WO2013054185A1 - Pyrimidine and pyridine derivatives useful in therapy - Google Patents

Pyrimidine and pyridine derivatives useful in therapy Download PDF

Info

Publication number
WO2013054185A1
WO2013054185A1 PCT/IB2012/002073 IB2012002073W WO2013054185A1 WO 2013054185 A1 WO2013054185 A1 WO 2013054185A1 IB 2012002073 W IB2012002073 W IB 2012002073W WO 2013054185 A1 WO2013054185 A1 WO 2013054185A1
Authority
WO
WIPO (PCT)
Prior art keywords
heterocyclyl
phenyl
heteroaryl
compound
pharmaceutically acceptable
Prior art date
Application number
PCT/IB2012/002073
Other languages
French (fr)
Inventor
Scott A. Long
Atli Thorarensen
Mark E. Schnute
Original Assignee
Pfizer, Inc.
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 Pfizer, Inc. filed Critical Pfizer, Inc.
Publication of WO2013054185A1 publication Critical patent/WO2013054185A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • This invention relates to pyrimidine and pyridine derivatives useful in therapy, to processes for the preparation of such derivatives, to compositions containing such derivatives, and to the uses of such derivatives.
  • the pyrimidine and pyridine derivatives of the present invention have a number of therapeutic applications, particularly in the treatment of pain. This is believed to result from them being inhibitors of the enzyme autotaxin.
  • Autotaxin is a member of the family of nucleotide pyrophosphatases/phosphodiesterases (NPPl-7) and is also referred to as NPP2 [Stefan et al, "NPP-type ectophosphodiesterases: unity in diversity," Trends in Biochemical Sciences, vol. 30, no. 10, pp. 542-550, 2005].
  • ATX is a constitutively active enzyme possessing activity of phospholipase D. It hydrolyzes the head groups of lysophospho lipids (LPC) to lysophosphatidic acid (1 or 2-acyl- sn-glycerol-3-phosphate, LPA) and also acts on sphingosylphosphorylcholine to produce sphingosine 1 -phosphate (SIP) [Yuelling et al, "Autotaxin (ATX): a multifunctional and multi-modular protein possessing enzymatic lysoPLD activity and matricellular properties," Biochimica et Biophysica Acta, vol. 1781, no. 9, pp.
  • LPC lysophospho lipids
  • SIP sphingosine 1 -phosphate
  • ATX is the main source of blood LPA ( ⁇ 0.1 ⁇ plasma and ⁇ 1 ⁇ serum), but not S IP [Tanaka et al, "Autotaxin stabilizes blood vessels and is required for embryonic vasculature by producing lysophosphatidic acid," Journal of Biological Chemistry, vol. 281 , no. 35, pp. 25822-25830, 2006; Alvarez et al, “Autocrine and paracrine roles of sphingosine- 1 -phosphate,” Trends in Endocrinology and Metabolism, vol. 18, no. 8, pp. 300-307, 2007].
  • ATX hydrolyzes ATP: however, the affinity to ATP is at least 50-fold lower than for lysophospholipids [Gijsbers et al, "The hydrolysis of lysophospholipids and nucleotides by autotaxin (NPP2) involves a single catalytic site," FEBS Letters, vol. 538, no. 1- 3, pp. 60-64, 2003; Van Meeteren et al, "Regulation and biological activities of the autotaxin- LPA axis," Progress in Lipid Research, vol. 46, no. 2, pp. 145-160, 2007].
  • LPA acts on target cells through specific G-protein-coupled receptors (LPAl /Edg2, LPA2/Edg4, LPA3/Edg7, LPA4/GPR23/P2Y9, LPA5/GPR92, LPA6/P2Y5).
  • LPA receptors are broadly expressed throughout the body (neuronal, peripheral and central; platelets), and up-regulated in pathological conditions including for example fibrosis (renal, liver and lung), cancer (ovarian and breast), and osteoarthritis.
  • ATX an excellent drug target for the treatment of a number of pathologies including fibrosis (eg renal, lung, and liver), cancer (including ovarian cancer, prostate cancer and breast cancer), pain, osteoarthritis, rheumatoid arthritis, multiple sclerosis, atherosclerosis, thrombosis, psoriasis, diabetic neuropathy, neuropathies and inflammatory conditions. Consequently, selective ATX inhibitors have the potential to treat a variety of diseases that involve the LPC/LPA ATX pathway.
  • the pyrimidine and pyridine derivatives of the present invention are potentially useful in the treatment of a wide range of disorders, particularly pain, acute pain, chronic pain, neuropathic pain, inflammatory pain, osteoarthritis, visceral pain, nociceptive pain including post-surgical pain, and mixed pain types involving the viscera, gastrointestinal tract, cranial structures, musculoskeletal system, spine, urogenital system, cardiovascular system and CNS, including cancer pain, back and orofacial pain.
  • cancer including ovarian cancer, prostate cancer and breast cancer
  • atherosclerosis including ovarian cancer, prostate cancer and breast cancer
  • thrombosis including psoriasis, multiple sclerosis
  • fibrotic diseases including pulmonary fibrosis, Cirrhosis, endomyocardial fibrosis, mediastinal fibrosis, myelofibrosis, retroperitoneal fibrosis, nephrogenic systemic fibrosis, Crohn's disease, Keloid, old myocardial infarction, scleroderma / systemic sclerosis, atherofibrosis and adhesive capsulitis, neurodegenerative disorders, irritable bowel syndrome, rheumatoid arthritis, neuropathological disorders, functional bowel disorders, inflammatory bowel diseases, pain associated with dysmenorrhea, pelvic pain, cystitis, pancreatitis, migraine, cluster and tension headaches, diabetic neuropathy, peripheral neuropathic pain, sci
  • ATX inhibitors that are good drug candidates.
  • preferred compounds should bind potently to the ATX enzyme whilst showing little affinity for other receptors and enzymes which could give rise to unwanted side-effects, and should show functional activity as ATX inhibitors. They should be well absorbed from the gastrointestinal tract, be metabolically stable and possess favourable pharmacokinetic properties. They should be non-toxic and demonstrate few side-effects.
