohydride (i.e. overall a reductive amination);
(v) amide coupling reactions, i.e. the formation of an amide from a carboxylic acid (or ester thereof), for example when R2 represents -C(0)OH (or an ester thereof), it may be converted to a -C(O)N(R10b )R l 1 group (in which R10b1 and R11b1 are as hereinbefore defined, and may be linked together, e.g. as defined above), and which reaction may (e.g. when R2 represents -C(O)OH) be performed in the presence of a suitable coupling reagent (e.g. 1 ,1'-carbonyldiimidazole, Λ/,/V- dicyclohexylcarbodiimide, or the like) or, in the case when R2 represents an ester (e.g. -C(0)OCH3 or -C(0)OCH2CH3), in the presence of e.g. trimethylaluminium, or, alternatively the -C(0)OH group may first be activated to the corresponding acyl halide (e.g -C(0)CI, by treatment with oxalyl chloride, thionyl chloride, phosphorous pentachloride, phosphorous oxychloride, or the like), and, in all cases, the relevant compound is reacted with a compound of formula HN(R10a)R11a (in which R10a and R 1a are as hereinbefore defined), under standard conditions known to those skilled in the art (e.g. optionally in the presence of a suitable solvent, suitable base and/or in an inert atmosphere);
(vi) conversion of a primary amide to a nitrile functional group, for example under dehydration reaction conditions, e.g. in the presence of POCI3, or the like;
(vii) nucleophilic substitution reactions, where any nucleophile replaces a leaving group, e.g. methylsulfonylpiperazine may replace a chloro leaving group;
(viii) transformation of a methoxy group to a hydroxy group, by reaction in the presence of an appropriate reagent, such as boron fluoride-dimethyl sulfide complex or BBr3 (e.g. in the presence of a suitable solvent such as dichloromethane); (ix) alkylation, acylation or sulfonylation reactions, which may be performed in the presence of base and solvent (such as those described hereinbefore in respect of preparation of compounds of formula I, process step (iv) above, for instance, a - N(H)- or -OH or -NH2 (or a protected version of the latter) moiety may be alkylated, acylated or sulfonylated by employing a reactant that is an alkyl, acyl or sulfonyl moiety attached to a leaving group (e.g. C^e alkyl-halide (e.g. ethylbromide), alkyl-C(0)-halide (e.g. H3C-C(0)CI), an anhydride (e.g. H3C-C(0)-0-C(0)-CH3, i.e. "-0-C(0)-CH3" is the leaving group), dimethylformamide (i.e. -N(CH3)2 is the leaving group) or a sulfonyl halide (e.g. H3C-S(0)2CI) and the like);
(x) formation of a urea functional group by reaction of an amine (e.g. a secondary amine, such as a -NH moiety that is a part of a heterocyclic group) with an alkyl isocyanate (e.g. ethyl isocyanate) to form a -N-C(0)-N(H)-alkyl (e.g. -N-C(0)-N(H)-CH2CH3 moiety), which transformation may be performed in the presence of a suitable solvent (e.g. acetonitrile) and base (e.g. N,N- diisopropylethylamine);
(xi) specific deprotection steps, such as deprotection of an /V-Boc protecting group by reaction in the presence of an acid (or another suitable method known to those skilled in the art or a specific method described in the experimental hereinafter), or, a hydroxy group protected as a silyl ether (e.g. a re/ -butyl- dimethylsilyl protecting group) may be deprotected by reaction with a source of fluoride ions, e.g. by employing the reagent tetrabutylammonium fluoride (TBAF). Other deprotection steps that may be mentioned include the removal of a carboxylic acid (or ester group) attached to a nitrogen atom (e.g. the conversion of a -N-(-C(0)0-ethyl) moiety to a -N(H) moiety, which nitrogen atom may be an integral part of a heterocycloalkyl ring system), for instance by reaction in the presence of a base (e.g. lithium hydroxide hydrate/monohydrate) in a suitable solvent (e.g. a mixture of methanol/isopropanol);
(xii) for compounds in which there is a halo (e.g. bromo, iodo or chloro) substituent present (e.g. one that is attached to an aromatic group), reaction of a corresponding compound, in which there is a hydrogen atom present, with a reagent that is a source of halide ions (a halogenating reagent). For instance, an electrophile that provides a source of iodide ions includes iodine, diiodoethane, diiodotetrachloroethane or, preferably, A/-iodosuccinimide, a source of bromide ions includes A -bromosuccinimide and bromine, and a source of chloride ions includes V-chlorosuccinimide, chlorine and iodine monochloride, for instance in the presence of a suitable solvent, such as CHCI3 or an alcohol (e.g. methanol), optionally in the presence of a suitable base, such as a weak inorganic base, e.g. sodium bicarbonate. Typically, the reaction maybe performed by heating at a convenient temperature, either by conventional heating under reflux or under microwave irradiation;
(xiii) for compounds in which there is a substituent (e.g. alkyl) other that hydrogen, or halo (e.g. bromo, iodo or chloro) present, reaction of a corresponding compound in which there is a halo (e.g. bromo, chloro or iodo) group present, with a compound containing the relevant substituent (e.g. alkyl) which has a suitable leaving group attached to it (e.g. halo, such as chloro);
(xiv) for compounds of formula I (or intermediates thereto) in which there is -C(O)- group present, adjacent a N atom (e.g. for compounds in which A2 represents -N(R6)- and A represents -C(O)-), oxidation of corresponding compounds in which there is a -CH2- present (e.g. in which AT represents -CH2-) for instance in accordance with the procedures described in e.g. Bioorg. Med. Chem. Lett., 2008, (18) 4054 or Tetrahedron Lett., 2009 (50) 3436.
Intermediate compounds described herein are either commercially available, are known in the literature, or may be obtained either by analogy with the processes described herein, or by conventional synthetic procedures, in accordance with standard techniques, from available starting materials using appropriate reagents and reaction conditions. Further, processes to prepare compounds of formula I may be described in the literature, for example in:
Werber.G. et al.; J. Heterocycl. Chem.; EN; 14; 1977; 823-827;
Andanappa K. Gadad et al. Bioorg. Med. Chem. 2004, 12, 5651-5659;
Paul Heinz et al. Monatshefte fur Chemie, 1977, 08, 665-680;
M.A. El-Sherbeny et al. Boll. Chim. Farm. 1997, 136, 253-256;
Nicolaou, K. C; Bulger, P. G.; Sarlah, D. Angew. Chem. Int. Ed. 2005, 44, 2-49; Bretonnet et al. J. Med. Chem. 2007, 50, 1872 ;
Asuncion Marin et al. Farmaco 1992, 47 (1), 63-75;
Severinsen, R. et al. Tetrahedron 2005, 61, 5565-5575;
Nicolaou, K. C; Bulger, P. G.; Sarlah, D. Angew. Chem. Int. Ed. 2005, 44, 2-49;
M. Kuwahara et al., Chem. Pharm Bull., 1996, 44, 122;
Wipf, P.; Jung, J.-K. J. Org. Chem. 2000, 65(20), 6319-6337; Shintani, R.; Okamoto, K. Org. Lett. 2005, 7 (21), 4757-4759;
Nicolaou, K. C; Bulger, P. G.; Sarlah, D. Angew. Chem. Int. Ed. 2005, 44, 2-49;
J. Kobe et al., Tetrahedron, 1968, 24, 239 ;
P.F. Fabio, A.F. Lanzilotti and S.A. Lang, Journal of Labelled Compounds and Pharmaceuticals, 1978, 15, 407;
F.D. Bellamy and K. Ou, Tetrahedron Lett, 1985, 25, 839;
M. Kuwahara et al., Chem. Pharm Bull. , 1996, 44, 122;
A.F. Abdel-Magid and C.A Maryanoff. Synthesis, 1990, 537;
M. Schlosser et al. Organometallics in Synthesis. A Manual, (M. Schlosser, Ed.), Wiley &Sons Ltd: Chichester, UK, 2002, and references cited therein;
L. Wengwei et al., Tetrahedron Lett, 2006, 47, 1941 ;
M. Plotkin et al. Tetrahedron Lett , 2000, 41, 2269;
Seyden-Penne, J. Reductions by the Alumino and Borohydrides, VCH, NY, 1991 ;
O. C. Dermer, Chem. Rev., 1934, 14, 385;
N. Defacqz, et al., Tetrahedron Lett, 2003, 44, 91 1 ;
S.J. Gregson et al., J. Med. Chem., 2004, 47, 1 161 ;
A. M. Abdel Magib, et al., J. Org. Chem., 1996, 61, 3849;
A.F. Abdel-Magid and C.A Maryanoff. Synthesis, 1990, 537;
T. Ikemoto and M. Wakimasu, Heterocycles, 2001 , 55, 99;
E. Abignente et al., II Farmaco, 1990, 45, 1075;
T. Ikemoto et al. , Tetrahedron, 2000, 56, 7915;
T. W. Greene and P. G. M. Wuts, Protective Groups in Organic Synthesis, Wiley, NY, 1999;
S. Y. Han and Y.-A. Kim. Tetrahedron, 2004, 60, 2447;
J. A. H. Lainton et al., J. Comb. Chem., 2003, 5, 400; or
Wiggins, J. M. Synth. Commun., 1988, 18, 741.
Those skilled in the art will appreciate that other synthetic routes may be used to synthesise the compounds of the invention. Although specific starting materials and reagents are depicted hereinbefore and discussed below, other starting materials and reagents can be easily substituted to provide a variety of derivatives and/or reaction conditions.
The substituents R3, B , B1a, B2, B2a, B3, B3a, B4, B4a, A1 t A2, A3 and (A^ in final compounds of the invention or relevant intermediates may be modified one or more times, after or during the processes described above by way of methods that are well known to those skilled in the art. Examples of such methods include substitutions, reductions, oxidations, alkylations, acylations, hydrolyses, esterifications, etherifications, halogenations or nitrations. Such reactions may result in the formation of a symmetric or asymmetric final compound of the invention or intermediate. The precursor groups can be changed to a different such group, or to the groups defined in formula I, at any time during the reaction sequence. For example, when substituents in the compounds of the invention (e.g. represented by R3, B1, B1a, B2, B2a, B3, B3a, B4, B4a, A1 t A2, A3 and (A4)n) such as C02Et, CHO, CN and/or CH2CI, are present, these groups can be further derivatized to other fragments described (e.g. by those integers mentioned above) in compounds of the invention, following synthetic protocols very well know to the person skilled in the art and/or according to the experimental part described in the patent. Other specific transformation steps that may be mentioned include: the reduction of a nitro or azido group to an amino group; the hydrolysis of a nitrile group to a carboxylic acid group; and standard nucleophilic aromatic substitution reactions, for example in which an iodo-, preferably, fluoro- or bromo-phenyl group is converted into a cyanophenyl group by employing a source of cyanide ions (e.g. by reaction with a compound which is a source of cyano anions, e.g. sodium, copper (I), zinc or potassium cyanide, optionally in the presence of a palladium catalyst) as a reagent (alternatively, in this case, palladium catalysed cyanation reaction conditions may also be employed).
Other transformations that may be mentioned include: the conversion of a halo group (preferably iodo or bromo) to a 1 -alkynyl group (e.g. by reaction with a 1 - alkyne), which latter reaction may be performed in the presence of a suitable coupling catalyst (e.g. a palladium and/or a copper based catalyst) and a suitable base (e.g. a tri-(Ci.6 alkyl)amine such as triethylamine, tributylamine or ethyldiisopropylamine); the introduction of amino groups and hydroxy groups in accordance with standard conditions using reagents known to those skilled in the art; the conversion of an amino group to a halo, azido or a cyano group, for example via diazotisation (e.g. generated in situ by reaction with NaN02 and a strong acid, such as HCI or H2S04, at low temperature such as at 0°C or below, e.g. at about -5°C) followed by reaction with the appropriate nucleophile e.g. a source of the relevant anions, for example by reaction in the presence of a halogen gas (e.g. bromine, iodine or chlorine), or a reagent that is a source of azido or cyanide anions, such as NaN3 or NaCN; the conversion of -C(0)OH to a -NH2 group, under Schmidt reaction conditions, or variants thereof, for example in the presence of HN3 (which may be formed in by contacting NaN3 with a strong acid such as H2S04), or, for variants, by reaction with diphenyl phosphoryl azide ((PhO)2P(0)N3) in the presence of an alcohol, such as tert-butanol, which may result in the formation of a carbamate intermediate; the conversion of -C(0)NH2 to -NH2, for example under Hofmann rearrangement reaction conditions, for example in the presence of NaOBr (which may be formed by contacting NaOH and Br2) which may result in the formation of a carbamate intermediate; the conversion of -C(0)N3 (which compound itself may be prepared from the corresponding acyl hydrazide under standard diazotisation reaction conditions, e.g. in the presence of NaN02 and a strong acid such as H2S04 or HCI) to -NH2, for example under Curtius rearrangement reaction conditions, which may result in the formation of an intermediate isocyanate (or a carbamate if treated with an alcohol); the conversion of an alkyl carbamate to -NH2, by hydrolysis, for example in the presence of water and base or under acidic conditions, or, when a benzyl carbamate intermediate is formed, under hydrogenation reaction conditions (e.g. catalytic hydrogenation reaction conditions in the presence of a precious metal catalyst such as Pd); halogenation of an aromatic ring, for example by an electrophilic aromatic substitution reaction in the presence of halogen atoms (e.g. chlorine, bromine, etc, or an equivalent source thereof) and, if necessary an appropriate catalyst/Lewis acid (e.g. AICI3 or FeCI3).
Compounds of the invention bearing a carboxyester functional group may be converted into a variety of derivatives according to methods well known in the art to convert carboxyester groups into carboxamides, N-substituted carboxamides, Ν,Ν-disubstituted carboxamides, carboxylic acids, and the like. The operative conditions are those widely known in the art and may comprise, for instance in the conversion of a carboxyester group into a carboxamide group, the reaction with ammonia or ammonium hydroxide in the presence of a suitable solvent such as a lower alcohol, dimethylformamide or a mixture thereof; preferably the reaction is carried out with ammonium hydroxide in a methanol/dimethyl- formamide mixture, at a temperature ranging from about 50°C to about 100°C. Analogous operative conditions apply in the preparation of N-substituted or N,N- disubstituted carboxamides wherein a suitable primary or secondary amine is used in place of ammonia or ammonium hydroxide. Likewise, carboxyester groups may be converted into carboxylic acid derivatives through basic or acidic hydrolysis conditions, widely known in the art. Further, amino derivatives of compounds of the invention may easily be converted into the corresponding carbamate, carboxamido or ureido derivatives.
Compounds of the invention may be isolated from their reaction mixtures using conventional techniques (e.g. recrystallisations).
It will be appreciated by those skilled in the art that, in the processes described above and hereinafter, the functional groups of intermediate compounds may need to be protected by protecting groups.
The need for such protection will vary depending on the nature of the remote functionality and the conditions of the preparation methods (and the need can be readily determined by one skilled in the art). Suitable amino-protecting groups include acetyl, trifluoroacetyl, t-butoxycarbonyl (BOC), benzyloxycarbonyl (CBz), 9-fluorenylmethyleneoxycarbonyl (Fmoc) and 2,4,4-trimethylpentan-2-yl (which may be deprotected by reaction in the presence of an acid, e.g. HCI in water/alcohol (e.g. MeOH)) or the like. The need for such protection is readily determined by one skilled in the art.
The protection and deprotection of functional groups may take place before or after a reaction in the above-mentioned schemes. Protecting groups may be removed in accordance with techniques that are well known to those skilled in the art and as described hereinafter. For example, protected compounds/intermediates described herein may be converted chemically to unprotected compounds using standard deprotection techniques. The type of chemistry involved will dictate the need, and type, of protecting groups as well as the sequence for accomplishing the synthesis.
The use of protecting groups is fully described in "Protective Groups in Organic Synthesis", 3rd edition, T.W. Greene & P.G.M. Wutz, Wiley-lnterscience (1999).
Medical and Pharmaceutical Uses
Compounds of the invention are indicated as pharmaceuticals. According to a further aspect of the invention there is provided a compound of the invention, as hereinbefore defined, for use as a pharmaceutical.
Compounds of the invention may inhibit protein or lipid kinases, such as a PI3 kinase (especially a class I PI3K), for example as may be shown in the tests described below (for example, the test for PI3Ka inhibition described below) and/or in tests known to the skilled person. The compounds of the invention may also inhibit mTOR. Thus, the compounds of the invention may be useful in the treatment of those disorders in an individual in which the inhibition of such protein or lipid kinases (e.g. PI3 , particularly class I PI3K, and/or mTOR) is desired and/or required (for instance compounds of the invention may inhibit PI3K, particularly class I PI3K and, optionally, may also inhibit mTOR).
The term "inhibit" may refer to any measurable reduction and/or prevention of catalytic kinase (e.g. PI3K, particularly class I PI3K, and/or mTOR) activity. The reduction and/or prevention of kinase activity may be measured by comparing the kinase activity in a sample containing a compound of the invention and an equivalent sample of kinase (e.g. PI3K, particularly class I PI3K, and/or mTOR) in the absence of a compound of the invention, as would be apparent to those skilled in the art. The measurable change may be objective (e.g. measurable by some test or marker, for example in an in vitro or in vivo assay or test, such as one described hereinafter, or otherwise another suitable assay or test known to those skilled in the art) or subjective (e.g. the subject gives an indication of or feels an effect). Compounds of the invention may be found to exhibit 50% inhibition of a protein or lipid kinase (e.g. PI3K, such as class I PI3K, and/or mTOR) at a concentration of 100 μΜ or below (for example at a concentration of below 50 μΜ, or even below 10 μΜ, such as below 1 μΜ), when tested in an assay (or other test), for example as described hereinafter, or otherwise another suitable assay or test known to the skilled person.
Compounds of the invention are thus expected to be useful in the treatment of a disorder in which a protein or lipid kinase (e.g. PI3K, such as class I PI3K, and/or mTOR) is known to play a role and which are characterised by or associated with an overall elevated activity of that kinase (due to, for example, increased amount of the kinase or increased catalytic activity of the kinase). Hence, compounds of the invention are expected to be useful in the treatment of a disease/disorder arising from abnormal cell growth, function or behaviour associated with the protein or lipid kinase (e.g. PI3 , such as class I PI3K, and/or mTOR). Such conditions/disorders include cancer, immune disorders, cardiovascular diseases, viral infections, inflammation, metabolism/endocrine function disorders and neurological disorders. The disorders/conditions that the compounds of the invention may be useful in treating hence includes cancer (such as lymphomas, solid tumours or a cancer as described hereinafter), obstructive airways diseases, allergic diseases, inflammatory diseases (such as asthma, allergy and Chrohn's disease), immunosuppression (such as transplantation rejection and autoimmune diseases), disorders commonly connected with organ transplantation, AIDS- related diseases and other associated diseases. Other associated diseases that may be mentioned (particularly due to the key role of kinases in the regulation of cellular proliferation) include other cell proliferative disorders and/or non- malignant diseases, such as benign prostate hyperplasia, familial adenomatosis, polyposis, neurofibromatosis, psoriasis, bone disorders, atherosclerosis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis glomerulonephritis and post-surgical stenosis and restenosis. Other disease states that may be mentioned include cardiovascular disease, stroke, diabetes, hepatomegaly, Alzheimer's disease, cystic fibrosis, hormone- related diseases, immunodeficiency disorders, destructive bone disorders, infectious diseases, conditions associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukaemia, liver disease, pathologic immune conditions involving T cell activation and CNS disorders. As stated above, the compounds of the invention may be useful in the treatment of cancer. More, specifically, the compounds of the invention may therefore be useful in the treatment of a variety of cancer including, but not limited to: carcinoma such as cancer of the bladder, breast, colon, kidney, liver, lung (including non-small cell cancer and small cell lung cancer), esophagus, gall- bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, skin, squamous cell carcinoma, testis, genitourinary tract, larynx, glioblastoma, neuroblastoma, keratoacanthoma, epidermoid carcinoma, large cell carcinoma, non-small cell lung carcinoma, small cell lung carcinoma, lung adenocarcinoma, bone, adenoma, adenocarcinoma, follicular carcinoma, undifferentiated carcinoma, papilliary carcinoma, seminona, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine, rectum, brain and central nervous system, Hodgkin's and leukaemia; hematopoietic tumors of lymphoid lineage, including leukemia, acute lymphocitic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell-lymphoma, Hodgkin's lymphoma, non- Hodgkin's lymphoma, hairy cell lymphoma and Burkett's lymphoma; hematopoietic tumors of myeloid lineage, including acute and chronic myelogenous leukemias, myelodysplastic syndrome and promyelocytic leukemia; tumors of mesenchymal origin, including fibrosarcoma and rhabdomyosarcoma; tumors of the central and peripheral nervous system, including astrocytoma, neuroblastoma, glioma and schwannomas; and other tumors, including melanoma, seminoma, teratocarcinoma, osteosarcoma, xeroderma pigmentosum, keratoxanthoma, thyroid follicular cancer and Kaposi's sarcoma.
Further, the protein or lipid kinases (e.g. PI3K, such as class I PI3K, and/or mTOR) may also be implicated in the multiplication of viruses and parasites. They may also play a major role in the pathogenesis and development of neurodegenerative disorders. Hence, compounds of the invention may also be useful in the treatment of viral conditions, parasitic conditions, as well as neurodegenerative disorders.
Compounds of the invention are indicated both in the therapeutic and/or prophylactic treatment of the above-mentioned conditions.
According to a further aspect of the present invention, there is provided a method of treatment of a disease (e.g. cancer or another disease as mentioned herein) which is associated with the inhibition of protein or lipid kinase (e.g. PI3K, such as class I PI3K, and/or mTOR) is desired and/or required (for example, a method of treatment of a disease/disorder arising from abnormal cell growth, function or behaviour associated with protein or lipid kinases, e.g. PI3K, such as class I PI3K, and/or mTOR), which method comprises administration of a therapeutically effective amount of a compound of the invention, as hereinbefore defined, to a patient suffering from, or susceptible to, such a condition.
"Patients" include mammalian (including human) patients. Hence, the method of treatment discussed above may include the treatment of a human or animal body. The term "effective amount" refers to an amount of a compound, which confers a therapeutic effect on the treated patient. The effect may be objective (e.g. measurable by some test or marker) or subjective (e.g. the subject gives an indication of or feels an effect). Compounds of the invention may be administered orally, intravenously, subcutaneously, buccally, rectally, dermally, nasally, tracheally, bronchially, sublingually, by any other parenteral route or via inhalation, in a pharmaceutically acceptable dosage form. Compounds of the invention may be administered alone, but are preferably administered by way of known pharmaceutical formulations, including tablets, capsules or elixirs for oral administration, suppositories for rectal administration, sterile solutions or suspensions for parenteral or intramuscular administration, and the like. The type of pharmaceutical formulation may be selected with due regard to the intended route of administration and standard pharmaceutical practice. Such pharmaceutically acceptable carriers may be chemically inert to the active compounds and may have no detrimental side effects or toxicity under the conditions of use. Such formulations may be prepared in accordance with standard and/or accepted pharmaceutical practice. Otherwise, the preparation of suitable formulations may be achieved non-inventively by the skilled person using routine techniques and/or in accordance with standard and/or accepted pharmaceutical practice. According to a further aspect of the invention there is thus provided a pharmaceutical formulation including a compound of the invention, as hereinbefore defined, in admixture with a pharmaceutically acceptable adjuvant, diluent and/or carrier. Depending on e.g. potency and physical characteristics of the compound of the invention (i.e. active ingredient), pharmaceutical formulations that may be mentioned include those in which the active ingredient is present in at least 1 % (or at least 10%, at least 30% or at least 50%) by weight. That is, the ratio of active ingredient to the other components (i.e. the addition of adjuvant, diluent and carrier) of the pharmaceutical composition is at least 1 :99 (or at least 10:90, at least 30:70 or at least 50:50) by weight.
The amount of compound of the invention in the formulation will depend on the severity of the condition, and on the patient, to be treated, as well as the compound(s) which is/are employed, but may be determined non-inventively by the skilled person.
The invention further provides a process for the preparation of a pharmaceutical formulation, as hereinbefore defined, which process comprises bringing into association a compound of the invention, as hereinbefore defined, or a pharmaceutically acceptable ester, amide, solvate or salt thereof with a pharmaceutically-acceptable adjuvant, diluent or carrier.
Compounds of the invention may also be combined with other therapeutic agents that are inhibitors of protein or lipid kinases (e.g. PI3K (such as class I PI3K), mTOR, a PI family kinase (e.g. PIM-1 , PI -2 or PIM-3), EGFR and/or MEK) and/or useful in the treatment of a cancer and/or a proliferative disease. Compounds of the invention may also be combined with other therapies. According to a further aspect of the invention, there is provided a combination product comprising:
(A) a compound of the invention, as hereinbefore defined; and
(B) another therapeutic agent that is useful in the treatment of cancer and/or a proliferative disease,
wherein each of components (A) and (B) is formulated in admixture with a pharmaceutically-acceptable adjuvant, diluent or carrier.
Such combination products provide for the administration of a compound of the invention in conjunction with the other therapeutic agent, and may thus be presented either as separate formulations, wherein at least one of those formulations comprises a compound of the invention, and at least one comprises the other therapeutic agent, or may be presented (i.e. formulated) as a combined preparation (i.e. presented as a single formulation including a compound of the invention and the other therapeutic agent).
Thus, there is further provided:
(1) a pharmaceutical formulation including a compound of the invention, as hereinbefore defined, another therapeutic agent that is useful in the treatment of cancer and/or a proliferative disease, and a pharmaceutically-acceptable adjuvant, diluent or carrier; and
(2) a kit of parts comprising components:
(a) a pharmaceutical formulation including a compound of the invention, as hereinbefore defined, in admixture with a pharmaceutically-acceptable adjuvant, diluent or carrier; and
(b) a pharmaceutical formulation including another therapeutic agent that is useful in the treatment of cancer and/or a proliferative disease in admixture with a pharmaceutically-acceptable adjuvant, diluent or carrier, which components (a) and (b) are each provided in a form that is suitable for administration in conjunction with the other.
The invention further provides a process for the preparation of a combination product as hereinbefore defined, which process comprises bringing into association a compound of the invention, as hereinbefore defined, or a pharmaceutically acceptable ester, amide, solvate or salt thereof with the other therapeutic agent that is useful in the treatment of cancer and/or a proliferative disease, and at least one pharmaceutically-acceptable adjuvant, diluent or carrier.
By "bringing into association", we mean that the two components are rendered suitable for administration in conjunction with each other. Thus, in relation to the process for the preparation of a kit of parts as hereinbefore defined, by bringing the two components "into association with" each other, we include that the two components of the kit of parts may be:
(i) provided as separate formulations (i.e. independently of one another), which are subsequently brought together for use in conjunction with each other in combination therapy; or
(ii) packaged and presented together as separate components of a "combination pack" for use in conjunction with each other in combination therapy.
Depending on the disorder, and the patient, to be treated, as well as the route of administration, compounds of the invention may be administered at varying therapeutically effective doses to a patient in need thereof. However, the dose administered to a mammal, particularly a human, in the context of the present invention should be sufficient to effect a therapeutic response in the mammal over a reasonable timeframe. One skilled in the art will recognize that the selection of the exact dose and composition and the most appropriate delivery regimen will also be influenced by inter alia the pharmacological properties of the formulation, the nature and severity of the condition being treated, and the physical condition and mental acuity of the recipient, as well as the potency of the specific compound, the age, condition, body weight, sex and response of the patient to be treated, and the stage/severity of the disease. Administration may be continuous or intermittent (e.g. by bolus injection). The dosage may also be determined by the timing and frequency of administration. In the case of oral or parenteral administration the dosage can vary from about 0.01 mg to about 1000 mg per day of a compound of the invention.
In any event, the medical practitioner, or other skilled person, will be able to determine routinely the actual dosage, which will be most suitable for an individual patient. The above-mentioned dosages are exemplary of the average case; there can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention.
Compounds of the invention may have the advantage that they are effective inhibitors of protein or lipid kinases (e.g. PI3K, such as class I PI3K, and/or mTOR). In an embodiment, compounds of the invention may have the advantage that they are both PI3K (e.g. class I PI3K, such as PI3Ka) inhibitors and mTOR inhibitors, i.e. they may exhibit dual kinase inhibition.
Compounds of the invention may also have the advantage that they may be more efficacious than, be less toxic than, be longer acting than, be more potent than, produce fewer side effects than, be more easily absorbed than, and/or have a better pharmacokinetic profile (e.g. higher oral bioavailability and/or lower clearance) than, and/or have other useful pharmacological, physical, or chemical properties over, compounds known in the prior art, whether for use in the above- stated indications or otherwise.
Compounds of the invention may be beneficial as they are medicaments with targeted therapy, i.e. which target a particular molecular entity by inferring or inhibiting it (e.g. in this case by inhibiting one or more protein or lipid kinases as hereinbefore described). Compounds of the invention may therefore also have the benefit that they have a new effect (for instance as compared to known compounds in the prior art), for instance, the new effect may be a particular mode of action or another effect resultant of the targeted therapy. Targeted therapies may be beneficial as they may have the desired effect (e.g. reduce cancer, by reducing tumor growth or carcinogenisis) but may also have the advantage of reducing side effects (e.g. by preventing the killing of normal cells, as may occur using e.g. chemotherapy).
Furthermore, compounds of the invention may selectively target particular protein or lipid kinases (e.g. the ones described herein) compared to other known protein or lipid kinases (as may be shown experimentally hereinafter). Accordingly, compounds of the invention may have the advantage that certain, specific, cancers may be treated selectively, which selective treatment may also have the effect of reducing side effects.
Compounds of the invention may have the advantage that they may exhibit multiple kinase inhibitory activity. In this respect, advantageously, compounds of the invention may be considered as multi-targeted kinase inhibitors. Compounds of the invention that exhibit single selectivity for a kinase may have the additional benefit that they exhibit less side effects, whereas compounds of the invention that exhibit multiple kinase selectivity may have the additional benefit that they exhibit better potency and/or efficacy.
To date, clinical development of PI3K inhibitors and dual PI3K/mTOR inhibitors have shown moderate activities, suggesting that either more potent/efficacious inhibitors are required or that inhibition of multiple targets or even pathways might be required for effective treatments (see e.g. Bunney, Tom D., Katan, Matilda, Phosphoinositide signalling in cancer: beyond PI3K and PTEN, Nature Reviews Cancer (2010), 10(5), 342-352; Cleary, James M. and Shapiro, Geoffrey I., Development of phosphoinositide-3 kinase pathway inhibitors for advanced cancer, Current Oncology Report (2010), 12, 87-94; and van der Heijden, Michiel S. and Bernards, Rene; Inhibition of the PI3K Pathway: Hope We Can Believe in? Clinical Cancer Research (2010), 16, 3094-3099). The compounds of the invention may have the benefit that they inhibit multiple targets (or even multiple pathways). In this respect, compounds of the invention may be considered to have an improved kinase inhibition cross-reactivity profile, e.g. by being selective against multiple kinases of therapeutic interest, for instance compared to compounds known in the prior art. Compounds of the invention may therefore additionally act on other key kinases, thereby allowing single-agent administration (or, potentially, combination products with reduced dosages) and providing the associated benefits, e.g. reducing the risk of drug-drug interactions, etc.
Examples/Biological Tests
Determination of PI3 kinase activity of compounds of the invention (such as those exemplified) is possible by a number of direct and indirect detection methods. Certain exemplary compounds described herein were prepared, characterized, and assayed for their PI3Ka and mTOR enzymatic activities using the methods described herein. The activities are expressed in IC50 values that range between 1 to 100 nM (**) and 100 nM to 10 μΜ (*), as shown in the Examples below (Table 1). The compounds may also be tested in cell-based assays.
PI3K activity assay
The kinase activity was measured by using the commercial ADP Hunter™ Plus assay available from DiscoveRx (#33-016), which is a homogeneous assay to measure the accumulation of ADP, a universal product of kinase activity. The enzyme, PI3K (p110a/p85o was purchased from Carna Biosciences (#07CBS- 0402A). The assay was done following the manufacturer recommendations with slight modifications: Mainly the kinase buffer was replace by 50 mM HEPES, pH 7.5, 3 mM MgCI2, 100 mM NaCI, 1 mM EGTA, 0.04% CHAPS, 2 mM TCEP and 0.01 mg/ml BGG. The PI3K was assayed in a titration experiment to determine the optimal protein concentration for the inhibition assay. To calculate the IC50 of the ETP-compounds, serial 1 :5 dilutions of the compounds were added to the enzyme at a fixed concentration (2.5 g/ml). The enzyme was preincubated with the inhibitor and 30 μΜ PIP2 substrate (P9763, Sigma) for 5 min and then ATP was added to a final 50 μΜ concentration. Reaction was carried out for 1 hour at 25°C. Reagent A and B were sequentially added to the wells and plates were incubated for 30 min at 37 °C. Fluorescence counts were read in a Victor instrument (Perkin Elmer) with the recommended settings (544 and 580 nm as excitation and emission wavelengths, respectively). Values were normalized against the control activity included for each enzyme (i.e., 100 % PI3 kinase activity, without compound). These values were plotted against the inhibitor concentration and were fit to a sigmoid dose-response curve by using the Graphad software. Cellular Mode of Action
Cell culture: The cell lines are obtained from the American Type Culture Collection (ATCC). U20S (human osteosarcoma) is cultured in Dulbecco's modified Eagle's medium (DME ). PC3 (human prostate carcinoma), MCF7 (human breast cardinoma), HCT116 (human colon carcinoma), 768-0 (human neuroblastoma), U251 (human glyoblastoma) are grown in RPMI. All media are supplemented with 10% fetal bovine serum (FBS) (Sigma) and antibiotics- antimycotics. Cells are maintained in a humidified incubator at 37°C with 5% C02 and passaged when confluent using trypsin/EDTA.
U2foxRELOC and U2nesRELOC assay: The U2nesRELOC assay and the U2foxRELOC assay have been described. Briefly, cells are seeded at a density of 1.0*105 cells/ml into black-wall clear-bottom 96-well microplates (BD Biosciences). After incubation at 37°C with 5% C02for 12 hours, 2μΙ of each test compound are transferred from the mother plates to the assay plates. Cells are incubated in the presence of the compounds for one hour. Then cells are fixed and the nucleus stained with DAPI (Invitrogen). Finally the plates are washed with X PBS twice and stored at 4°C before analysis. Image acquirement and processing: Assay plates are read on the BD Pathway™ 855 Bioimager equipped with a 488/10 nm EGFP excitation filter, a 380/10 nm DAPI excitation filter, a 515LP nm EGFP emission filter and a 435LP nm DAPI emission filter. Images are acquired in the DAPI and GFP channels of each well using 10x dry objective. The plates are exposed 0.066 ms (Gain 31) to acquire DAPI images and 0.55 ms (Gain 30) for GFP images.
Data analysis: The BD Pathway Bioimager outputs its data in standard text files. Data are imported into the data analysis software BD Image Data Explorer. The nuclear/cytoplasmic (Nuc/Cyt) ratios of fluorescence intensity are determined by dividing the fluorescence intensity of the nucleus by the cytoplasmic. A threshold ratio of greater than 1.8 is employed to define nuclear accumulation of fluorescent signal for each cell. Based on this procedure we calculate the percentage of cells per well displaying nuclear translocation or inhibition of nuclear export. Compounds that induce a nuclear accumulation of the fluorescent signal greater than 60% of that obtained from wells treated with 4nM LMB are considered as hits. In order to estimate the quality of the HCS assay, the Z' factor is calculated by the equation: Z* = 1 - [(3 χ std. dev. of positive controls) + (3 χ std. dev. of negative controls) / (mean of positive controls) - (mean of negative controls)].
PI3K signalling
AKT phosphorylation Inhibition.Western Blot Analysis: Subconfluent cells are incubated under different conditions and are washed twice with TBS prior to lysis. Lysis buffer is added containing 50 mM Tris HCI, 150 mM NaCI, 1% NP-40, 2mM Na3V04, 100 mM NaF, 20 mM Na„P207 and protease inhibitor cocktail (Roche Molecular Biochemicals). The proteins are resolved on 10% SDS-PAGE and are transferred to nitrocellulose membrane (Schleicher & Schuell, Dassel, Germany). The membranes are incubated overnight at 4°C with antibodies specific for Akt, phospho-Ser-473-Akt (Cell Signaling Technology) and ct-tubulin (Sigma), they are washed and then incubated with IRDye800 conjugated anti- mouse and Alexa Fluor 680 goat anti-rabbit IgG secondary antibodies. The bands are visualized using an Odyssey infrared imaging system (Li-Cor Biosciences).
Cytotoxicity assessment
The compounds are tested on 96-well trays. Cells growing in a flask are harvested just before they became confluent, counted using a haemocytometer and are diluted down with media adjusting the concentration to the required number of cells per 0.2 ml (volume for each well). Cells are then seeded in 96- well trays at a density between 1000 and 4000 cells/well, depending of the cell size. Cells are left to plate down and grow for 24 hours before adding the drugs. Drugs are weighed out and diluted with DMSO to get them into solution to a concentration of 10mM. From here a "mother plate" with serial dilutions is prepared at 200X the final concentration in the culture. The final concentration of DMSO in the tissue culture media should not exceed 0.5%. The appropriate volume of the compound solution (usually 2 microlitres) is added automatically (Beckman FX 96 tip) to media to make it up to the final concentration for each drug. The medium is removed from the cells and replaced with 0.2 ml of medium dosed with drug. Each concentration is assayed in triplicate. Two sets of control wells are left on each plate, containing either medium without drug or medium with the same concentration of DMSO. A third control set is obtained with the cells untreated just before adding the drugs (seeding control, number of cells starting the culture). Cells are exposed to the drugs for 72 hours and then processed for MTT colorimetric read-out. mTOR assay
The enzymatic mTOR activity was measured using a LanthaScreen™ kinase activity assay (Invitrogen). The enzyme was purchased from Invitrogen (PV4754), as well as the GFP-labeled substrate (4EBP1-GFP; PV4759) and the Tb-anti- p4EBP1(pThr46) antibody (PV4757). The assay was performed in 50 mM HEPES buffer, pH 7.5, containing 1.5 mM MnCI2, 10 mM MgCI2, 1 mM EGTA, 2.5 mM DTT and 0.01% Tween-20. The concentration of the assay components were the following: 0.24 nM mTOR kinase, 400 nM 4EBP1-GFP, 10 mM ATP and serial dilutions of the compound (inhibitor) to be evaluated. After 1 h incubation at room temperature, 20 mM EDTA was used to stop the reaction and terbium- labeled antibody (4 nM) added to detect phosphorylated product. The antibody associates with the phosphorylated product resulting in an increased TR-FRET value. The TR-FRET value (a dimensionless number) was calculated as the ratio of the acceptor signal (GFP, emission at 520 nm) to the donor signal (terbium, emission at 495 nm). Values were plotted against the inhibitor concentration and fitted to a sigmoid dose-response curve using GraphPad software PI3K cellular activity (Elisa assay)
Activity was measured as endogenous levels of phospho-Aktl (Ser473) protein. Osteosarcoma U20S cells were plated in 96 Poly-D-Lysine coating tissue culture 10 plates (18.000 cells/well). After the treatment with serial dilutions of the compound during 3h, the cells were fixed directly in the wells with 4% paraformaldehyde. After fixing, individual wells go through the same series of steps used for a conventional immunoblot: including blocking with 5% BSA, incubation with 15 1/1000 of primary antibody-AKT (Ser 74) in PBS containing 5% BSA at 4°C overnight (Cell Signalling), washing and incubation with second antibody HRPanti-mouse IgG for 1h at RT (Amersham). After the addition of SuperSignal ELISA Femto maximum sensitivity chemiluminescent substrate (Pierce) the results were read using a luminescence plate reader (Victor).