  • the ideal drug candidate will exist in a physical form that is stable, non-hygroscopic and easily formulated. The invention therefore provides a compound of formula I,
  • W represents a Ci-6 alkylene group which may be straight or branched
  • R 1 represents H, phenyl, naphthyi, heteroaryl 1 , heterocyclyl 1 or C3.6 cycloalkyl
  • each phenyl, naphthyi, heteroaryl 1 , heterocyclyl 1 and C3-5 cycloalkyl group is optionally substituted with from 1 to 3 substituents selected from halo, alkyl optionally substituted with 1-3 halogen atoms, phenyl, C e alkoxy optionally substituted with 1 -3 halogen atoms, cyano, heteroaryl la and heterocyclyl la ;
  • each phenyl, naphthyi, heteroaryl 1 , heterocyclyl 1 and C3.6 cycloalkyl group is optionally fused to a 5- or 6-membered heteroaromatic or heterocyclic ring containing from 1 to 3 heteroatoms (selected from N, O and S); and each heteroaryl 1 , heterocyclyl 1 and C 3- 6 cycloalkyl group is optionally fused to a benzene ring; and when the group is substituted the substitution may occur anywhere on the optionally fused ring system as a who le;
  • heterocyclyl 1 and heterocyclyl 1 a may additionally be substituted with
  • n 1 or 2;
  • n 0, 1 or 2;
  • V represents a bond or alkylene which may be straight or branched
  • R 2 represents H, -S-(Ci -6 alkyl), -S0 2 N(C, -6 alkyl) 2 , -C0 2 -(Ci. 6 alkyl), -NHCOCH 2 -phenyl,
  • each phenyl, naphthyi, heteroaryl 2 , heterocyclyl 2 and cycloalkyl group is optionally substituted with from 1 to 3 substituents selected from halo, Ci -6 alkyl optionally substituted with 1-3 halogen atoms, phenyl (optionally substituted with CN or Ci.6 alkyl), phenoxy, benzyloxy, N0 2 , -NHS0 2 (Ci -6 alkyl), -S0 2 (Ci.6 alkyl), pyridylmethyl, C .e alkoxy optionally substituted with 1-3 halogen atoms, cyano, heteroaryl 2a and heterocyclyl 23 ;
  • each phenyl, naphthyl, heteroaryl 2 , heterocyclyl 2 and cycloalkyl group is optionally fused to a 5- or 6-membered heteroaromatic or heterocyclic ring containing from 1 to 3 heteroatoms (selected from N, O and S); and wherein each heteroaryl 2 , heterocyclyl 2 and C3-6 cycloalkyl group is optionally fused to a benzene ring; and when the group is substituted the substitution may occur anywhere on the optionally fused ring system as a whole;
  • R represents heterocyclyl 2 ;
  • R 4 represents phenyl-(Ci.6 alkylene)-, wherein the phenyl group is optionally substituted with from 1 to 3 substituents selected from halo, Cj.6 alkyl optionally substituted with 1-3 halogen atoms, Ci-6 alkoxy optionally substituted with 1-3 halogen atoms, and cyano; heteroaryl 1 , heteroaryl 13 , heteroaryl 2 and heteroaryl 23 independently represent a 5- or 6-membered heteroaryl group containing from 1 to 3 heteroatoms (selected from N, O and S); and
  • heterocyclyl 1 , heterocyclyl 13 , heterocyclyl 2 and heterocyclyl 23 independently represent a 5- or 6-membered heterocyclyl group containing from 1 to 3 heteroatoms (selected from N, O and S);
  • halo means fluoro, chloro, bromo or iodo.
  • Alkyl, alkylene, and alkoxy groups, containing the requisite number of carbon atoms, can be unbranched or branched.
  • alkyl include methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, sec-butyl and t-butyl.
  • alkoxy include methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, i-butoxy, sec-butoxy and t-butoxy.
  • alkylene include methylene,
  • cycloalkyl examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • Heteroaryl is a radical formed from a heteroaromatic ring, for example thienyl, furanyl, pyrrolyl, pyrazolyl, imidazoyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl and tetrazolyl.
  • Heterocyclyl is a radical formed from a saturated or partially saturated heterocyclic ring, for example tetrahydrofuranyl, tetrahydrothiophenyl, pyrrolidinyl, tetrahydropyranyl, piperazinyl and imidazolidinyl.
  • Preferred groups of compounds are those in which:
  • W is C3 ⁇ 4, and pharmaceutically acceptable salts and solvates thereof;
  • n 1 , and pharmaceutically acceptable salts and solvates thereof;
  • n 1 , and pharmaceutically acceptable salts and solvates thereof;
  • A is CO, and pharmaceutically acceptable salts and solvates thereof;
  • R 1 is phenyl substituted by F or OCF 3 , and pharmaceutically acceptable salts and solvates thereof;
  • V is as defined herein;
  • R2 is as defined herein.
  • V is is selected from a bond or C1.5 alkylene which may be straight or branched.
  • V is C3 ⁇ 4 or CH2CH2, and pharmaceutically acceptable salts and solvates thereof;
  • R 2 is selected from H, -S-(Ci-6 alkyl), -S02N(Ci-6 alkyl)2, -C0 2 -(C,.
  • phenyl is optionally fused to tetrahydrofuran and said phenyl or dihydrobenzofuran is optionally substituted by 1 -2 substituents selected from CI, F, OCF 3 , N0 2 , OCH3, NS0 2 CH 3 , SO2CH3, OCH 2 Ph and OPh
  • heteroaryl 2 is a 5-6 membered heteroaryl group containing from 1 to 2 heteroatoms (selected from N, O and S) said heteroaryl group being optionally fused to a benzene ring and the optionally fused system may be substituted at any position by OPh;
  • R 2 is selected from H, -SCH 3 , -S0 2 N(CH3) 2 , -C0 2 -(C, -2 alkyl), -NHCOCH 2 -phenyl, -OCH 2 -phenyl, -S0 2 -phenyl, -NR 3 R 4 , phenyl, naphthyl, heteroaryl 2 , heterocyclyl 2 , -CO-4-methylpiperazin-l -yl or €3.5 cycloalkyl;
  • phenyl is optionally substituted by 1-2 substituents selected from CI, F, OCF3, N0 2 , OCH 3 , NSO2CH3, SO2CH3, OCH 2 Ph and OPh; or is optionally fused with tetrahydrofuran to form a 2,3-dihydrobenzofuran moiety.
  • heteroaryl 2 is selected from furanyl, 1 ,3-benzothiazolyl, pyridinyl substituted by a phenoxy group and an indazolyl;
  • heterocyclyl 2 is selected from oxoimidazolidinyl, isoindolin-l-one and pyrrolidin-2-one substituted by CH 2 -pyridine.
  • R 3 is tetrahydropyranyl
  • ⁇ R 4 is benzyl
  • Pharmaceutically acceptable salts of the compounds of formula I include the acid addition and base salts thereof. Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydro iodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, sac
  • Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts.
  • Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts.
  • the resulting salt may precipitate out and be collected by filtration or may be recovered by evaporation of the solvent.
  • the degree of ionisation in the resulting salt may vary from completely ionised to almost non- ionised.
  • the compounds of the invention may exist in both unsolvated and solvated forms.
  • the term 'solvate' is used herein to describe a molecular complex comprising the compound of the invention and a stoichiometric amount of one or more pharmaceutically acceptable solvent molecules, for example, ethanol.
  • the term 'hydrate' is employed when said solvent is water.
  • complexes such as clathrates, drug-host inclusion complexes wherein, in contrast to the aforementioned solvates, the drug and host are present in stoichiometric or non-stoichiometric amounts.
  • complexes of the drug containing two or more organic and/or inorganic components which may be in stoichiometric or non-stoichiometric amounts.
  • the resulting complexes may be ionised, partially ionised, or non-ionised.
  • references to compounds of formula I below include references to salts, solvates and complexes thereof and to solvates and complexes of salts thereof.
  • the compounds of the invention include all polymorphs and crystal habits thereof, prodrugs and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labeled compounds of formula I.
  • 'pro-drugs' of the compounds of formula I are also within the scope of the invention.
  • certain derivatives of compounds of formula I which may have little or no pharmacological activity themselves can, when administered into or onto the body, be converted into compounds of formula I having the desired activity, for example, by hydro lytic cleavage.
  • Such derivatives are referred to as 'prodrugs'.
  • Further information on the use of prodrugs may be found in Pro-drugs as Novel Delivery Systems, Vol. 14, ACS Symposium Series (T. Higuchi and W. Stella) and Bioreversible Carriers in Drug Design, Pergamon Press, 1987 (ed. E. B. Roche, American Pharmaceutical Association).
  • Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities present in the compounds of formula I with certain moieties known to those skilled in the art as 'pro-moieties' as described, for example, in Design of Prodrugs by H. Bundgaard (Elsevier, 1985).
  • prodrugs in accordance with the invention include
  • the compound of formula I contains a primary or secondary amino functionality (-N3 ⁇ 4 or -NHR where R ⁇ H), an amide thereof, for example, a compound wherein, as the case may be, one or both hydrogens of the amino functionality of the compound of formula I is/are replaced by (Ci-Cio)alkanoyl.
  • metabolites of compounds of formula I that is, compounds formed in vivo upon administration of the drug.
  • Compounds of formula I containing one or more asymmetric carbon atoms can exist as two or more stereoisomers. Where a compound of formula I contains an alkenyl or alkenylene group, geometric cisltrans (or Z/E) isomers are possible. Where structural isomers are interconvertible via a low energy barrier, tautomeric isomerism ('tautomerism') can occur. This can take the form of proton tautomerism in compounds of formula I containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism.
  • Cisltrans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallisation.
  • Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC).
  • HPLC high pressure liquid chromatography
  • the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound of formula I contains an acidic or basic moiety, a base or acid such as 1-phenylethylamine or tartaric acid.
  • the resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person.
  • Chiral compounds of the invention may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1 % diethylamine. Concentration of the eluate affords the enriched mixture.
  • Stereoisomeric conglomerates may be separated by conventional techniques known to those skilled in the art - see, for example, Stereochemistry of Organic Compounds by E. L. Eliel and S. H.
  • the present invention includes all pharmaceutically acceptable isotopically-labelled compounds of formula I wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number which predominates in nature.
  • isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2 H and 3 H, carbon, such as U C, ,3 C and 14 C, chlorine, such as 36 C1, fluorine, such as 18 F, iodine, such as 123 I and 125 1, nitrogen, such as 13 N and 15 N, oxygen, such as , 5 0, l 7 0 and l 8 0, phosphorus, such as 32 P, and sulphur, such as 3 S.
  • Certain isotopically-labelled compounds of formula I for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e.
  • Isotopically- labeled compounds of formula I can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically- labeled reagent in place of the non- labeled reagent previously employed.
  • Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D 2 O, de-acetone, d 6 -DMSO. All of the compounds of the formula (I) can be prepared by the procedures described in the general methods presented below or by the specific methods described in the Examples section and the Preparations section, or by routine modifications thereof.
  • the present invention also encompasses any one or more of these processes for preparing the compounds of formula (I), in addition to any novel intermediates used therein.
  • R , W, X, Y, Z, m and n are as defined above,
  • A, V and R are as defined above, and L is a leaving group.
  • suitable leaving groups are halogen, such as CI.
  • suitable leaving groups include OH, and peptide-coupling type procedures may be used, for example using HATU (2-(7-Aza-lH-benzotriazole-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate) and DIPEA (diisopropyl ethylamine) in THF at room temperature.
  • Hal is a halogen (such as Br or CI) and Pg is a protecting group, with a compound of formula V,
  • R 1 and W are as defined above, followed by removal of Pg.
  • the zincate coupling is carried out in the presence of a palladium(O) catalyst, such as Pd2(dba)3 [tris(dibenzylideneacetone)dipalladium(0)] in the presence of P(o-tolyl)3, in THF, at around 40-50°C.
  • a palladium(O) catalyst such as Pd2(dba)3 [tris(dibenzylideneacetone)dipalladium(0)] in the presence of P(o-tolyl)3, in THF, at around 40-50°C.