The invention is illustrated by way of the following examples, in which the following abbreviations (or chemical symbols) may be employed:
"DCM" means dichloromethane, "CHCI3" means chloroform, "MeOH" means methanol, "EtOH" means ethanol, "EtOAc" means ethyl acetate, "THF" means tetrahydrofuran, "ACN" means acetonitrile, "DMAP" means 4,4- dimethylaminopyridine, "DMF" means dimethylformamide, "DME" means dimethoxyethane, "DMSO" means dimethylsulfoxide, "Et20" means diethyl ether, "Hex" means hexane, "EtOAc" means ethyl acetate, "BA/BE" means boronic acid/ester, "Pd(PPh3)4" means tetrakis(triphenylphosphine)palladium, "Pd(Ph3P)2CI2" means dichlorobis(triphenylphosphine)palladium(ll),
"Pd(dppf)CI2.DCM" means 1 ,1'-bis(diphenylphosphino)ferrocenepalladium(ll) dichloride, dichloromethane complex, "NIS" means N-iodosuccinimide, "Na2S04" means disodium sulphate, "MgS04" means magnesium sulphate, "K2C03" means dipotassium carbonate, "Na2C03" means disodium carbonate, "NaHC03" means sodium bicarbonate, "TEA" means triethylamine, "TFA" means trifluoroacetic acid, "TsCI" means toluenesulfonyl chloride, "sat." means saturated, "aq." means aqueous, "HPLC" means high performance liquid chromatography, ' means retention time, "MS" means mass spectrometry, "TLC" means thin layer chromatography, "Rf" means retardation factor, "g" means gram(s), "mmol" means millimole(s), "eq" means equivalent(s), "ml." means milliliter(s), "min" means minute(s), "h" means hour(s), "RT" means room temperature.
Analytical analysis. NMR spectra were recorded on a Bruker Avance II 300 spectrometer and Bruker Avance II 700 spectrometer fitted with 5mm QXI 700 S4 inverse phase, Z-gradient unit and variable temperature controller. The HPLC measurements were performed using a HP 1100 from Agilent Technologies comprising a pump (binary) with degasser, an autosampler, a column oven, a diode-array detector (DAD) and a column as specified in the respective methods below. Flow from the column was split to a MS spectrometer. The MS detector was configured with an electrospray ionization source or API/APCI. Nitrogen was used as the nebulizer gas. Data acquisition was performed with ChemStation LC/MSD quad, software. HPLC- ethod 1
Reversed phase HPLC was carried out on a Gemini-NX C18 (100 x 2.0 mm; 5um), Solvent A: water with 0.1% formic acid; Solvent B: acetonitrile with 0.1% formic acid. Gradient: 5% of B to 100% of B within 8 min at 50 °C, DAD.
HPLC-Method 2
Reversed phase HPLC was carried out on a Gemini-NX C18 (100 x 2.0 mm; 5um), Solvent A: water with 0.1 % formic acid; Solvent B: acetonitrile with 0.1 % formic acid. Gradient: 50% of B to 100% of B within 8 min at 50 °C, DAD.
HPLC-Method 3
Reversed phase HPLC was carried out on a Gemini-NX C18 (100 x 2.0 mm; 5um), Solvent A: water with 0.1 % formic acid; Solvent B: acetonitrile with 0.1 % formic acid. Gradient: 5% of B to 40% of B within 8 min at 50 °C, DAD.
HPLC-Method 4
Reversed phase HPLC was carried out on a Gemini C18 column (50 x 2 mm, 3 urn); Solvent A: water with 0.1 % formic acid; Solvent B: acetonitrile with 0.1 % formic acid. Gradient: 10-95 % of B within 4 min at a flow rate of 0.5 mlJmin followed by 2 min of 100 % of B at 0.8 mlJmin, controlled temperature at 50 °C, DAD.
HPLC-Method 5
Reversed phase HPLC was carried out on a Gemini C18 column (50 x 2 mm, 3 urn); Solvent A: water with 10mM ammonium bicarbonate; Solvent B: acetonitrile. Gradient: 20-100 % of B within 3 min at a flow rate of 0.5 mL/min followed by 2 min of 100 % of B at 0.8 mL/min, controlled temperature at 40 °C, DAD.
HPLC-Method 6
Reversed phase HPLC was carried out on a Gemini-NX C 8 (100 x 2.0 mm; 5um), Solvent A: water with 0.1% formic acid; Solvent B: acetonitrile with 0.1 % formic acid. Gradient: 0% of B to 30% of B within 8 min at 50 °C, DAD. "Found mass" refers to the most abundant isotope detected in the HPLC-MS.
Compound preparation
The compound names given above were generated in accordance with lUPAC using the AutoNom naming program in MDL ISIS Draw. Intermediate 1
3-Amino-6,7-dihydro-4H-thieno[3,2-c]pyridine-2,5-dicarboxylic acid 5-tert- butyl ester 2-methyl ester
To a solution of fert-butyl 5-cyano-4-hydroxy-3,6-dihydropyridine-1(2H)- carboxylate (1.74 g, 7.76 mmol) and TEA (1.6 mL, 11.64 mmol) in dry DCM (40 mL), MsCI (0.66 mL, 8.54 mmol) was added at 0°C under Ar atmosphere. After stirring for 15 min at 0°C and 30 min at RT, a previously mixed solution of methyl thioglycolate (0.76 mL, 8.54 mmol) and sodium methoxide (62 mL of 0.5N sol in MeOH) was added. The reaction mixture was stirred at RT for 1 h and more sodium methoxide (31 mL of 0.5N sol in MeOH) was added. The mixture was stirred at RT for 2h and evaporated. The residue was partitioned between water and DCM, and extracted twice with DCM. The organics were dried, filtered and evaporated to afford the desired product (1.40 g, 58%) as a brown solid. It was used in the next experiment without further purification. 1H NMR (300 MHz, CDCI3) δ 5.31 (s, 2H), 4.22 (s, 2H), 3.79 (s, 3H), 3.67 (m, 2H), 2.73 (t, J = 5.5 Hz, 2H), 1.48 (s, 9H) ppm.
Intermediate 2
3-Amino-6,7-dihydro-4H-thieno[3,2-c]pyridine-2,5-dicarboxylic acid 5-ethyl ester 2-methyl ester
A solution of 3-amino-6,7-dihydro-4H-thieno[3,2-c]pyridine-2,5-dicarboxylic acid 5-tert-butyl ester 2-methyl ester (0.710 g, 2.27 mmol) in dry DCM (5 mL) and TFA (1.5 mL) was stirred at RT for 2h. The mixture was evaporated and the residue was taken up in sat aq NaHC03 and extracted with CHCl3/i-PrOH 1 :1. The combined organic phases were dried, filtered and evaporated. The residue (0.443 g) was dissolved in dry DCM (10 mL) and treated with TEA (1 mL) and ethyl chloroformate (0.26 mL) at RT overnight. After quenching with sat aq NaHC03 the product was extracted with DCM, washed with 1M aq HCI and brine/NaHC03, dried, filtered and evaporated to give the desired product (0.423 g, 66%). 1H NMR (300 MHz, CDCI3) δ 5.28 (s, 2H), 4.22 (s, 2H), 4.12 (q, J = 7.1 Hz, 2H), 3.75 (s, 3H), 3.67 (m, 2H), 2.70 (t, J = 5.4 Hz, 2H), 1.22 (t, J = 7.1 Hz, 3H) ppm. Intermediate 3
3-(3-Methoxy-phenyl)-1-oxo-1,2,7,8-tetrahydro-5H-9-thia-2,4,6-triaza- fluorene-6-carboxylic acid ethyl ester
3-Amino-6,7-dihydro-4H-thieno[3,2-c]pyridine-2,5-dicarboxylic acid 5-ethyl ester 2-methyl ester (315 mg, 1.11 mmol) and 3-methoxybenzonitrile (0.27 mL, 2.22 mmol) were placed in a pressure tube, and 4M HCI/dioxane (5 mL) was added dropwise. The tube was closed and left in ultrasonic bath for 0.5h at RT, then heated at 130°C with stirring during the weekend (shows mainly open intermediate). The mixture was evaporated and the residue was taken in dry toluene and treated with TEA (0.5 mL). The reaction mixture was refluxed for 4h and stirred at RT overnight. Et20 was added and a precipitate was filtered off and washed with Et20. The solid was purified by column chromatography (EtOAc:cyclohexane, 30:70 to 100:0; and MeOH:DCM 0:100 to 10:90) to give the desired product (0.100 g, 23%).
Intermediate 4
3-(3-Methoxy-phenyl)-1-morpholin-4-yl-7,8-dihydro-5H-9-thia-2,4,6-triaza- fluorene-6-carboxylic acid ethyl ester
p-Toluenesulfonyl chloride (0.130 g, 0.680 mmol, 2.6 eq) and a catalytic amount of DMAP (few mg) were added to a previously sonicated suspension of 3-(3- methoxy-phenyl)-1-oxo-1 ,2,7,8-tetrahydro-5H-9-thia-2,4,6-triaza-fluorene-6- carboxylic acid ethyl ester (0.100 g, 0.259 mmol, 1 eq) and TEA (0.18 mL, 1.30 mmol, 5 eq) in dry acetonitrile (2 mL). The reaction mixture was stirred overnight. Then morpholine (0.10 mL, 1.1 mmol, 4.2 eq) was added, and stirring was continued at 45-50°C for 3h. The precipitate which had formed was filtered off, washed with CH3CN and dried. The crude (0.068 g) was purified by flash chromatography (Si02; DCM to DCM/MeOH 95:5) affording the desired product (0.040g, 34 %). HPLC-MS (Method 4): tR= 6.25 min, [M+H]+ m/z 455.2, 1H NMR (300 MHz, CDCI3) δ 8.08 - 7.93 (m, 2H), 7.61 (s, 1H), 7.31 (t, J = 7.9 Hz, 1 H), 7.04 (d, J = 4.8 Hz, 1 H), 6.97 - 6.87 (m, 1 H), 4.76 (s, 2H), 4.16 (q, J = 7.1 Hz, 2H), 3.94 (d, J = 5.1 Hz, 4H), 3.82 (d, J = 12.8 Hz, 9H), 2.87 (s, 2H) ppm. Example 1
3-(3-Hydroxy-phenyl)-1-morpholin-4-yl-7,8-dihydro-5H-9-thia-2,4,6-triaza- fluorene-6-carboxylic acid ethyl ester
1 BBr3/DC (1 mL, 10 eq) was added at -15°C (ice in MeOH) to a suspension of 3-(3-methoxy-phenyl)-1 -morpholin-4-yl-7,8-dihydro-5H-9-thia-2,4,6-triaza- fluorene-6-carboxylic acid ethyl ester (0.040 g, 0.09 mmol, 1 eq) in dry DCM (1 mL). The reaction mixture was kept in the freezer (-20°C) for 2.5 days and was then poured onto ice and extracted with DCM. The organic phase was concentrated, and the resulting crude was purified by flash chromatography (Si02; DCM/MeOH 98:2 to 96:4) affording the desired product (0.015 g, 34 %). HPLC-MS (Method 1 ): t*= 5.14 min, [M+H]+ m/z 441.2, 1H NMR (300 MHz, DMSO) δ 9.52 (s, 1 H), 7.86 (d, J = 7.2 Hz, 2H), 7.28 (t, J = 8.1 Hz, 1 H), 6.93 - 6.81 (m, 1 H), 4.68 (s, 2H), 4.14 (q, J = 7.1 Hz, 2H), 3.96 (d, J = 5.0 Hz, 4H), 3.80 (d, J = 4.8 Hz, 6H), 2.97 (s, 2H), 1.24 (t, J - 7.1 Hz, 4H) ppm.
Example 2
3-(1-Morpholin-4-yl-5,6,7,8-tetrahydro-9-thia-2,4,6-triaza-fluoren-3-yl)-phenol
A suspension of 3-(3-hydroxy-phenyl)-1-morpholin-4-yl-7,8-dihydro-5H-9-thia- 2,4,6-triaza-fluorene-6-carboxylic acid ethyl ester (0.015 g, 0.034 mmol, 1 eq) and lithium hydroxide hydrate (0.014 g, 0.34 mmol, 10 eq) in MeOH/i-PrOH 1 :1 (1 mL) was heated at 160°C for 1.5 h in a microwave oven. Volatiles were removed and the residue taken up in aq NaHC03. A solid formed and was filtered off, washed with water and ether, dried and purified (together with the crude resulting from extraction of the filtrate) by prep HPLC affording the desired product (0.002 g, 16 %). HPLC-MS (Method 1 ): t*= 2.38 min, [M+H]+ m/z 369.1. 1H NMR (300 MHz, DMSO) δ 8.24 (s, 1 H), 7.85 (m, 2H), 7.26 (m, 1 H), 6.85 (m, 1 H), 4.01 (s, 2H), 3.95 (m, 4H), 3.79 (m, 4H), 3.12 (m, 2H), 2.86 (m, 2H) ppm.
Intermediate 5
3-Ureido-6,7-dihydro-4H-thieno[3,2-c]pyridine-2,5-dicarboxylic acid 5-ethyl ester 2-methyl ester
A solution of 3-amino-6,7-dihydro-4H-thieno[3,2-c]pyridine-2,5-dicarboxylic acid 5-ethyl ester 2-methyl ester (0.488 g, 1.72 mmol, 1 eq) in dichloromethane (7.8 mL) was cooled at -78 °C. Chlorosulfonyl isocyanate (0.26 ml, 2.92 mmol, 1.7 eq) was added, and the reaction mixture was slowly warmed to RT and stirred for additional 45 min. The reaction mixture was concentrated, and the residue was taken up in 6N aq. HCI and stirred at 100 °C for 20 min, then neutralized with sat aq NaHC03 at RT. The suspension was filtered and the solid rinsed with water and dried to afford the desired product (0.431 g, 77%) as a white solid. 1H NMR (300 MHz, DMSO) δ 8.57 (s, 1 H), 6.51 (s, 2H), 4.30 (m, 2H), 4.08 (q, J = 7.1 Hz, 2H), 3.76 (s, 3H), 3.66 (m, 2H), 2.82 (t, J = 5.5 Hz, 2H), 1.19 (t, J = 7.1 Hz, 3H) ppm.
Intermediate 6
1 ,3-Dioxo-1 ,2,3,4,7,8-hexahydro-5H-9-thia-2,4,6-triaza-f luorene-6-carboxylic acid ethyl ester
To a solution of 3-ureido-6,7-dihydro-4H-thieno[3,2-c]pyridine-2,5-dicarboxylic acid 5-ethyl ester 2-methyl ester (0.431 g, 1.317 mmol, 1 eq) in methanol (8.3 mL) was added 1.5 M aq. KOH (3.63 mL) at RT. The reaction mixture was refluxed for 3.5 h. After cooling, the reaction mixture was quenched by addition of HCI (6N solution, 0.7 mL; up to pH 5). The solid was filtered off, rinsed with water and dried to afford the desired product (0.292 g, 75%) as an orange solid.
Intermediate 7
1-Morpholin-4-yl-3-(toluene-4-sulfonyloxy)-7,8-dihydro-5H-9-thia-2,4,6-triaza- fluorene-6-carboxylic acid ethyl ester
To a solution of 1 ,3-dioxo-1 ,2,3,4,7, 8-hexahydro-5H-9-thia-2,4,6-triaza-fluorene- 6-carboxylic acid ethyl ester (2.06 g, 6.97 mmol) and TEA (2.43 mL, 17.44 mmol) in DCM (100 mL) was added p-toluenesulfonyl chloride (3.0 g, 15.35 mmol) and 4-dimethylaminopyridine (0.085 g, 0.7 mmol). The reaction mixture was stirred at RT for 12 h. Morpholine (0.92 mL, 10.46 mmol) was added, and stirring was continued for 2 h. The solvent was removed under reduced pressure, and the residue was taken up in DCM and washed with sat aq NaHC03. The organic layer was dried with Na2S04, filtered and evaporated. The residue was purified by column chromatography (EtOAc:cyclohexane, 10:90 to 60:40) to afford the desired product (1.95 g, 54 %) as a white solid. HPLC-MS (Method 4): tR= 4.81 min, [M+H]+ m/z 519.0. 1H NMR (300 MHz, DMSO) δ 7.93 (d, J = 8.3 Hz, 2H), 7.50 (d, J = 8.2 Hz, 2H), 4.46 (s, 2H), 4.13 (q, J = 7.1 Hz, 2H), 3.75 (t, J = 5.5 Hz, 2H), 3.67 (m, 8H), 2.93 (m, 2H), 2.44 (s, 3H), 1.23 (t, J = 7.1 Hz, 3H) ppm. Example 3
3-(2-Amino-pyrimidin-5-yl)-1-morpholin^-yl-7,8-dihydro-5H-9-thia-2,4,6- triaza-fluorene-6-carboxylic acid ethyl ester
To a mixture of 1-morpholin-4-yl-3-(toluene-4-sulfonyloxy)-7,8-dihydro-5H-9-thia- 2,4,6-triaza-fluorene-6-carboxylic acid ethyl ester (0.1 g, 0.193 mmol, 1 eq), Pd(dppf)CI2.DCM (0.016 g, 0.019 mmol, 0.1 eq) and 2-aminopyrimidine-5-boronic acid pinacol ester (0.064 g, 0.289 mmol, 1.6 eq) in D E (2.5 mL) was added Na2C03 (0.62 mL, 2.0 M solution in water). The reaction mixture was heated in the microwave oven at 130 °C for 1 h. On cooling, the mixture was concentrated, and the residue was redissolved in DCM and washed with water. The organic layer was dried over Na2S04, filtered and concentrated. The residue was purified by column chromatography (MeOH:DCM, 1 :99 to 7:93) to afford the desired product (0.066 g, 77%) as a yellow solid. HPLC-MS (Method 1): t*= 4.58 min, [M+H]+ m/z 442.2. 1H NMR (300 MHz, DMSO) δ 9.12 (s, 2H), 7.11 (s, 2H), 4.65 (s, 2H), 4.13 (q, J = 7.0 Hz, 2H), 3.92 (m, 4H), 3.74 (m, 6H), 2.94 (m, 2H), 1.23 (t, J = 7.1 Hz, 3H) ppm.
Example 4
5-(1-Morpholin-4-yl-5,6,7,8-tetrahydro-9-thia-2,4,6-triaza-fluoren-3-yl)- pyrimidin-2-ylamine
3-(2-Amino-pyrimidin-5-yl)-1-morpholin-4-yl-7,8-dihydro-5H-9-thia-2,4,6-triaza- fluorene-6-carboxylic acid ethyl ester (0.050 g, 0.11 mmol) was suspended in MeOH/i-PrOH (1 mL, 1 :1) and treated with lithium hydroxide monohydrate (71 mg, 1.70 mmol). The reaction mixture was heated under microwave conditions at 160 °C for 1 h. On cooling, the reaction mixture was concentrated, and the residue was suspended in a mixture of acetonitrile and methanol (1:1). The solid was filtered off, rinsed with methanol and dried to afford the desired product (0.040 g, 95%). HPLC-MS (Method 1): t*= 0.42 and 2.20 min, [M+H]+ m/z 370.0. H NMR (300 MHz, DMSO) δ 9.12 (s, 2H), 7.07 (s, 2H), 3.93 (m, 6H), 3.76 (m, 4H), 3.06 (m, 2H), 2.80 (m, 2H) ppm. Example 5