  • Suitable Pg groups include benzyl, which may be removed by hydrogenation in methanol containing aqueous HCI in the presence of Pd/C at a temperature in the range 20°C to the reflux temperature of the solvent, and a pressure of 1-100 atm (lxlO 5 - IxlO 7 N/m 2 ) hydrogen.
  • reaction by the action of PO(Hal)3.
  • the reaction is carried out in dichloromethane at room temperature.
  • the invention further provides a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, for use in the treatment of a disorder selected from: pain, acute pain, chronic pain, neuropathic pain, inflammatory pain, visceral pain, nociceptive pain including post-surgical pain, and mixed pain types involving the viscera, gastrointestinal tract, cranial structures, musculoskeletal system, spine, urogenital system, cardiovascular system and CNS, including cancer pain, back and orofacial pain.
  • a disorder selected from: pain, acute pain, chronic pain, neuropathic pain, inflammatory pain, visceral pain, nociceptive pain including post-surgical pain, and mixed pain types involving the viscera, gastrointestinal tract, cranial structures, musculoskeletal system, spine, urogenital system, cardiovascular system and CNS, including cancer pain, back and orofacial pain.
  • the invention further provides a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, for use in the treatment of a disorder selected from: cancer (including ovarian cancer, breast cancer and prostate cancer), atherosclerosis, thrombosis, psoriasis, multiple sclerosis, neurodegenerative disorders, irritable bowel syndrome, osteoarthritis, rheumatoid arthritis, neuropathological disorders, functional bowel disorders, inflammatory bowel diseases, pain associated with , dysmenorrhea, pelvic pain, cystitis, pancreatitis, migraine, cluster and tension headaches, diabetic neuropathy, peripheral neuropathic pain, sciatica, fibromyalgia, causalgia, and conditions of lower urinary tract dysfunction.
  • cancer including ovarian cancer, breast cancer and prostate cancer
  • atherosclerosis including ovarian cancer, breast cancer and prostate cancer
  • thrombosis psoriasis
  • multiple sclerosis neurodegenerative disorders
  • the invention further provides a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, for use in the treatment of fibrotic diseases.
  • fibrotic diseases include pulmonary fibrosis, Cirrhosis, endomyocardial fibrosis, mediastinal fibrosis, myelofibrosis, retroperitoneal fibrosis, nephrogenic systemic fibrosis, Crohn's disease, Keloid, old myocardial infarction, scleroderma / systemic sclerosis, atherofibrosis and adhesive capsulitis,
  • the invention further provides a pharmaceutical formulation comprising a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable adjuvant, diluent or carrier.
  • the pharmaceutical formulation may further comprise one or more additional active agents for the treatment of a disorder mentioned above.
  • the invention further provides a pharmaceutical kit comprising a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, and one or more additional active agents, as a combined preparation for separate, simultaneous or sequential administration in the treatment of a disorder mentioned above.
  • the invention further provides a method of treatment of a disorder mentioned above in a mammal (especially a human), comprising administration of a therapeutically effective amount of a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, to a mammal in need of such treatment.
  • Compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
  • excipients may be administered alone or in combination with one or more other compounds of the invention or in combination with one or more other drugs (or as any combination thereof). Generally, they will be administered as a formulation in association with one or more pharmaceutically acceptable excipients.
  • excipient' is used herein to describe any ingredient other than the compound(s) of the invention. The choice of excipient will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
  • compositions suitable for the delivery of compounds of the present invention and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company, 1995).
  • the compounds of the invention may be administered orally.
  • Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the blood stream directly from the mouth.
  • Formulations suitable for oral administration include solid formulations such as tablets, capsules containing particulates, liquids, or powders, lozenges (including
  • Liquid formulations include suspensions, solutions, syrups and elixirs. Such formulations may be employed as fillers in soft or hard capsules and typically comprise a carrier, for example, water, ethanol, polyethylene glycol, propylene glycol, methylcellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a solid, for example, from a sachet.
  • the compounds of the invention may also be used in fast-dissolving, fast-disintegrating dosage forms such as those described in Expert Opinion in Therapeutic Patents, ⁇ . (6), 981-986, by Liang and Chen (2001).
  • the drug may make up from 1 weight % to 80 weight % of the dosage form, more typically from 5 weight % to 60 weight % of the dosage form.
  • tablets generally contain a disintegrant.
  • disintegrants include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methyl cellulose, microcrystalline cellulose, lower alkyl-substituted hydroxypropyl cellulose, starch, pregelatinised starch and sodium alginate.
  • the disintegrant will comprise from 1 weight % to 25 weight %, preferably from 5 weight % to 20 weight % of the dosage form.
  • Binders are generally used to impart cohesive qualities to a tablet formulation. Suitable binders include microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinised starch, hydroxypropyl cellulose and hydroxypropyl methylcellulose. Tablets may also contain diluents, such as lactose (monohydrate, spray-dried monohydrate, anhydrous and the like), mannitol, xylitol, dextrose, sucrose, sorbitol, microcrystalline cellulose, starch and dibasic calcium phosphate dihydrate.
  • lactose monohydrate, spray-dried monohydrate, anhydrous and the like
  • mannitol xylitol
  • dextrose sucrose
  • sorbitol microcrystalline cellulose
  • starch dibasic calcium phosphate dihydrate
  • Tablets may also optionally comprise surface active agents, such as sodium lauryl sulfate and polysorbate 80, and glidants such as silicon dioxide and talc.
  • surface active agents such as sodium lauryl sulfate and polysorbate 80
  • glidants such as silicon dioxide and talc.
  • surface active agents may comprise from 0.2 weight % to 5 weight % of the tablet, and glidants may comprise from 0.2 weight % to 1 weight % of the tablet.
  • Tablets also generally contain lubricants such as magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, and mixtures of magnesium stearate with sodium lauryl sulphate.
  • Lubricants generally comprise from 0.25 weight % to 10 weight %, preferably from 0.5 weight % to 3 weight % of the tablet.
  • ingredients include anti-oxidants, colourants, flavouring agents, preservatives and taste-masking agents.
  • Exemplary tablets contain up to about 80% drug, from about 10 weight % to about 90 weight % binder, from about 0 weight % to about 85 weight % diluent, from about 2 weight % to about 10 weight % disintegrant, and from about 0.25 weight % to about 10 weight % lubricant.
  • Tablet blends may be compressed directly or by roller to form tablets. Tablet blends or portions of blends may alternatively be wet-, dry-, or melt-granulated, melt congealed, or extruded before tabletting.
  • the final formulation may comprise one or more layers and may be coated or uncoated; it may even be encapsulated.
  • the formulation of tablets is discussed in Pharmaceutical Dosage Forms: Tablets. Vol. 1, by H. Lieberman and L. Lachman (Marcel Dekker, New York, 1980).
  • Consumable oral films for human or veterinary use are typically pliable water-soluble or water-swellable thin film dosage forms which may be rapidly dissolving or mucoadhesive and typically comprise a compound of formula I, a film-forming polymer, a binder, a solvent, a humectant, a plasticiser, a stabiliser or emulsifier, a viscosity-modifying agent and a solvent. Some components of the formulation may perform more than one function.
  • the compound of the invention may be water-soluble or insoluble.
  • a water-soluble compound typically comprises from 1 weight % to 80 weight %, more typically from 20 weight % to 50 weight %, of the solutes. Less soluble compounds may comprise a greater proportion of the composition, typically up to 88 weight % of the solutes.
  • the compound of the invention may be in the form of multiparticulate beads.
  • the film-forming polymer may be selected from natural polysaccharides, proteins, or synthetic hydrocolloids and is typically present in the range 0.01 to 99 weight %, more typically in the range 30 to 80 weight %.
  • ingredients include anti-oxidants, colorants, flavourings and flavour enhancers, preservatives, salivary stimulating agents, cooling agents, co-solvents (including oils), emollients, bulking agents, anti-foaming agents, surfactants and taste-masking agents.
  • Films in accordance with the invention are typically prepared by evaporative drying of thin aqueous films coated onto a peelable backing support or paper. This may be done in a drying oven or tunnel, typically a combined coater dryer, or by freeze-drying or vacuuming.
  • Solid formulations for oral administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • Suitable modified release formulations for the purposes of the invention are described in US Patent No. 6,106,864. Details of other suitable release technologies such as high energy dispersions and osmotic and coated particles are to be found in Pharmaceutical Technology On-line. 25(2), 1-14, by Verma et al (2001). The use of chewing gum to achieve controlled release is described in WO 00/35298.
  • the compounds of the invention may also be administered directly into the blood stream, into muscle, or into an internal organ.
  • Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous.
  • Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
  • Parenteral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water.
  • excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9)
  • a suitable vehicle such as sterile, pyrogen-free water.
  • parenteral formulations under sterile conditions may readily be accomplished using standard pharmaceutical techniques well known to those skilled in the art.
  • solubility of compounds of the invention used in the preparation of parenteral solutions may be increased by the use of appropriate formulation techniques, such as the incorporation of solubility-enhancing agents.
  • Formulations for parenteral administration may be formulated to be immediate and/or modified release.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • compounds of the invention may be formulated as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active compound.
  • examples of such formulations include drug-coated stents and poly(i#-lactic-coglycolic)acid (PGLA) microspheres.
  • the compounds of the invention may also be administered topically to the skin or mucosa, that is, dermally or transdermal ly.
  • Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used.
  • Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporated - see, for example, J Pharm Sci, 88 (10), 955-958, by Finnin and Morgan (October 1999).
  • topical administration include delivery by electroporation, iontophoresis, phonophoresis, sonophoresis and microneedle or needle-free (e.g. PowderjectTM, BiojectTM, etc.) injection.
  • Formulations for topical administration may be formulated to be immediate and/or modified release.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • the compounds of the invention can also be administered intranasally or by inhalation, typically in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurised container, pump, spray, atomiser (preferably an atomiser using electrohydrodynamics to produce a fine mist), or nebuliser, with or without the use of a suitable propellant, such as 1,1 ,1,2-tetrafluoroethane or 1 , 1, 1,2,3,3,3-heptafluoropropane.
  • the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
  • the pressurised container, pump, spray, atomizer, or nebuliser contains a solution or suspension of the compound(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilising, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
  • a solution or suspension of the compound(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilising, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
  • the drug product Prior to use in a dry powder or suspension formulation, the drug product is micronised to a size suitable for delivery by inhalation (typically less than 5 microns). This may be achieved by any appropriate comminuting method, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenisation, or spray drying.
  • comminuting method such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenisation, or spray drying.
  • Capsules made, for example, from gelatin or hydroxypropylmethylcellulose
  • blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound of the invention, a suitable powder base such as lactose or starch and a performance modifier such as /-leucine, mannitol, or magnesium stearate.
  • the lactose may be anhydrous or in the form of the monohydrate, preferably the latter.
  • Other suitable excipients include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose and trehalose.
  • a suitable solution formulation for use in an atomiser using electrohydrodynamics to produce a fine mist may contain from ⁇ g to 20mg of the compound of the invention per actuation and the actuation volume may vary from ⁇ ⁇ to ⁇ .
  • a typical formulation may comprise a compound of formula I, propylene glycol, sterile water, ethanol and sodium chloride.
  • Alternative solvents which may be used instead of propylene glycol include glycerol and polyethylene glycol.
  • Suitable flavours such as menthol and Ievomenthol, or sweeteners, such as saccharin or saccharin sodium, may be added to those formulations of the invention intended for inhaled/intranasal administration.
  • Formulations for inhaled/intranasal administration may be formulated to be immediate and/or modified release using, for example, PGLA.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • the dosage unit is determined by means of a valve which delivers a metered amount.
  • Units in accordance with the invention are typically arranged to administer a metered dose or "puff containing from 1 to 10,000 ⁇ g of the compound of the invention.
  • the overall daily dose will typically be in the range ⁇ g to 10 mg which may be administered in a single dose or, more usually, as divided doses throughout the day.
  • the compounds of the invention may be administered rectally or vaginally, for example, in the form of a suppository, pessary, or enema. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate.