3-(2-Amino-pyrimidin-5-yi)-lHiiorpholin-4-yl-7,8-dihydro-5H-9-thia-2,4,6- triaza-fluorene-6-carboxylic acid ethylamide
5-(1-Morpholin-4-yl-5,6,7,8-tetrahydro-9-thia-2,4,6-tri^
ylamine (0.035 g, 0.08 mmol) was suspended in acetonitrile (1.5 mL) and treated with ethyl isocyanate (0.008 mL, 0.095 mmol) and N,N-diisopropylethylamine (0.035 mL, 0.198 mmol). The reaction mixture was stirred at RT overnight. More ethyl isocyanate (0.02 mL) was added and the reaction mixture was stirred at RT overnight. The reaction mixture was diluted with EtOAc and washed with water. The organic layer was dried over Na2S04, filtered and evaporated. The residue was purified by column chromatography (MeOH:DCM, 0:100 to 10:90) to afford the desired product (0.015 g, 45%) as a white solid. HPLC-MS (Method 1):
Figure imgf000073_0001
3.84 min, [M+H]+ m/z 441.3. 1H NMR (300 MHz, DMSO) δ 9.16 (s, 2H), 7.11 (s, 2H), 6.77 (m, 1H), 4.58 (s, 2H), 3.93 (m, 4H), 3.75 (m, 6H), 3.10 (m, 2H), 2.89 (m, 2H), 1.04 (t, J = 7.1 Hz, 3H) ppm.
Example 6
5-[6-(4-Fluoro-benzenesulfonyl)-1-morpholin-4-yl-5,6l7,8-tetrahydro-9-thia- 2,4,6-triaza-fluoren-3-yl]-pyrimidin-2 -ylamine
5-(1-Morpholin-4-yl-5,6,7l8-tetrahydro-9-thia-2,4,6-triaza-fluoren-3-yl)-pyrimidin-2- ylamine (0.050 g, 0.14 mmol) was suspended in acetonitrile (1.5 mL) and treated with 4-fluorobenzenesulfonyl chloride (0.032 g, 0.16 mmol) and N,N- diisopropylethylamine (0.06 mL, 0.34 mmol). The reaction mixture was stirred at RT overnight and then diluted with acetonitrile: methanol (1:1, 2 mL). The solid that had formed was filtered off and rinsed with Et20 to afford the desired product (0.049 g, 69%) as a white solid. HPLC-MS (Method 1): t«= 5.14 min, [M+H]+ m/z 528.0. H NMR (300 MHz, DMSO) δ 9.14 (s, 2H), 7.98 (m, 2H), 7.47 (m, 2H), 7.12 (s, 2H), 4.36 (s, 2H), 3.91 (m, 4H), 3.76 (m, 4H), 3.52 (m, 2H), 2.99 (m, 2H) ppm.
Example 7
[3-(2-Amino-pyrimidin-5-yl)-1-morpholin-4-yl-7,8-dihydro-5H-9-thia-2,4,6- triaza-fluoren-6-yl]-(4-fluoro-phenyl)-methanone
5-(1-Morpholin-4-yl-5,6,7,8-tetrahydro-9-thia-2,4,6-triaza-fluoren-3-yl)-pyrimidin-2- ylamine (0.050 g, 0.14 mmol, 1 eq) was suspended in acetonitrile (1.5 mL) and treated with 4-fluorobenzoyl chloride (0.026 g, 0.162 ,mmol, 1.2 eq) and N,N- diisopropylethylamine (0.059 mL, 0.338 mmol, 2.5 eq). The reaction mixture was stirred at RT overnight and then diluted with DCM and washed with water and sat. aq. NaHC03. The organic layer was dried over Na2S04, filtered and concentrated. The residue was purified by column chromatography (MeOH:DCM, 0:100 to 10:90) to afford the desired product (0.034 g, 52%) as a yellow solid. HPLC-MS (Method 1): .«= 4.70 min, [M+H]+ m/z 492.0. H NMR (300 MHz, DMSO) δ 9.11 (s, 2H), 7.59 (dd, J = 8.7, 5.5 Hz, 2H), 7.32 (t, J = 8.9 Hz, 2H), 7.02 (s, 2H), 4.80 (s, 2H), 3.94 (m, 4H), 3.78 (m, 6H), 3.03 (m, 2H) ppm.
Example 8
1 -[3-{2-Amino-pyrimidin-5-yl)-1 -morp
triaza-fluoren-6-yl]-ethanone
S-il-Morpholin^- l-S.e .S-tetrah dro-S-thia^^.e-triaza-fluoren-S-y -pyrimidin^- ylamine (0.050 g, 0.14 mmol, 1 eq) was suspended in acetonitrile (1.5 mL) and treated with acetyl chloride (0.012 ml, 0.162 mmol, 1.2 eq) and N,N- diisopropylethytamine (0.059 ml, 0.338 mmol, 2.5 eq). The reaction mixture was stirred at RT overnight and then diluted with DCM and washed with water and sat. aq. NaHC03. The organic layer was dried over Na2S04, filtered and concentrated. The residue was purified by column chromatography (MeOH:DCM, 0:100 to 15:85) to afford the desired product (0.012 g, 22%) as a white solid. HPLC-MS (Method 1): t«= 3.65 min, [M+H]+ m/z 412.0. H NMR (300 MHz, DMSO) δ 9.14 (s, 2H), 7.11 (s, 2H), 4.71 (m, 2H), 3.93 (m, 4H), 3.84 (m, 2H), 3.78 (m, 4H), 3.02 (m, 1 H), 2.90 (m, 1 H), 2.16 (s, 3H) ppm.
Example 9
5-(6-Ethyl-1-morpholin-4-yl-5,6,7.8-tetrahydro-9-thia-2,4,6-triaza-fluoren-3- yl)-pyrimidin-2-ylamine
5-(1-Morpholin-4-yl-5,6,7,8-tetrahydro-9-thia-2,4,6-triaza-fluoren-3-yl)-pyrimidin-2- ylamine (0.050 g, 0.14 mmol, 1 eq) and acetaldehyde (0.009 ml, 0.162 mmol, 1.2 eq) in methanol (2 mL) was treated with sodium cyanoborohydride (0.013 g, 0.203 mmol, 1.5 eq) followed by acetic acid (0.023 mL, 0.406 mmol, 3 eq). The reaction mixture was stirred at RT overnight. The solvent was removed under reduced pressure, and the residue was suspended in sat aq NaHC03 and extracted with DCM. The organic layer was dried over Na2S04 and evaporated. The residue was purified by column chromatography (MeOH:DCM, 0:100 to 10:90) to afford the desired product (0.038 g, 72%) as a yellow solid. HPLC-MS (Method 1): t«= 0.44 and 2.41 min, [M+HJ+ m/z 398.0. 1H NMR (300 MHz, DMSO) δ 9.12 (s, 2H), 7.08 (s, 2H), 3.92 (m, 4H), 3.76 (m, 4H), 3.65 (s, 2H), 2.93 (m, 2H), 2.83 (m, 2H), 2.63 (q, J = 7.1 Hz, 2H), 1.13 (t, J - 7.1 Hz, 3H) ppm.
Example 10
5-[6-(4-Fluoro-benzyl)-1-morpholin-4-yl-5,6,7,8-tetrahydro-9-thia-2(4,6-triaza- fluoren-3-yl]-pyrimidin-2-ylamine
A solution of S-i -morpholin^-yl-S.ej.S-tetrahydro-g-thia^^.e-triaza-fluoren-S- yl)-pyrimidin-2-ylamine (0.050 g, 0.14 mmol, 1 eq) and 4-fluorobenzaldehyde (0.018 mL, 0.162 mmol, 1.2 eq) in MeOH (2 mL) was treated with sodium cyanoborohydride (0.013 g, 0.203 mmol, 1.5 eq) followed by acetic acid (0.023 mL. 0.406 mmol, 3 eq). The reaction mixture was stirred at RT overnight. Additional 4-fluorobenzaldehyde (1.2 eq), sodium cyanoborohydride (1.5 eq) and acetic acid (3 eq) were added, and the reaction mixture was stirred at RT for 12 h. The solvent was removed under reduced pressure, and the residue was suspended in sat. aq. NaHC03, and extracted with DCM. The organic layer was dried over Na2S04, filtered and evaporated. The residue was purified by column chromatography (MeOH: DCM, 0:100 to 10:90) to afford the desired product (0.048 g, 74%) as a white solid. HPLC-MS (Method 1): t*= 2.93 and 3.04 min, [M+H]+ m/z 478.0. 1H NMR (300 MHz, DMSO) δ 9.07 (s, 2H), 7.43 (dd, J = 8.3, 5.8 Hz, 2H), 7.18 (t, J = 8.8 Hz, 2H), 7.08 (s, 2H), 3.91 (m, 4H), 3.76 (m, 6H), 3.68 (s, 2H), 2.92 (m, 2H), 2.83 (m, 2H) ppm.
Example 11
3- (2-Amino-pyrimidin-5-yl)-1-morpholin-4-yl-7,8-dihydro-5H-9-thia-2,4,6- triaza-fluorene-6-carboxylic acid (4-fluoro-phenyl)-amide
5-(1-Morpholin-4-yl-5,6,7,8-tetrahydro-9-thia-2,4,6-triaza-fluoren-3-yl)-pyrimidin-2- ylamine (0.050 g, 0.14 mmol) was suspended in ACN (1.5 mL) and treated with
4- fluorophenyl isocyanate (0.018 mL, 0.16 mmol) and N,N-diisopropylethylamine (0.059 mL, 0.34 mmol). The reaction mixture was stirred at RT overnight. The solvent was removed under reduced pressure, and the residue was purified by column chromatography (MeOH:DCM, 0:100 to 10:90) to afford the desired product (0.015 g 22%) as a white solid. HPLC-MS (Method 1): \R= 5.33 min, [M+H]+ m/z 507.0. 1H NMR (300 MHz, DMSO) δ 9.14 (s, 2H), 8.85 (s, 1 H), 7.49 (dd, J = 9.1 , 5.1 Hz, 2H), 7.08 (m, 4H), 4.76 (s, 2H), 3.94 (m, 4H), 3.84 (m, 2H), 3.78 (m, 4H), 2.99 (m, 2H) ppm. Intermediate 8
3-Amino-4,6-dihydro-thieno[2,3-c]pyrrole-2,5-dicarboxylic acid 5-tert-butyl ester 2-methyl ester
To a solution of 1-Boc-3-cyano-4-oxopyrrolidine (2.0 g, 9.51 mmol) and TEA (2.0 mL, 14.27 mmol) in dry DCM (50 mL) was added MsCI (0.8 mL, 10.46 mmol) at 0°C under N2 atmosphere. The mixture was stirred at 0°C for 15 min and at RT for 30 min. A solution of methyl thioglycolate (0.93 mL, 10.46 mmol) and sodium methoxide (38 mL of 0.5N sol in MeOH) was added, and stirring was continued for 1h at RT. More sodium methoxide (38 mL of 0.5N sol in MeOH) was added, and the reaction mixture was stirred at RT for 2 h and then evaporated. The residue was partitioned between water and DCM, and extracted twice with DCM. The organic layer was washed with brine, dried, filtered and evaporated to afford the desired product (1.1 g, 40%). It was used in the next step without further purification. HPLC-MS (Method 4): tR= 4.24 min, [M+H]+ m/z 299.2. 1H NMR (300 MHz, CDCI3) δ 4.52 (m, 2H), 4.31 (dt, J = 11.1 , 3.0 Hz, 2H), 3.75 (s, 3H), 1.42 (s, 9H) ppm.
Intermediate 9
3-Amino-4,6-dihydro-thieno[2,3-c]pyrrole-2,5-dicarboxylic acid 5-ethyl ester 2-methyl ester
A solution of 3-amino-4,6-dihydro-thieno[2,3-c]pyrrole-2,5-dicarboxylic acid 5-tert- butyl ester 2-methyl ester (1.1 g, 3.68 mmol) in dry DCM (10 mL) and TFA (1.7 mL, 22.20 mmol) was stirred at RT for 3h. The mixture was concentrated, and the residue was taken up in sat aq NaHC03 and extracted with DCM and with CHCl3/i-PrOH 1 :1. The combined organic layers were dried, filtered and evaporated. The residue (0.44 g) was dissolved in dry DCM (15 mL) and treated with TEA (1.54 mL, 11.06 mmol) and ethyl chloroformate (0.43 mL, 4.44 mmol). The reaction mixture was stirred at RT overnight. Sat aq NaHC03 was added, and the product was extracted with DCM, washed with 1M aq HCI and brine/NaHC03. The organics were dried, filtered and evaporated. The residue was purified by column chromatography (EtOAc/cyclohexane 30:70 to 50:50) to give the desired product (0.270 g, 27%). 1H NMR (300 MHz, DMSO) δ 6.67 (s, 2H), 4.54 (m, 2H), 4.33 (m, 2H), 4.09 (m, 2H), 3.70 (s, 3H), 1.21 (m, 3H) ppm.
Intermediate 10
5-(3-Methoxy-phenyl)-7-oxo-1,3,6,7-tetrahydro-8-thia-2,4,6-tria2a- cyclopenta[a]indene-2-carboxylic acid ethyl ester
3-Amino-4,6-dihydro-thieno[2,3-c]pyrrole-2,5-dicarboxylic acid 5-ethyl ester 2- methyl ester (0.080 g, 0.29 mmol) and 3-methoxybenzonitrile (0.06 mL, 0.44 mmol) were placed in a pressure tube, and 4M HCI/dioxane (2 mL) was added dropwise. The tube was closed, sonicated for 10 min at RT and then stirred at 103°C overnight. More 3-methoxybenzonitrile (0.04 mL) was added, and stirring was continued at 110°C for 6h. The reaction mixture was concentrated, sat aq NaHC03 was added and the precipitate was filtered off and washed with water and EtOAc to give the desired product (0.052 g, 47%). HPLC-MS (Method 4): t*= 3.90 min, [M+H]+ m/z 372.1.
Intermediate 11
5-(3-Methoxy-phenyl)-7-morpholin-4-yl-1,3-dihydro-8-thia-2,4,6-triaza- cyclopenta[a]indene-2-carboxylic acid ethyl ester
TsCI (0.050 g, 0.24 mmol) and DMAP (cat.) were added to a previously sonicated suspension of 5-(3-methoxy-phenyl)-7-oxo-1 ,3,6,7-tetrahydro-8-thia-2,4,6-triaza- cyclopenta[a]indene-2-carboxylic acid ethyl ester (0.030 g, 0.08 mmol) and ΤΕΞΑ (0.06 mL, 0.24 mmol) in dry acetonitrile (1 mL). The mixture was stirred at RT for 4h. Morpholine (0.03 mL, 0.32 mmol) was added, and the reaction mixture was stirred at 45°C overnight. On cooling, the precipitate was filtered off and rinsed with CH3CN to give the desired product (0.021 g, 60%). HPLC-MS (Method 4): tR= 4.71 min, [M+H]+ m/z 441.2. 1H NMR (300 MHz, DMSO) δ 7.96 (m, 1H), 7.71 (m, 1 H), 7.41 (m, 1H), 7.08 (m, 1 H), 4.72 (m, 4H), 4.13 (m, 2H), 3.98 (m, 4H), 3.83 (m, 4H), 1.25 (m, 3H) ppm.
Example 12
5-(3-Hydroxy-phenyl)-7-morpholin-4-yl-1,3-dihydro-8-thia-2,4,6-triaza- cyclopenta[a]indene-2-carboxylic acid ethyl ester
To a suspension of 5-(3-methoxy-phenyl)-7-morpholin-4-yl-1 ,3-dihydro-8-thia- 2,4,6-triaza-cyclopenta[a]indene-2-carboxylic acid ethyl ester (0.020 g, 0.045 mmol) in dry DCM (1 mL) was added 1 M BBrj/DCM (0.7 mL) at -15°C (ice in MeOH). The reaction mixture was kept in the freezer at -20°C for 18h and was then poured into ice. The solid that had formed was filtered off and rinsed with water. When the organic solvent was removed from the filtrate, a new crop precipitated that was filtered off and rinsed with water. The solids were combined and purified by prep-HPLC to give the desired product (0.004 g, 21%). HPLC-MS (Method 1): = 5.23 min, [M+H]+ m/z 427.2. 1H NMR (300 MHz, DMSO) δ 9.50 (s, 1 H), 7.85 (m, 2H), 7.27 (m, 1H), 6.87 (m, 1H), 4.76 (m, 2H), 4.68 (m, 2H), 4.13 (m, 2H), 3.97 (m, 4H), 3.80 (m, 4H), 1.25 (m, 3H) ppm.
Intermediate 12
3-Ureido-4,6-dihydro-thieno[2,3-c]pyrrole-2,5-dicarboxylic acid 5-ethyl ester 2-methyl ester
To a solution of 3-amino-4,6-dihydro-thieno[2,3-c]pyrrole-2,5-dicarboxylic acid 5- ethyl ester 2-methyl ester (0.785 g, 2.90 mmol) in dry DCM (15 mL) was added chlorosulfonyl isocyanate (0.4 mL, 4.65 mmol) at -78°C under Ar. The reaction mixture was allowed to warm to RT and stirred for 1 h. The solvent was evaporated, and the residue was taken up in 6N aq HCI and stirred at 100°C for 40 min, then neutralized with sat aq NaHC03 at RT. The suspension was filtered, and the solid was rinsed with water and dried to give the desired product (0.833 g, 92%). HPLC-MS (Method 4): t«= 3.79 min, [M+H]+ m/z 314.1. 1H NMR (300 MHz, DMSO) δ 9.16 (s, 1 H), 6.70 (s, 2H), 4.58 (m, 4H), 4.08 (q, J = 6.5 Hz, 2H), 3.80 (s, 3H), 1.21 (t, J = 6.6 Hz, 3H) ppm. Intermediate 13
5,7-Dioxo-1,3,4,5,6,7-hexahydro-8-thia-2,4,6-triaza-cyclopenta[a]indene-2- carboxylic acid ethyl ester
To a suspension of 3-ureido-4,6-dihydro-thieno[2,3-c]pyrrole-2,5-dicarboxylic acid 5-ethyl ester 2-methyl ester (0.833 g, 2.66 mmol) in metanol (10 mL) was added aq 1.5M KOH (4 mL), and the reaction mixture was stirred at RT for 2h. Water was added followed by 6M aq HCI (up to pH~4-5). The suspension was filtered, and the solid was rinsed with water and dried to give the desired product (0.609 g, 81 %). HPLC-MS (Method 4): t^ 2.81 min, [M+H]+ m/z 282.0. 1H NMR (300 MHz, DMSO) δ 11.61 (s, 1 H), 11.24 (s, 1H), 4.64 (s, 2H), 4.43 (d, J = 10.7 Hz, 2H), 4.11 (q, J = 6.3 Hz, 2H), 1.22 (t, J = 6.1 Hz, 3H) ppm. Intermediate 14
7-Morpholin-4-yl-5-(toluene-4-sulfonyloxy)-1,3-dihydro-8-thia-2,4,6-triaza- cyclopenta[a]indene-2-carboxylic acid ethyl ester
To a suspension of 5,7-dioxo-1 , 3,4,5, 6,7-hexahydro-8-thia-2,4,6-triaza- cyclopenta[a]indene-2-carboxylic acid ethyl ester (0.609 g, 2.16 mmol), TEA (0.75 mL, 5.41 mmol) and DMAP (0.030 g, 0.22 mmol) in DCM (30 mL) was added p- toluenesulfonyl chloride (0.908 g, 4.76 mmol). The reaction mixture was stirred at RT for 20 h. More TEA (0.5 mL), DMAP (0.030 g) and TsCI (0.460 g) were added, and stirring was continued for 4 h. Morpholine (0.3 mL, 3.25 mmol) was added, and the reaction mixture was stirred for 2 h. The solvent was removed under reduced pressure, and the residue was taken up in DCM and washed with sat aq NaHC03. The organic layer was dried over Na2S04, filtered and evaporated. The residue was purified by column chromatography (MeOH:DCM, 1 :99 to 10:90) to afford the desired product (0.100 g, 9%). 1H NMR (300 MHz, DMSO) δ 7.92 (d, J = 8.0 Hz, 2H), 7.50 (d, J = 8.1 Hz, 2H), 4.76 (m, 2H), 4.53 (m, 2H), 4.13 (q, J = 7.0 Hz, 2H), 3.62 (m, 8H), 2.41 (s, 3H), 1.24 (m, 3H) ppm.
Example 13
5-(2-Amino-pyrimidin-5-yl)-7-morpholin-4-yl-1,3-dihydro-8-thia-2,4,6-triaza- cyclopenta[a]indene-2-carboxylic acid ethyl ester