  • Formulations for rectal/vaginal administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
  • the compounds of the invention may also be administered directly to the eye or ear, typically in the form of drops of a micronised suspension or solution in isotonic, pH-adjusted, sterile saline.
  • Other formulations suitable for ocular and aural administration include ointments, biodegradable (e.g. absorbable gel sponges, collagen) and non-biodegradable (e.g. silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes.
  • a polymer such as crossed-linked polyacrylic acid, polyvinylalcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methyl cellulose, or a heteropoly saccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride.
  • a preservative such as benzalkonium chloride.
  • Such formulations may also be delivered by iontophoresis.
  • Formulations for ocular/aural administration may be formulated to be immediate and/or modified release.
  • Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted, or programmed release.
  • the compounds of the invention may be combined with soluble macromolecular entities, such as cyclodextrin and suitable derivatives thereof or polyethylene gly col-containing polymers, in order to improve their solubility, dissolution rate, taste-masking, bioavailability and/or stability for use in any of the aforementioned modes of administration.
  • Drug-cyclodextrin complexes for example, are found to be generally useful for most dosage forms and administration routes. Both inclusion and non- inclusion complexes may be used.
  • the cyclodextrin may be used as an auxiliary additive, i.e. as a carrier, diluent, or solubiliser.
  • compositions may conveniently be combined in the form of a kit suitable for coadministration of the compositions.
  • the kit of the invention comprises two or more separate pharmaceutical compositions, at least one of which contains a compound of formula I in accordance with the invention, and means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet.
  • a container, divided bottle, or divided foil packet An example of such a kit is the familiar blister pack used for the packaging of tablets, capsules and the like.
  • the kit of the invention is particularly suitable for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another.
  • the kit typically comprises directions for administration and may be provided with a so-called memory aid.
  • Navl .7 channel modulator such as a compound disclosed in WO 2009/012242
  • Navl .3 modulator e.g. as disclosed in WO2008/1 18758
  • Navl.8 modulator e.g. as disclosed in WO 2008/135826, more particularly N-[6-Amino-5-(2-chIoro-5-methoxyphenyl)pyridin-2-yl]-l-methyl-lH-pyrazole-5 -carboxamide
  • an inhibitor of nerve growth factor signaling such as: an agent that binds to NGF and inhibits NGF biological activity and/or downstream pathway(s) mediated by NGF signaling (e.g. tanezumab), a TrkA antagonist or a p75 antagonist;
  • a compound which increases the levels of endocannabinoid such as a compound with fatty acid amid hydrolase inhibitory (FAAH) activity, in particular those disclosed in WO 2008/047229 (e.g.
  • FAAH fatty acid amid hydrolase inhibitory
  • an opioid analgesic e.g. morphine, heroin, hydromorphone, oxymorphone, levorphanol, levallorphan, methadone, meperidine, fentanyl, cocaine, codeine, dihydrocodeine, oxycodone, hydrocodone, propoxyphene, nalmefene, nalorphine, naloxone, naltrexone, buprenorphine, butorphanol, nalbuphine or pentazocine; a nonsteroidal antiinflammatory drug (NSAID), e.g.
  • NSAID nonsteroidal antiinflammatory drug
  • a barbiturate sedative e.g. amobarbital, aprobarbital, butabarbital, butabital, mephobarbital, metharbital, methohexital, pentobarbital, phenobartital, secobarbital, talbutal, theamylal or thiopental;
  • a benzodiazepine having a sedative action e.g. chlordiazepoxide, clorazepate, diazepam, flurazepam, lorazepam, oxazepam, temazepam or triazolam;
  • an Hi antagonist having a sedative action e.g. diphenhydramine, pyrilamine, promethazine, chlorpheniramine or chlorcyclizine; a sedative such as glutethimide, meprobamate, methaqualone or dichloralphenazone;
  • a skeletal muscle relaxant e.g. baclofen, carisoprodol, chlorzoxazone, cyclobenzaprine, methocarbamol or orphrenadine;
  • an NMDA receptor antagonist e.g. dextromethorphan ((+)-3-hydroxy-N-methylmorphinan) or its metabolite dextrorphan ((+)-3-hydroxy-N-methylmorphinan), ketamine, memantine, pyrroloquinoline quinine, cis-4-(phosphonomethyl)-2-piperidinecarboxylic acid, budipine, EN-3231 (MorphiDex®, a combination formulation of morphine and dextromethorphan), topiramate, neramexane or perzinfotel including an NR2B antagonist, e.g. ifenprodil, traxoprodil or
  • an alpha-adrenergic e.g. doxazosin, tamsulosin, clonidine, guanfacine, dexmetatomidine, modafinil, or
  • a tricyclic antidepressant e.g. desipramine, imipramine, amitriptyline or nortriptyline;
  • an anticonvulsant e.g. carbamazepine, lamotrigine, topiratmate or valproate
  • a tachykinin (NK) antagonist particularly an NK-3, NK-2 or NK- antagonist, e.g. (aR,9R)-7-[3,5-bis(trifluoromethyl)benzyl]-8,9,10,l -tetrahydro-9-methyl-5-(4-m ethylphenyl)-7H-[l,4]diazocino[2,l-g][l,7]-naphthyridine-6-13-dione (TAK-637),
  • a muscarinic antagonist e.g oxybutynin, tolterodine, propiverine, tropsium chloride, darifenacin, solifenacin, temiverine and ipratropium; • a COX-2 selective inhibitor, e.g. celecoxib, rofecoxib, parecoxib, valdecoxib, deracoxib, etoricoxib, or lumiracoxib;
  • a neuroleptic such as droperidol, chlorpromazine, haloperidol, perphenazine, thioridazine, mesoridazine, trifluoperazine, fluphenazine, clozapine, olanzapine, risperidone, ziprasidone, quetiapine, sertindole, aripiprazole, sonepiprazole, blonanserin, iloperidone, perospirone, raclopride, zotepine, bifeprunox, asenapine, lurasidone, amisulpride, balaperidone, palindore, eplivanserin, osanetant, rimonabant, meclinertant, Miraxion® or sarizotan;
  • ⁇ a vanilloid receptor agonist e.g. resinferatoxin
  • antagonist e.g. capsazepine
  • a beta-adrenergic such as propranolol
  • a corticosteroid such as dexamethasone
  • a 5-HT receptor agonist or antagonist particularly a 5-HTIB / ID agonist such as eletriptan, sumatriptan, naratriptan, zolmitriptan or rizatriptan;
  • a 5-HT2A receptor antagonist such as R(+)-alpha-(2,3-dimethoxy-phenyl)-l -[2-(4-fluorophenylethyl)]-4-piperidinemeth anol (MDL- 100907);
  • a 5-HT3 antagonist such as ondansetron
  • a cholinergic (nicotinic) analgesic such as ispronicline (TC-1734), (E)-N-methyl-4-(3-pyridinyl)-3-buten-l-amine (RJR-2403), (R)-5-(2-azetidinylmethoxy)-2-chloropyridine (ABT-594) or nicotine;
  • a PDEV inhibitor such as 5-[2-ethoxy-5-(4-methyl-l-piperazinyl-sulphonyl)phenyl]-l-methyl-3-n-propyl-l ,
  • 6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one (sildenafil), (6R, 12aR)-2,3,6,7, 12,12a-hexahydro-2-methyl-6-(3,4-methylenedioxyphenyl)-pyr azino[2',l':6,l]-pyrido[3,4-b]indole-l,4-dione (IC-351 or tadalafil), 2-[2-ethoxy-5-(4-ethyl-piperazin- 1 -yl- 1 -sulphonyl)-phenyl]-5-methy l-7-propyl-3 H-imidazo[5,l-fJ[l,2,4]triazin-4-one (vardenafil), 5-(5-acetyl-2-butoxy-3-pyridinyl)-3-ethyl-2-(l-ethyl-3-azetidinyl)-2,6-d
  • an alpha-2-delta ligand such as gabapentin, pregabalin, 3-methylgabapentin, ( l a,3a,5a)(3-amino-methyl-bicyclo[3.2.0]hept-3-yl)-acetic acid, (3S,5R)-3-aminomethyl-5-methyl-heptanoic acid, (3S,5R)-3-amino-5-methyl-heptanoic acid, (3S,5R)-3-amino-5-methyl-octanoic acid, (2S,4S)-4-(3-chlorophenoxy)proline, (2S,4S)-4-(3-fluorobenzyl)-proline, [(lR,5R,6S)-6-(aminomethyl)bicyclo[3.2.0]hept-6-yl]acetic acid, 3-(l-ammomethyl-cyclohexylmethyl)-4H-[l,2,4]oxadiazol-5-one,
  • metabotropic glutamate subtype 1 receptor (mGluRl) antagonist metabotropic glutamate subtype 1 receptor (mGluRl) antagonist
  • a serotonin reuptake inhibitor such as sertraline, sertraline metabolite demethylsertraline, fluoxetine, norfluoxetine (fluoxetine desmethyl metabolite), fluvoxamine, paroxetine, citalopram, citalopram metabolite desmethylcitalopram, escitalopram, d,l- fenfluramine, femoxetine, ifoxetine, cyanodothiepin, litoxetine, dapoxetine, nefazodone, cericlamine and trazodone;
  • noradrenaline (norepinephrine) reuptake inhibitor such as maprotiline, lofepramine, mirtazepine, oxaprotiline, fezolamine, tomoxetine, mianserin, buproprion, buproprion metabolite hydroxybuproprion, nomifensine and viloxazine (Vivalan®), especially a selective noradrenaline reuptake inhibitor such as reboxetine, in particular (S,S)-reboxetine;
  • a dual serotonin-noradrenaline reuptake inhibitor such as venlafaxine, venlafaxine metabolite O-desmethylvenlafaxine, clomipramine, clomipramine metabolite desrnethylclomipramine, duloxetine, milnacipran and imipramine;
  • an inducible nitric oxide synthase (iNOS) inhibitor such as S-[2-[(l -iminoethyl)amino]ethyl]-L-homocysteine,
  • an acetylcholinesterase inhibitor such as donepezil
  • a prostaglandin E2 subtype 4 (EP4) antagonist such as N-[( ⁇ 2-[4-(2-ethyl-4,6-dimethyl-lH-imidazo[4,5-c]pyridin- l-yl)phenyl]ethyl ⁇ ami no)-carbonyl]-4-methylbenzenesulfonamide or 4-[(lS)-l -( ⁇ [5-chloro-2-(3-fluorophenoxy)pyridin-3-yl]carbonyl ⁇ amino)ethyl]ben zoic acid;
  • microsomal prostaglandin E synthase type 1 (mPGES-1) inhibitor • a microsomal prostaglandin E synthase type 1 (mPGES-1) inhibitor
  • a leukotriene B4 antagonist such as l -(3-biphenyl-4-ylmethyl-4-hydroxy-chroman-7-yl)-cyclopentanecarboxylic acid (CP- 105696), 5-[2-(2-Carboxyethyl)-3-[6-(4-methoxyphenyl)-5E- hexenyl]oxyphenoxy]-valeric acid ( ⁇ -4057) or DPC- 1870; and • a 5-lipoxygenase inhibitor, such as zileuton, 6-[(3-fluoro-5-[4-methoxy-3,4,5,6-tetrahydro-2H-pyran-4-yl])phenoxy-methyl]-l- methyl-2-quinolone (ZD-2138), or 2,3,5-trimethyl-6-(3-pyridylmethyl),l,4-benzoquinone (CV-6504).
  • a leukotriene B4 antagonist such as l -(3-biphen
  • additional active agents which may be part of a formulation or pharmaceutical kit according to the present invention, particularly for the treatment of cancer, include:
  • an anti-tumour platinum-based compound such as cisplatin or carboplatin
  • an anti-estrogen or selective estrogen receptor modulator such as tamoxifen, afimoxifene, arzoxifene, avalycoxifene, thioprine, thioprine, thioprine, thioprine, thioprine, thioprine, thioprine, thioprine, thioprine, thioprine, thioprine, thiothione, and others.
  • a tyrosine kinase inhibitor such as afatinib, imatinib, gefitinib, sorafenib, sunitinib, vandetanib, crizotinib or lapatinib.