To a suspension of 7-morpholin-4-yl-5-(toluene-4-sulfonyloxy)-1 ,3-dihydro-8-thia- 2,4,6-triaza-cyclopenta[a]indene-2-carboxylic acid ethyl ester (0.090 g, 0.178 mmol), 2-aminopyrimidine-5-boronic acid pinacol ester (0.063 g, 0.285 mmol) and Pd(dppf)CI2.DCM (0.015 g, 0.018 mmol) in 1 ,2-DME (2.5 mL), an aq solution of Na2C03 (2M, 0.6 mL) was added. The reaction mixture was heated under microwave irradiation at 130°C for 1h. On cooling, the mixture was concentrated and the residue was purified by column chromatography (MeOH:DCM 2:98 to 7:93). The product obtained was triturated with acetonitrile and Et20 and removed by filtration to give the desired product (0.029 g, 38%). HPLC-MS (Method 1): = 4.98 min, [M+H]+ m/z 428.2. 1H NMR (300 MHz, DMSO) δ 9.12 (s, 2H), 7.12 (s, 2H), 4.78 (m, 2H), 4.68 (m, 2H), 4.14 (q, J = 7.1 Hz, 2H), 3.95 (m, 4H), 3.79 (m, 4H), 1.26 (t, J = 7.1 Hz, 3H) ppm. Preparation of Example 14
Example 14 and its corresponding intermediates have been prepared following the synthetic route as described for 5-(2-Amino-pyrimidin-5-yl)-7-morpholin-4-yl- 1,3-dihydro-8-thia-2,4,6-triaza-cyclopenta[a]indene-2-carboxylic acid ethyl ester (Example 13).
Intermediate 15
3-Amino-4-methyl-4,6-dihydro-thieno[2,3-c]pyrrole-2,5-dicarboxylic acid 5- tert-butyl ester 2-methyl ester
Synthesis as described for Intermediate 8 starting from 10 g of tert-butyl-3-cyano-
2- methyl~4-oxopyrrolidine-1-carboxylate.
Yield: 4.72 g of crude product.
HPLC-MS (Method 4): tR= 4.61 min.
Intermediate 16
3- Amino-4-methyl-4,6-dihydro-thieno[2,3-c]pyrrole-2,5-dicarboxylic acid 5- ethyl ester 2-methyl ester
Synthesis as described for Intermediate 9.
Yield: 2.53 g
HPLC-MS (Method 4): t*= 4.22 min, [M+H]+ m/z 285.0.
Intermediate 17
4- Methyl-3-ureido-4,6-dihydro-thieno[2,3-c]pyrrole-2,5-dicarboxylic acid 5- ethyl ester 2-methyl ester
Synthesis as described for Intermediate 12.
Yield: 2.08 g (69 %)
HPLC-MS (Method 4): t«= 3.87 min, [M+HJ+ m/z 228.0. Intermediate 18
3-Methyl-5,7-dioxo-1,3,4,5,6,7-hexahydro-8-thia-2,4,6-triaza- cyclopenta[a]indene-2-carboxylic acid ethyl ester
Synthesis as described for Intermediate 13.
Yield: 1.61 g (86 %) HPLC-MS (Method 4): t#r= 3.24 min. 1H NMR (300 MHz, DMSO) δ 11.61 (bs, 1 H), 11.26 (bs, 1H), 4.94 (s, 1H), 4.65 (m, 2H), 4.12 (m, 2H), 1.42/1.40 (2s, 3H), 1.22 (2t, 3H) ppm. Intermediate 19
3-Methyl-7-morpholin-4-yl-5-(toluene-4-sulfonyloxy)-1,3-dihydro-8-thia-2,4,6- triaza-cyclopenta[a]indene-2-carboxylic acid ethyl ester
Synthesis as described for Intermediate 14.
Yield: 0.755 g (27 %)
HPLC-MS (Method 4): t*= 4.81 min, [M+H]+ m/z 519.2. 1H NMR (300 MHz, DMSO) δ 7.90 (d, 2H), 7.50 (d, 2H), 4.79 (m, 1 H), 4.75 (m, 2H), 4.12 (m, 2H), 3.68 (m, 8H), 2.43 (s, 3H), 1.42/1.40 (2s, 3H), 1.25 (m, 3H) ppm.
Example 14
5-(2-Amino-pyrimidin-5-yl)-3-methyl-7-morpholin-4-yl-1 ,3-dihydro-8-thia- 2,4,6-triaza-cyclopenta[a]indene-2-carboxylic acid ethyl ester
Synthesis as described for Example 13.
Yield: 0.060 g (9 %)
HPLC-MS (Method 1): t/?= 4.70 min, [M+H]+ m/z 442.2. 1H NMR (300 MHz, DMSO) δ 9.12 (s, 2H), 7.12 (s, 2H), 5.13 (s, 1 H), 4.77 (m, 2H), 4.14 (m, 2H), 3.94 (m, 4H), 3.79 (m, 4H), 1.62/1.60 (2s, 3H), 1.26 (m, 3H) ppm.
Example 15
5-(7-Morpholin-4-yl-2,3-dihydro-1H-8-thia-2,4,6-triaza-cyclopenta[a]inden-5- yl)-pyrimidin-2-ylamine (HCI salt)
To a suspension of 5-(2-amino-pyrimidin-5-yl)-7-morpholin-4-yl-1 ,3-dihydro-8- thia-2,4,6-triazacyclopenta[a]indene-2-carboxylic acid ethyl ester (0.20 g, 0.468 mmol, 1 eq) in MeOH/2-propanol (2 mL each), lithium hydroxide monohydrate (0.216 g, 5.146 mmol, 11 eq) was added, and the reaction mixture was heated in the microwave oven at 160°C for 1 h. The solvents were removed, and the residue was redissolved in water, neutralized with 1N HCI to pH~7 and extracted with CHCIs/i-PrOH 1 :1. The organic phase was separated (slow!) and concentrated, and the crude was redissolved in DCM and little MeOH. Drops of ~4M HCI in 1 ,4- dioxane were slowly added until the color turned from brown to grey and a precipitate appeared. The solid was filtered off and dried affording the title product (dark grey solid, 0.037 g, 20%).
HPLC-MS (Method 6): t«= 3.57 min, [M+H]+ m/z 356.2. 1H NMR (300 MHz, DMSO) δ 10.34 (s, 2H), 9.13 (s, 2H), 7.20 (s, 2H), 4.69 (s, 2H), 4.58 (s, 2H), 3.95 (d, J = 4.7 Hz, 4H), 3.80 (d, J = 4.7 Hz, 4H) ppm.
Example 16
[5-(2-Amino-pyrimidin-5-yl)-7-morpholin^-yl-1,3-dihydro-8-thia-2,4,6-triaza- cyclopenta[a]inden-2-yl]-(4-fluoro-phenyl)-methanone
4-Fluorobenzoyl chloride (0.03 mL, 0.17 mmol. 1.2 eq) was added at RT to a mixture of crude 5-(7-morpholin-4-yl-2,3-dihydro-1 H-8-thia-2,4,6-triaza- cyclopenta[a]inden-5-yl)-pyrimidin-2-ylamine (0.050 g, 0.14 mmol), TEA (0.06 mL, 0.42 mmol, 3.0 eq) and DMAP (catalytic) in dry acetonitrile (1 mL). The reaction mixture was stirred at RT for 6h, then quenched with sat aq NaHC03 and extracted with CHCl3/i-PrOH 1 :1. The organic phase was dried and concentrated, and the residue was purified by silica gel chromatography (DCM/MeOH 95:5 to 9:1) affording the title compound (0.020 g, 30%).
HPLC-MS (method 1): Rt= 4.98 min, [M+1]+ m/z 478.1. 1H NMR (300 MHz, DMSO) δ 9.14 (s, 0.8H, conformer A), 9.06 (s, 1.2H, conformer B), 7.78-7.73 (m, 2H), 7.36-7.31 (m, 2H), 7.11 (bd, 2H), 5.01-4.82 (m, 4H), 3.94 (bs, 4H), 3.79 (bs, 4H) ppm.
Example 17
5-[2-(4-Fluoro-benzenesulfonyl)-7-morpholin-4-yl-2,3-dihydro-1H-8-thia- 2,4,6-triaza-cyclopenta[a]inden-5-yl]-pyrimidin-2-ylamine
4-Fluorobenzenesulfonyl chloride (33 mg, 0.17 mmol) was added at RT to a suspension of 5-(7-morpholin-4-yl-2,3-dihydro-1 H-8-thia-2,4,6-triaza- cyclopenta[a]inden-5-yl)-pyrimidin-2-ylamine (50 mg, 0.14 mmol) and TEA (0.06 mL, 0.42 mmol) in dry acetonitrile (1 mL). The reaction mixture was stirred at RT overnight. Acetonitrile and Et20 were added and the suspension was filtered and rinsed with DCM/MeOH and CHCI PrOH. The solid was purified by column chromatography (DCM/MeOH 100:0 to 9:1) to afford the title compound (25 mg, 35%).
HPLC-MS (Method 1): R, = 5.38 min, [M+H]+ m/z 514.2. H NMR (300 MHz, DMSO) δ 9.09 (s, 2H), 8.03 (dd, J = 8.7, 5.1 Hz, 2H), 7.46 (t, J = 8.7 Hz, 2H), 7.14 (s, 2H), 4.78 (s, 2H), 4.67 (s, 2H), 3.90 (m, 4H), 3.76 (m, 4H) ppm. Example 18
5-(2-Amino-pyrimidin-5-yl)-7-morpholin^-yl-1,3-d^
cyclopenta[a]indene-2-carboxylic acid methyl ester
To a suspension of 5-(2-amino-pyrimidin-5-yl)-7-morpholin-4-yl-1 ,3-dihydro-8- thia-2,4,6-triaza-cyclopenta[a]indene-2-carboxylic acid ethyl ester (0.2 g, 0.468 mmol) in MeOH/'PrOH (4:4 mL) was added lithium hydroxide monohydrate (0.216 g, 5.146 mmol). The reaction mixture was heated under microwave irradiation at 160°C for 1h. Solvents were removed and the residue was redissolved in water, neutralised with 1N HCI to pH~7 and extracted with MeOH/'PrOH 1:1. The organic layer was dried, filtered and evaporated. The residue was purified by column chromatography (DCM/MeOH 100:0 to 95:5) and by prep-HPLC to give the deprotected product (37 mg, 20%), the i-propyl carbamate derivative (3 mg, 1 %) and the title compound (6 mg, 3%).
HPLC-MS (Method 1): R, = 3.85 min, [M+H]+ m/z 414.1.
1H NMR (300 MHz, DMSO) δ 9.11 (d, J = 1.9 Hz, 2H), 7.13 (s, 2H), 4.76 (m, 2H), 4.65 (m, 2H), 3.95 (m, 4H), 3.78 (m, 5H), 3.69 (s, 3H) ppm.
Example 19
5-[2^4-Fluoro-benzyl)-7-morpholin-4-yl-2,3-dihydro-1 H-8-thia-2,4,6-triaza- cyclopenta[a]inden-5-yl]-pyrimidin-2-ylamine
To a suspension of 5-(7-morpholin-4-yl-2,3-dihydro-1 H-8-thia-2,4,6-triaza- cyclopenta[a]inden-5-yl)-pyrimidin-2-ylamine (50 mg, 0.14 mmol) and 4- fluorobenzaldehyde (0.04 mg, 0.28 mmol) in MeOH (1.5 mL) was added NaCNBH3 (few crystalls) and AcOH (0.03 mL, 0.42 mmol). The reaction mixture was stirred at RT overnight. Sat aq NaHC03 was added and the mixture was extracted with DCM. The organic layer was dried, filtered and evaporated. The residue was purified by column chromatography (DCM/MeOH 10:0 to 9:1) and by prep-HPLC to afford the title compound (10 mg, 14%).
HPLC-MS (Method 1): R, = 2.83 min, [M+H]+ m/z 464.2. H N R (300 MHz, DMSO) δ 9.08 (s, 2H), 8.18 (s, 1 H), 7.46 (dd, J = 8.2, 5.6 Hz, 2H), 7.19 (t, J = 8.3 Hz, 2H), 7.10 (s, 2H), 4.09 (m, 2H), 3.98 (m, 2H), 3.94 (m, 4H), 3.78 (m, 4H) ppm. Example 20
5-(2-Amino-4 rifluoromethyl-pyrimidin-5-yl)-7-morpholin-4-yl-1 ,3-dihydro-8- thia-2,4,6-triaza-cyclopenta[a]indene-2-carboxylic acid ethyl ester
To a suspension of 7-morpholin-4-yl-5-(toluene-4-sulfonyloxy)-1 ,3-dihydro-8-thia- 2,4,6-triaza-cyclopenta[a]indene-2-carboxylic acid ethyl ester (80 mg, 0.16 mmol) and 5-(4,4,5,5-tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-4-trifluoromethyl-pyrimidin-2- ylamine (60% pure, 140 mg, 0.24 mmol) in 1 ,2-DME (2 mL) was added aq 2M Na2C03 (0.4 mL) and PdCI2(dppf) DCM (13 mg, 0.016 mmol). The reaction mixture was heated under microwave irradiation at 130°C for 30 min. On cooling, the mixture was purified by column chromatography (DCM/MeOH 98:2 to 9:1) and by prep-HPLC to afford the title compound ( 2 mg, 15%).
HPLC-MS (Method 1): Rf = 5.07 min, [M+H]+ m/z 496.3.
1H NMR (300 MHz, DMSO) δ 8.87 (s, 1 H), 7.59 (s, 2H), 4.80 (m, 2H), 4.64 (m, 2H), 4.14 (q, J = 7.1 Hz, 2H), 3.93 (m, 4H), 3.76 (m, 4H), 1.25 (t, J = 7.1 Hz, 3H) ppm.
Example 21
5-(2-Amino^yrimidin-5-yl)-7-morpholin^-yl-1,3-dihydro-8-thia-2,4,6-triaza- cyclopenta[a]indene-2-carboxylic acid isopropyl ester
To a suspension of 5-(2-amino-pyrimidin-5-yl)-7-morpholin-4-yl-1 ,3-dihydro-8- thia-2,4,6-triaza-cyclopenta[a]indene-2-carboxylic acid ethyl ester (0.2 g, 0.468 mmol) in MeOH/'PrOH (4:4 mL) was added lithium hydroxide monohydrate (0.216 g, 5.146 mmol). The reaction mixture was heated under microwave irradiation at 160°C for 1 h. Solvents were removed and the residue was redissolved in water, neutralised with 1 N HCI to pH~7 and extracted with MeOH/'PrOH 1 :1. The organic layer was dried, filtered and evaporated. The residue was purified by column chromatography (DCM/MeOH 100:0 to 95:5) and by prep-HPLC to give the deprotected product (37 mg, 20%), the methyl carbamate derivative (6 mg, 3%) and the title compound (3 mg, 1%).
HPLC-MS (Method 1 ): R, = 4.61 min, [M+H]+ m/z 442.2. H NMR (300 MHz, DMSO) δ 9.11 (s, 2H), 7.13 (s, 2H), 4.86 (m, 1H), 4.75 (s, 2H), 4.63 (s, 2H), 3.93 (m, 4H), 3.79 (m, 4H), 1.26 (m, 6H) ppm.
Example 22
1-[5^2-Amino-pyrimidin-5-yl)-7-morpholin^-yl-1,3-dihydro-8-thia-2,4,6- triaza-cyclopenta[a]inden-2-yl]-ethanone
To a suspension of 5-(7-morpholin-4-yl-2,3-dihydro-1 H-8-thia-2,4,6-triaza- cyclopenta[a]inden-5-yl)-pyrimidin-2-ylamine (29 mg, 0.074 mmoi) in acetonitrile (1.5 mL) was added acetyl chloride (0.006 mL, 0.089 mmoi) and DIPEA (0.051 mL, 0.296 mmoi). The reaction mixture was stirred at RT overnight. Solvent was removed, DCM was added and the mixture was washed with water and sat. sol. NaHC03. Aq. phases were extracted twice with CHCI PrOH 1:1. The organic layers were dried, filtered and evaporated. The residue was purified by prep- HPLC to give the title compound (8 mg, 27%).
HPLC-MS (Method 1 ): R, = 3.24 min, [M+H]+ m/z 398.2.
1H NMR (300 MHz, DMSO) δ 9.12 (s, 2H), 7.14 (s, 2H), 4.99 (s, 0.8H), 4.89 (s, 1.2H), 4.75 (s, 1.2H), 4.62 (s, 0.8H), 3.96 (m, 4H), 3.79 (m, 4H), 2.11 (s, 1.8H), 2.08 (s, 1.2H) ppm. Example 23
5-(2-Amino-pyrimidin-5-yl)-7-morpholin-4-yl-1,3-dihydro-8-thia-2,4,6-triaza- cyclopenta[a]indene-2-carboxylic acid ethylamide
To a suspension of 5-(7-morpholin-4-yl-2,3-dihydro- H-8-thia-2,4,6-triaza- cyclopenta[a]inden-5-yl)-pyrimidin-2-ylamine (66 mg, 0.186 mmoi) in acetonitrile (4 mL) was added ethyl isocyanate (0.018 mL, 0.223 mmoi) and DIPEA (0.081 mL, 0.465 mmoi). The reaction mixture was stirred at RT overnight. Solvent was removed and the residue was extracted twice with CHCI PrOH 1:1. The organic layers were dried, filtered and evaporated. The residue was purified by prep- HPLC to give the title compound (6 mg, 8%).
HPLC-MS (Method 1 ): R, = 3.46 min, [M+H]+ m/z 427.2.
1H NMR (300 MHz, DMSO) δ 9.10 (s, 2H), 7.12 (s, 2H), 6.47 (t, J = 5.2 Hz, 1 H), 4.71 (m, 2H), 4.61 (m, 2H), 3.95 (m, 4H), 3.78 (m, 4H), 3.12 (m, 2H), 1.07 (t, J = 7.1 Hz, 3H) ppm. Example 24
5-(2-Ethyl-7-morpholin-4-yl-2,3-dihydro-1 H-8-thia-2,4,6-triaza- cyclopenta[a]inden-5-yl)-pyrimidin-2-ylamine
To a suspension of 5-(7-morpholin-4-yl-2,3-dihydro-1H-8-thia-2,4,6-triaza- cyclopenta[a]inden-5-yl)-pyrimidin-2-ylamine (60 mg, 0.17 mmol) and acetaldehyde (few drops) in 1 ,2-DCE (2 mL) was added Na(OAc)3BH (54 mg, 0.25 mmol) and AcOH (0.01 mL). The reaction mixture was stirred at RT overnight. Sat aq NaHC03 was added and the mixture was extracted with CHCI PrOH 1 :1. The organic layers were dried, filtered and evaporated. The residue was purified by column chromatography (DCM/MeOH 95:5, 9:1, then 9:1 with 1 %TEA) and by prep-HPLC to give the title compound (5 mg, 8%).
HPLC-MS (Method 2): R, = 2.24 min, [M+H]+ m/z 384.2.
1H NMR (300 MHz, DMSO) 6 9.10 (s, 2H), 8.37 (s, 1H), 7.11 (s, 2H), 4.07 (m, 2H), 3.96 (m, 6H), 3.78 (m, 4H), 2.81 (q, J = 7.1 Hz, 2H), 1.13 (t, J = 7.1 Hz, 3H) ppm.
Example 25
5-(2-Ethanesulfonyl-7-morpholin-4-yl-2,3-dihydro-1 H-8-thia-2,4,6-triaza- cyclopenta[a]inden-5-yl)-pyrimidin-2-ylamine
A mixture of 5-(7-morpholin-4-yl-2,3-dihydro-1H-8-thia-2,4,6-triaza- cyclopenta[a]inden-5-yl)-pyrimidin-2-ylamine (60 mg, 0.169 mmol) and ethanesulfonyl chloride (0.019 mL, 0.202 mmol) in dry Py (2 mL) was stirred at RT for overnight. More ethanesulfonyl chloride (0.016 mL, 0.169 mmol) and 4- DMAP (5 mg, 0.034 mmol) were added and reaction mixture was stirred at RT overnight and heated at 45°C for 1h. More ethanesulfonyl chloride (0.032 mL, 0.338 mmol) was added and the mixture was heated at 60°C overnight (still SM observed). H20 was added and the mixture was extracted with CHCI PrOH 1 :1. The organic layers were washed with 1.2M HCI (x2), H20 (x2) and brine (x2), dried, filtered and evaporated. The residue was purified by column chromatography (DCM/MeOH 100:0 to 97:3) to give the title compound (8 mg, 1 %).
HPLC-MS (Method 1): R, = 3.78 min, [M+H]+ m/z 448.2.
1H NMR 1H NMR (300 MHz, DMSO) δ 9.11 (s, 2H), 7.14 (s, 2H), 4.84 (s, 2H), 4.73 (s, 2H), 3.95 (m, 4H), 3.78 (m, 4H), 3.29 (q, J = 7.5 Hz, 2H), 1.25 (t, J = 7.3 Hz, 3H) ppm. Example 26
Biological activity in PI3-K and/or mTOR for certain examples is represented in Table 1 by semi-quantative results: between 100 nM and 10 μΜ (0.1-10μΜ) (*) and between 1 and 100 nM (0.001-0.1 μΜ) (**). For instance, selected exemplary compounds displayed the following specific IC50 values (in μΜ): Example 1 (PI3Kcc 0.530; mTOR 0.112), Example 5 (PI3Ka 0.061 ; mTOR 0.200), Example 12 (PI3Ka 0.132; mTOR 0.081). Biological activity for certain examples is also represented by quantitative results in the table below. Cellular activity is also represented in the table below, which measures the cellular activity of certain compounds of the invention/examples by the inhibition of phosphorylation of AKT using ELISA assay (as described hereinbefore).
Figure imgf000087_0001

Claims

Claims
1. A compound of formula I,
Figure imgf000088_0001
wherein: n represents 0, 1 or 2; A,, A2, A3 and each A4 (if present) independently represents -C(R )R5-, -N(R6)-, -C(O)-, -0-, -S-, -S(O)- or -S(0)2-; the dotted lines represent the presence of an optional double bond, which may be present between A, and A2, A2 and A3, A3 and A4 (if the latter is present, i.e. when n does not represent 0) and/or between two A groups (if present, i.e. when n represents 2), provided that the ΑΊ to A-i-containing ring is not aromatic; each B1, B1a, B2, B2a, B3, B3a, B4 and B4a independently represent hydrogen or a substituent selected from halo, -C(=Y)-R10a, -C(=Y)-OR10a, -C(=Y)N(R10a)R1 a, -S(O)2N(R10a)R11a, d.12 alkyl, heterocycloalkyl (which latter two groups are optionally substituted by one or more substituents selected from =0 and E1), aryl and/or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from E2); or any two B1, B1a, B2, B2a, B3, B3a, B4 and B4a substituents that are attached to the same carbon atom (i.e. B1 and B a; B2 and B2a; B3 and B3a; and/or B4 and B4a) may together form a =0 group; or, any two B\ B a, B2, B2a, B3, B3a, B4 and B a substituents may be linked together to form a further 3- to 12- membered ring, optionally containing (in addition to the atom(s) of the morpholine ring) one or more heteroatom(s), which ring optionally contains one or more double bonds, and which ring is itself optionally substituted by one or more substituents selected from halo, =0 and C1-3 alkyl optionally substituted by one or more fluoro atoms;
R3 represents aryl or heteroaryl (both of which are optionally substituted by one or more substituents selected from E4);
R4 and R5 independently represent hydrogen, halo, -OR10C, -N(R10d)R 1d, -N(R10e)-C(O)-R10f, -C(O)R10g, -C(O)OR10h, -C(O)N(R10i)R1 i, -N(R10')-C(O)OR10k, -N(R10m)-C(O)-N(R10n)R11n, -N[-C(O)-T -R10p]-C(O)-T2-R10q, C1-12 alkyl, heterocycloalkyi (which latter two groups are optionally substituted by one or more substituents selected from E5 and =0), aryl or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from E6 and =0); or
R4 and R5 may be linked together to form a 3- to 6-membered ring, optionally containing one or more heteroatom(s), which ring optionally contains one or more double bonds, and which ring is itself optionally substituted by one or more substituents selected from halo, =0 and C1-3 alkyl optionally substituted by one or more fluoro atoms; T1 and T2 independently represent a single bond, -N(R10*)- or -0-;
R6 represents hydrogen, -C(O)-R 0r, -C(O)-OR10s, -C(O)-N(R 0l)R11, l -S(O)2R 0u, Cv alkyl, heterocycloalkyi (which latter two groups are optionally substituted by one or more substituents selected from E7 and =0), aryl or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from E8 and =0); each R10a, R1 a, R10c, R10d, R1 d, R10e, R10', R1°9, R10h, R10i, R11i, R10j, R10k, R10m, R10n, R11n, R10p, R10q, R10r, R10s, R10t, R11t, R10u and R10x independently represent, on each occasion when used herein, hydrogen, d.12 alkyl, heterocycloalkyi (which latter two groups are optionally substituted by one or more substituents selected from =0, =S, =N(R20) and E10), aryl or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from E11); or any relevant pair of R 0a and R11a and/or any pair of R10d and R11d, R10i and Rl1i, R10n and R11n and R10t and R11t may be linked together to form a 4- to 20- membered ring, optionally containing one or more heteroatoms, optionally containing one or more unsaturations, and which ring is optionally substituted by one or more substituents selected from =0, =S, =N(R20) and E12; each E\ E2, E4, E5, E6, E7, E8, E10, E11 and E12 independently represents, on each occasion when used herein:
(i) Q";
(ii) alkyl optionally substituted by one or more substituents selected from =0 and Q5; or any two E1, E2, E4, E5, E6, E7, E8, E10, E11 or E12 groups may be linked together to form a 3- to 12-membered ring, optionally containing one or more unsaturations, and which ring is optionally substituted by one or more substituents selected from =0 and J1; each Q4 and Q5 independently represent, on each occasion when used herein: halo, -CN, -N02, -N(R20)R21, -OR20, -C(=Y)-R20, -C(=Y)-OR20, -C(=Y)N(R20)R21, -OC(=Y)-R20, -OC(=Y)-OR20, -OC(=Y)N(R20)R21, -OS(0)2OR2°, -OP(=Y)(OR20)(OR21), -OP(OR20)(OR21), -N(R22)C(=Y)R21, -N(R2 )C(=Y)OR21, -N(R22)C(=Y)N(R20)R21, -NR22S(0)2R20, -NR 2S(O)2N(R20)R21, -S(O)2N(R20)R21, -SC(=Y)R20, -S(0)2R20, -SR20, -S(0)R20, C1-6 alkyl, heterocycloalkyl (which latter two groups are optionally substituted by one or more substituents selected from =0 and J2), aryl or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from J3); each Y independently represents, on each occasion when used herein, =0, =S, =NR23 or =N-CN; each R20, R21, R22 and R23 independently represent, on each occasion when used herein, hydrogen, d-e alkyl, heterocycloalkyl (which latter two groups are optionally substituted by one or more substituents selected from J4 and =0), aryl or heteroaryl (which latter two groups are optionally substituted by one or more substituents selected from J5); or any relevant pair of R20, R21 and R22, may be linked together to form a 4- to 20- membered ring, optionally containing one or more heteroatoms, optionally containing one or more unsaturations, and which ring is optionally substituted by one or more substituents selected from J6 and =0; each J1, J2, J3, J4, J5 and J6 independently represents, on each occasion when used herein:
(i) Q7;
(ii) Ci_6 alkyl or heterocycloalkyl, both of which are optionally substituted by one or more substituents selected from =0 and Q8; each Q7 and Q8 independently represents, on each occasion when used herein: halo, -N(R50)R51, -OR50, -C(=Ya)-R50, -C(=Ya)-OR50, -C(=Ya)N(R50)R51, -N(R52)C(=Ya)R51, -NR52S(0)2R50, -S(O)2N(R50)R51, -N(R52)-C(=Ya)-N(R50)R51, -S(0)2R5°, -SR50, -S(0)R50 or C1-6 alkyl optionally substituted by one or more fluoro atoms; each Ya independently represents, on each occasion when used herein, =0, =S, =NR53 or =N-CN; each R50, R51, R52 and R53 independently represents, on each occasion when used herein, hydrogen or C,^ alkyl optionally substituted by one or more substituents selected from fluoro, -OR60 and -N(R61)R62; or
any relevant pair of R50, R51 and R52 may be linked together to form, a 3- to 8- membered ring, optionally containing one or more heteroatoms, optionally containing one or more unsaturations, and which ring is optionally substituted by one or more substituents selected from =0 and alkyl; R60, R61 and R62 independently represent hydrogen or d-e alkyl optionally substituted by one or more fluoro atoms; or a pharmaceutically acceptable ester, amide, solvate or salt thereof.
2. A compound as claimed in Claim 1, wherein: the , to A^-containing rings represents one of the following formulae:
Figure imgf000092_0001
; and/or R3 represents optionally substituted phenyl or pyrimidinyl (e.g. 5-pyrimidinyl).
3. A compound as claimed in Claim 1 or Claim 2, wherein: R3 represents aryl (e.g. phenyl) or heteroaryl (e.g. a 5- or 6-membered monocyclic heteroaryl group; which may contain one to four, e.g 3 or, preferably, 1 or 2, heteroatoms preferably selected from nitrogen, oxygen and sulfur) both of which are optionally substituted by one or more (e.g. two, or, preferably, one) substituent(s) selected from E4 (e.g. -OH and/or -N(R20)R21 (e.g. -NH2)); each R4 and R5 independently represent hydrogen or Ci.6 alkyl (optionally substituted as defined herein; but preferably unsubstituted); R4 and R5 may be linked, but are more preferably not linked together; each R6 (when/if present) independently represents hydrogen, -C(O)R10r, -C(O)OR10s, -C(O)N(R10,)R11', -S(O)2R10u or C1-6 (e.g. C1-4) alkyl (e.g. ethyl or methyl) optionally substituted by one or more (e.g. two or, preferably, one) E7 substituents; R10r represents aryl (e.g. phenyl; which aryl group is optionally substituted by one or more E substituent, so forming e.g. a fluorophenyl group) or C1.3 alkyl (e.g. methyl); R10s represents Ci.3 alkyl (e.g. ethyl); R10t represents hydrogen; R11t represents aryl (e.g. phenyl; which aryl group is optionally substituted by one or more E 1 substituent, so forming e.g. a fluorophenyl group) or C1-3 alkyl (e.g. ethyl); R10u represents aryl (e.g. phenyl; which aryl group is optionally substituted by one or more E11 substituent, so forming e.g. a fluorophenyl group) or d.3 (e.g. Ci-2) alkyl (e.g. methyl); E7 represents Q4; when E7 represents Q4, then Q4 preferably represents aryl (e.g. phenyl) optionally substituted by one or more substituents selected from J3 (so forming e.g. a fluorophenyl group); E1 represents Q4; when E1 represents Q4, then Q4 represents halo (e.g. fluoro); J3 represents Q7, in which Q7 preferably represents halo (e.g. fluoro); E4 represents Q4; Q4 represents halo, -OR20, -N(R20)R21 or aryl (optionally substituted by one or more substituents selected from J3); Q5 represents -OR20, -N(R 0)R21 or, preferably, halo (e.g. fluoro); Y represents =0; R20 and R21 independently represent hydrogen or d.3 alkyl (e.g. methyl or ethyl); and/or Q7 represents halo (e.g. fluoro).
4. A compound as claimed in any one of the preceding claims, wherein:
R3 represents hydroxyphenyl (e.g. 3-hydroxyphenyl) or pyrimidinyl (e.g. 5- pyrimidinyl, such as 2-amino-5-pyrimidinyl (i.e. 2-[-N(R20)(R21)]-pyrimidin-5-yl such as 2-NH2-pyrimidin-5-yl)); A represents -C(R4)R5-; one of A2 and A3 (preferably A2) represents -N(R6)- and the other (preferably A3) represents -C(R4)R5-; n represents 0 or 1 ; At represents -C(R4)R5-; only a maximum of two of A^ A2, A3 and, if present, A4, represents -C(O)- (which, if there are two -C(O)- moieties, are preferably not adjacent to one another); and/or the dotted lines do not represent the presence of an optional double bond (i.e. the A to A4-containing ring does not contain a double bond, other than that double bond that is integral to the requisite imidazopyrazine of formula I). 5. A compound as claimed in any one of the preceding claims, wherein:
B1, B a, B2, B2a, B3, B3a, B4 and B4a independently represent hydrogen; each R4 and R5 independently represent hydrogen; and/or each R6 (when/if present) independently represents hydrogen, -C(0)OCH2CH3,
-C(0)N(H)CH2CH3, -S(0)2-[4-fluorophenyl], -C(0)-[4-fluorophenyl], -C(0)CH3, ethyl, -CH2-[4-fluorophenyl], -C(0)-N(H)-[4-fluorophenyl] or -S(0)2CH3.
6. A compound of formula I as defined in any one of Claims 1 to 5, or a pharmaceutically acceptable ester, amide, solvate or salt thereof, for use as a pharmaceutical.
7. A pharmaceutical formulation including a compound of formula I, as defined in any one of Claims 1 to 5, or a pharmaceutically acceptable ester, amide, solvate or salt thereof, in admixture with a pharmaceutically acceptable adjuvant, diluent or carrier.
8. A compound, as defined in any one of Claims 1 to 5, or a pharmaceutically acceptable ester, amide, solvate or salt thereof, for use in the treatment of a disease in which inhibition of a PI3-K and/or mTOR is desired and/or required.
9. Use of a compound of formula I, as defined in any one of Claims 1 to 5, or a pharmaceutically acceptable ester, amide, solvate or salt thereof, for the manufacture of a medicament for the treatment of a disease in which inhibition of a PI3-K and/or mTOR is desired and/or required.
10. A compound as claimed in Claim 8 or a use as claimed in Claim 9, wherein the disease is cancer, an immune disorder, a cardiovascular disease, a viral infection, inflammation, a metabolism/endocrine function disorder, a neurological disorder, an obstructive airways disease, an allergic disease, an inflammatory disease, immunosuppression, a disorder commonly connected with organ transplantation, an AIDS-related disease, benign prostate hyperplasia, familial adenomatosis, polyposis, neuro-fibromatosis, psoriasis, a bone disorder, atherosclerosis, vascular smooth cell proliferation associated with atherosclerosis, pulmonary fibrosis, arthritis glomerulonephritis and post-surgical stenosis, restenosis, stroke, diabetes, hepatomegaly, Alzheimer's disease, cystic fibrosis, a hormone-related disease, an immunodeficiency disorder, a destructive bone disorder, an infectious disease, a condition associated with cell death, thrombin-induced platelet aggregation, chronic myelogenous leukaemia, liver disease, a pathologic immune condition involving T cell activation, CNS disorders, and other associated diseases.
11. A method of treatment of a disease in which inhibition of a PI3-K and/or mTOR is desired and/or required, which method comprises administration of a therapeutically effective amount of a compound of formula I as defined in any one of Claims 1 to 5, or a pharmaceutically-acceptable ester, amide, solvate or salt thereof, to a patient suffering from, or susceptible to, such a condition. 2. A combination product comprising:
(A) a compound of formula I as defined in any one of Claims 1 to 5, or a pharmaceutically-acceptable ester, amide, solvate or salt thereof; and (B) another therapeutic agent that is useful in the treatment of in the treatment of cancer and/or a proliferative disease,
wherein each of components (A) and (B) is formulated in admixture with a pharmaceutically-acceptable adjuvant, diluent or carrier.
13. A process for the preparation of a compound of formula I as defined in Claim 1 , which process comprises:
(i) reaction of a compound of formula II,
Figure imgf000095_0001
wherein L1 represents a suitable leaving group, and A1, A2, A3, A4, n, the dotted lines, B1, B1a, B2, B2a, B3, B3a, B4, B4a and R2 are as defined in Claim 1 , with a compound of formula III,
R3-L2 III
wherein L2 represents a suitable group and R3 is as defined in Claim 1 ;
(ii) reaction of a compound of formula IV,
Figure imgf000095_0002
wherein L represents a suitable leaving group, and A , A , A , A , n, the dotted lines and R3 are as defined in Claim 1 , with a compound of formula V,
Figure imgf000095_0003
wherein L4 may represent hydrogen (so forming an amine group), and L1, B1, B1a, B2, B2a, B3, B3a, B4 and B4a are as defined in Claim 1 ;
(iii) reaction of a compound of formula VI,
Figure imgf000096_0001
wherein A1, A2, A3, A4, n, the dotted lines and R3 are as hereinbefore defined, with a compound of formula V in which L4 represents hydrogen (so forming optionally substituted morpholine), which reaction may proceed by the reaction initially with a reagent that may convert the oxo moiety into a leaving group (and hence form a compound of formula IV in situ), for instance para-toluenesulfonyl chloride (e.g. in the presence of base (such as triethylamine or the like), a catalytic amount of DMAP, and the compound of formula VI in a suitable solvent such as dry acetonitrile or the like), followed by the addition of the compound of formula V in which L4 represents hydrogen;
(iv) for certain compounds of formula I, reaction of a compound of formula VII,
Figure imgf000096_0002
wherein (Ax) and (Ay) denotes the optional presence of the relevant A to A groups that are/may be present in the compound of formula I, and FG1 and FG2 independently represent mutually compatible functional groups, which may undergo an intramolecular reaction to form the requisite Ai to A^containing ring of formula I (and L1R3 represents R3 or L1, and R3, L1, B1, B1a, B2, B2a, B3, B3a, B4 and B4a are as defined in Claim 1 ; when L1R3 represents L1, then this step is followed by reaction with a compound of formula III as defined above); (v) for compounds of formula I in which there is a -N(R6)- moiety present, in which R6 represents Cv12 alkyl optionally substituted as hereinbefore defined (i.e. by one or more substituent(s) selected from E7 and =0), reaction of a corresponding compound of formula I in which R6 represents hydrogen, with either: a compound of formula VIII,
R6a-C(0)H VIII
wherein R6a represents CLU alkyl optionally substituted by one or more substituent(s) selected from E7 and =0, under reductive amination reaction conditions; or a compound of formula IX,
R6 -L1c IX
in which L1c represents a suitable leaving group and R6b represents CM2 alkyl optionally substituted by one or more substituents selected from =0 and E7;
(vi) for compounds of formula I in which there is a -N(R6)- moiety present, in which R6 represents -C(0)N(H)R11t, reaction of a corresponding compound of formula I in which R6 represents hydrogen, with a compound of formula X,
R1 ,-N=C=0 X wherein R11t is as defined in Claim 1 ;
(vii) for compounds of formula I in which there is a -N(R6)- moiety present, in which R6 represents -C(O)R10r or -S(O)2R10u, reaction of a corresponding compound of formula I in which R6 represents hydrogen, with a compound of formula XI,
G1-L1b XI
wherein G1 represents either -C(O)R10r or -S(O)2R10u, and L b (attached to the -C(O)- or -S(0)2 moieties) represents a suitable leaving group;
(viii) for compounds of formula I in which there is a -N(R6)- moiety present, in which R6 represents hydrogen, deprotection of a corresponding compound of formula I in which R6 represents a carboxylic acid (or ester group);
(ix) for compounds of formula I in which there is a hydroxy substituent present, transformation of a corresponding compound in which there is a methoxy group present;
(x) for compounds of formula I in which n represents 1 or 2, in which a (or the) A4 moiety adjacent to the requisite bicycle represents -C(O)-, A, represents -C(R )(R5)- and A2 represents -N(R6)-, reaction of a compound of formula XIA,
Figure imgf000098_0001
wherein (A4) denotes the optional presence of a further A4 group, and L1R3, B1, B1a, B2, B2a, B3, B3a, B4 and B a, A3, A4 and R6 are as defined in Claim 1 or above, with a compound of formula XIB,
R4-C(0)-R5 XIB
wherein R4 and R5 are as defined in Claim 1 followed by, if necessary, reaction with a compound of formula III as defined above.
14. A process for the preparation of a pharmaceutical formulation as defined in Claim 7, which process comprises bringing into association a compound of formula I, as defined in any one of one of Claims 1 to 5, or a pharmaceutically acceptable ester, amide, solvate or salt thereof with a pharmaceutically- acceptable adjuvant, diluent or carrier. 15. A process for the preparation of a combination product as defined in Claim 12, which process comprises bringing into association a compound of formula I, as defined in any one of Claims 1 to 5, or a pharmaceutically acceptable ester, amide, solvate or salt thereof with the other therapeutic agent that is useful in the treatment of cancer and/or a proliferative disease, and at least one pharmaceutically-acceptable adjuvant, diluent or carrier.
PCT/GB2011/001202 2010-08-11 2011-08-10 Tricyclic compounds for use as kinase inhibitors WO2012020227A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP10380104 2010-08-11
GB10380104.9 2010-08-11

Publications (1)

Publication Number Publication Date
WO2012020227A1 true WO2012020227A1 (en) 2012-02-16

Family

ID=43216221

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2011/001202 WO2012020227A1 (en) 2010-08-11 2011-08-10 Tricyclic compounds for use as kinase inhibitors

Country Status (1)

Country Link
WO (1) WO2012020227A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SU371209A1 (en) 1971-04-29 1973-02-22 Институт органической химии Н. Д. Зелинского METHOD OF OBTAINING 2-CYANOCYCLOGEXANDION-1,3
WO2002012242A2 (en) 2000-08-10 2002-02-14 Pharmacia Italia S.P.A. Bicyclo-pyrazoles active as kinase inhibitors, process for their preparation and pharmaceutical compositions comprising them
WO2009040552A2 (en) 2007-09-27 2009-04-02 Centro Nacional De Investigaciones Oncológicas (Cnio) Substituted imidazo (2, 1-b) -1, 3, 4-thiazole compounds, their pharmaceutical compositions and uses thereof
WO2009060197A1 (en) 2007-11-08 2009-05-14 Centro Nacional De Investigaciones Oncologicas (Cnio) Imidazopyridazines for use as protein kinase inhibitors

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SU371209A1 (en) 1971-04-29 1973-02-22 Институт органической химии Н. Д. Зелинского METHOD OF OBTAINING 2-CYANOCYCLOGEXANDION-1,3
WO2002012242A2 (en) 2000-08-10 2002-02-14 Pharmacia Italia S.P.A. Bicyclo-pyrazoles active as kinase inhibitors, process for their preparation and pharmaceutical compositions comprising them
WO2009040552A2 (en) 2007-09-27 2009-04-02 Centro Nacional De Investigaciones Oncológicas (Cnio) Substituted imidazo (2, 1-b) -1, 3, 4-thiazole compounds, their pharmaceutical compositions and uses thereof
WO2009060197A1 (en) 2007-11-08 2009-05-14 Centro Nacional De Investigaciones Oncologicas (Cnio) Imidazopyridazines for use as protein kinase inhibitors

Non-Patent Citations (52)

* Cited by examiner, † Cited by third party
Title
A. M. ABDEL MAGIB ET AL., J. ORG. CHEM., vol. 61, 1996, pages 3849
A.F. ABDEL-MAGID, C.A MARYANOFF., SYNTHESIS, 1990, pages 537
AKHREM ET AL.: "Bulletin of Academy of Sciences of USSR", DIVISION OF CHEMICAL SCIENCE (ENGLISH TRANSLATION, vol. 22, 1973, pages 836
ANDANAPPA K. GADAD ET AL., BIOORG. MED. CHEM., vol. 12, 2004, pages 5651 - 5659
ASUNCION MARIN ET AL., FARMACO, vol. 47, no. 1, 1992, pages 63 - 75
BIOORG. MED. CHEM. LETT., 2008, pages 4054
BRETONNET ET AL., J. MED. CHEM., vol. 50, 2007, pages 1872
BUNDEGAARD, H.: "Design of Prodrugs", 1985, ELESEVIER, pages: 1 - 92
BUNNEY, TOM D., KATAN, MATILDA: "Phosphoinositide signalling in cancer: beyond PI3K and PTEN", NATURE REVIEWS CANCER, vol. 10, no. 5, 2010, pages 342 - 352, XP002635531, DOI: doi:10.1038/NRC2842
CLEARY, JAMES M., SHAPIRO, GEOFFREY I.: "Development of phosphoinositide-3 kinase pathway inhibitors for advanced cancer", CURRENT ONCOLOGY REPORT, vol. 12, 2010, pages 87 - 94, XP055004477, DOI: doi:10.1007/s11912-010-0091-6
CURRENT OPINION IN CHEMICAL BIOLOGY, vol. 3, 1999, pages 459 - 465
E. ABIGNENTE, LL FARMACO, vol. 45, 1990, pages 1075
EASTON ET AL.: "mTOR and cancer therapy", ONCOGENE, vol. 25, no. 48, 2006, pages 6436 - 6446
F.D. BELLAMY, K. OU, TETRAHEDRON LETT., vol. 25, 1985, pages 839
FOLKES ADRIAN J ET AL: "The identification of 2-(1H-indazol-4-yl)-6-(4-methanesulfonyl-pipera zin-1-ylmethyl)-4-morpholin-4-yl-thieno[3,2-d]pyrimidine (GDC-0941) as a potent, selective, orally bioavailable inhibitor of class I PI3 kinase for the treatment of cancer", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, WASHINGTON, US, vol. 51, no. 18, 25 September 2008 (2008-09-25), pages 5522 - 5532, XP002592636, ISSN: 0022-2623, [retrieved on 20080829], DOI: DOI:10.1021/JM800295D *
HENNESSEY ET AL., NATURE REV. DRUG DISCOVERY, vol. 4, 2005, pages 988 - 1004
J. A. H. LAINTON, J. COMB. CHEM., vol. 5, 2003, pages 400
J. AM. CHEM. SOC., vol. 80, 1958, pages 1385 - 1387
J. KOBE ET AL., TETRAHEDRON, vol. 24, 1968, pages 239
KATSO, ANNU. REV. CELL. DEV. BOIL., vol. 17, 2001, pages 615 - 675
L. WENGWEI ET AL., TETRAHEDRON LETT., vol. 47, 2006, pages 1941
LESLIE ET AL., CHEM. REV., vol. 101, no. 8, 2001, pages 2365 - 2380
M. KUWAHARA ET AL., CHEM. PHARM BULL., vol. 44, 1996, pages 122
M. PLOTKIN ET AL., TETRAHEDRON LETT., vol. 41, 2000, pages 2269
M. SCHLOSSER ET AL.: "Organometallics in Synthesis. A Manual", 2002, WILEY &SONS LTD
M.A. EI-SHERBENY ET AL., BOLL. CHIM. FARM., vol. 136, 1997, pages 253 - 256
N. DEFACQZ ET AL., TETRAHEDRON LETT., vol. 44, 2003, pages 9111
NICOLAOU, K. C., BULGER, P. G., SARLAH, D., ANGEW. CHEM. INT. ED., vol. 44, 2005, pages 2 - 49
O. C. DERMER, CHEM. REV., vol. 14, 1934, pages 385
P.F. FABIO, A.F. LANZILOTTI, S.A. LANG, JOURNAL OF LABELLED COMPOUNDS AND PHARMACEUTICALS, vol. 15, 1978, pages 407
PARSONS ET AL., NATURE, vol. 436, 2005, pages 792
PAUL HEINZ ET AL., MONATSHEFTE FUR CHEMIE, vol. 108, 1977, pages 665 - 680
R. G. GLUSHKOV, T. V. STEZHKO, CHEMISTRY OF HETEROCYCLIC COMPOUNDS, vol. 14, no. 9, 1978, pages 1013 - 1016
S. Y. HAN, Y.-A. KIM., TETRAHEDRON, vol. 60, 2004, pages 2447
S.J. GREGSON ET AL., J. MED. CHEM., vol. 47, 2004, pages 1161
SEVERINSEN, R. ET AL., TETRAHEDRON, vol. 61, 2005, pages 5565 - 5575
SEYDEN-PENNE, J.: "Reductions by the Alumino and Borohydrides", 1991, VCH
SHINTANI, R., OKAMOTO, K., ORG. LETT., vol. 7, no. 21, 2005, pages 4757 - 4759
T. IKEMOTO ET AL., TETRAHEDRON, vol. 56, 2000, pages 7915
T. IKEMOTO, M. WAKIMASU, HETEROCYCLES, vol. 55, 2001, pages 99
T. W. GREENE, P. G. M. WUTS: "Protective Groups in Organic Synthesis", 1999, WILEY
T.W. GREENE, P.G.M. WUTZ: "Protective Groups in Organic Synthesis", 1999, WILEY-INTERSCIENCE
TETRAHEDRON LETT., 2009, pages 3436
TOKER, CELL. MOL. LIFE SCI., vol. 59, no. 5, 2002, pages 761 - 779
VAN DER HEIJDEN, MICHIEL S., BERNARDS, RENE: "Inhibition of the P13K Pathway: Hope We Can Believe in?", CLINICAL CANCER RESEARCH, vol. 16, 2010, pages 3094 - 3099, XP055004476, DOI: doi:10.1158/1078-0432.CCR-09-3004
VANHAESEBROECK, KATSO ET AL., EXP. CELL. RES., vol. 25, no. 1, 1999, pages 239 - 254
VANHAESEBROECK, TRENDS BIOCHEM. SCI, vol. 22, no. 87, 1997, pages 267 - 272
VANHAESEBROECK, TRENDS BIOCHEM. SCI., vol. 22, no. 7, 1997, pages 267 - 272
WERBER,G. ET AL., J. HETEROCYCL. CHEM.; EN, vol. 14, 1977, pages 823 - 827
WIGGINS, J. M., SYNTH. COMMUN., vol. 18, 1988, pages 741
WIPF, P., JUNG, J.-K., J. ORG. CHEM., vol. 65, no. 20, 2000, pages 6319 - 6337
ZH. KHIM., vol. 7, no. N245, 1974

Similar Documents

Publication Publication Date Title
US20130131057A1 (en) New bicyclic compounds as pi3-k and mtor inhibitors
JP5805623B2 (en) Imidazopyrazines for use as kinase inhibitors
CA2787714C (en) Inhibitors of pi3 kinase
KR101953210B1 (en) Macrocyclic Compounds as Protein Kinase Inhibitors
EP2414369B1 (en) Imidazo[2,1-b][1,3,4]thiadiazole derivatives
WO2013001310A1 (en) Macrocyclic compounds and their use as cdk8 inhibitors
EP2524918A1 (en) Imidazopyrazines derivates as kinase inhibitors
EP2536720A1 (en) Triazolo [4, 5 - b]pyridin derivatives
WO2012020215A1 (en) Amino- imidazolothiadiazoles for use as protein or lipid kinase inhibitors
WO2013005057A1 (en) New compounds
EP2480549B1 (en) Fused imidazo[3,2-d]pyrazines as PI3 kinase inhibitors
WO2012098387A1 (en) 6, 7-ring-fused triazolo [4, 3 - b] pyridazine derivatives as pim inhibitors
WO2013004984A1 (en) Tricyclic compounds for use as kinase inhibitors
WO2012020227A1 (en) Tricyclic compounds for use as kinase inhibitors
WO2013005041A1 (en) Tricyclic heterocyclic compounds as kinase inhibitors
WO2012020217A1 (en) Amino- imidazolothiadiazoles for use as protein or lipid kinase inhibitors
GM et al. the description, and pharmaceutically-acceptable esters, amides, solvates or salts thereof, which compounds are useful in the treat-ment of diseases in which inhibition of a protein or lipid kinase (eg a PIM family kinase, such as PIM-1, PIM-2 and/or PIM-3, and/or Flt-3) is desired and/or required, and particularly in the treatment of cancer or a proliferative disease.

Legal Events

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

Ref document number: 11757396

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 11757396

Country of ref document: EP

Kind code of ref document: A1