  • the total daily dose of the compounds of the invention is typically in the range 0.5 mg to 3000 mg depending, of course, on the mode of administration.
  • oral administration may require a total daily dose of from 3 mg to 3000 mg, while an intravenous dose may only require from 0.5 mg to 500 mg.
  • the total daily dose may be administered in single or divided doses and may, at the physician's discretion, fall outside of the typical range given herein.
  • These dosages are based on an average human subject having a weight of about 60kg to 70kg. The physician will readily be able to determine doses for subjects whose weight falls outside this range, such as infants and the elderly.
  • references herein to "treatment” include references to curative, palliative and prophylactic treatment.
  • the biological activity of the compounds of the invention may be measured using the assay described below.
  • FS-3 is an LPC analogue that is conjugated with both a fluorophore and a quencher. In its native state the quencher interferes with the fluorophore's fluorescence. Once autotaxin cleaves FS-3, the fluorophore becomes liberated from the quencher, resulting in increased fluorescence. Thus increase in fluorescence is a measure of ATX activity. Any compound that inhibits the activity of Autotaxin will show less fluorescence read in presence of FS-3, which could be measured by fluorimetric analysis. See US Patent 7,989,663.
  • the FS-3 assay to identify ATX inhibitors was preformed as follows: 3 ⁇ of standard inhibitor (referred to as PF-8380 in Gierse et al above) and test compounds were added to an assay plate. To each assay well, containing test compounds or standard, 24 ⁇ of human Autotaxin enzyme (2 nM) was added. The assay plate was then centrifuged at 1000 rpm for 1 minute and allowed to incubate at 37°C for 30 minutes. Following the incubation period each plate was read in a fluorescence plate reader (Spectra Max M5: excitation: 494nm and emission: 520nm) and IC50 values were derived from inhibition of FS-3 fluorescence (as described above).
  • UV - Acquity PDA Varian ELSD (evaporative light scattering detector)

Landscapes

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

Abstract

The invention provides compounds of formula (I), wherein one of X, Y and Z represents N and the other two represent CH, or X=Y-Z represents N=CH-N; W represents a CI-6 alkylene group which may be straight or branched; R1 represents H, phenyl, naphthyl, heteroaryl1, heterocyclyl1 or C3-6 cycloalkyl; wherein each phenyl, naphthyl, heteroaryl', heterocyclyl1 and C3-6 cycloalkyl group is optionally substituted, and wherein each phenyl, naphthyl, heteroaryl1, heterocyclyl1 and C3-6 cycloalkyl group is optionally fused to a 5- or 6-membered heteroaromatic or heterocyclic ring containing from 1 to 3 heteroatoms (selected from N, O and S); and each heteroaryl1, heterocyclyl1 and C3-6 cycloalkyl group is optionally fused to a benzene ring; m is 1 or 2; n is 0, 1 or 2; A is C=O or SO2; V represents a bond or C1-8 alkylene which may be straight or branched; R2 represents H, -S-(C1-6 alkyl), -SO2N(C1-6 alkyl)2, -CO2-(C1-6 alkyl), - NHCOCH2-phenyl, -OCH2- phenyl, -S02-phenyl, -NR3R4, phenyl, naphthyl, heteroaryl2, heterocyclyl2, -CO-heterocyclyl2 or C3-6 cycloalkyl; wherein each phenyl, naphthyl, heteroaryl2, heterocyclyl2 and C3-6 cycloalkyl group is optionally substituted, and wherein each phenyl, naphthyl, heteroaryl2, heterocyclyl2 and C3-6 cycloalkyl group is optionally fused to a 5- or 6-membered heteroaromatic or heterocyclic ring containing from 1 to 3 heteroatoms (selected from N, O and S); and wherein each heteroaryl2, heterocyclyl2 and C3-6 cycloalkyl group is optionally fused to a benzene ring; and pharmaceutically acceptable salts and solvates thereof. The compounds are useful as pharmaceuticals, particularly in the treatment of fibrotic diseases, cancer and pain.

Description

Pyrimidine and pyridine derivatives useful in therapy
This invention relates to pyrimidine and pyridine derivatives useful in therapy, to processes for the preparation of such derivatives, to compositions containing such derivatives, and to the uses of such derivatives.
The pyrimidine and pyridine derivatives of the present invention have a number of therapeutic applications, particularly in the treatment of pain. This is believed to result from them being inhibitors of the enzyme autotaxin.
Autotaxin (ATX) is a member of the family of nucleotide pyrophosphatases/phosphodiesterases (NPPl-7) and is also referred to as NPP2 [Stefan et al, "NPP-type ectophosphodiesterases: unity in diversity," Trends in Biochemical Sciences, vol. 30, no. 10, pp. 542-550, 2005]. It is a glycoprotein with four possible N-glycosylation sites, synthesized as a pre-proenzyme and is secreted to extracellular space following two N-terminal cleavages (27 and 8 amino acids) [Stracke et al, "Autotaxin is an N-linked glycoprotein but the sugar moieties are not needed for its stimulation of cellular motility," Melanoma Research, vol. 5, no. 4, pp. 203-209, 1995; Jansen et al, "Proteolytic maturation and activation of autotaxin (NPP2), a secreted metastasis-enhancing lysophospholipase D," Journal of Cell Science, vol. 1 18, no. 14, pp. 3081-3089, 2005].
ATX is a constitutively active enzyme possessing activity of phospholipase D. It hydrolyzes the head groups of lysophospho lipids (LPC) to lysophosphatidic acid (1 or 2-acyl- sn-glycerol-3-phosphate, LPA) and also acts on sphingosylphosphorylcholine to produce sphingosine 1 -phosphate (SIP) [Yuelling et al, "Autotaxin (ATX): a multifunctional and multi-modular protein possessing enzymatic lysoPLD activity and matricellular properties," Biochimica et Biophysica Acta, vol. 1781, no. 9, pp. 525-530, 2008; Nakanaga et al, "Autotaxin- An LPA producing enzyme with diverse functions," Journal of Bio chemistry, vol. 148, no. 1, pp. 13-24, 2010]. Both LPA and SIP are strong inhibitors of ATX with affinity to enzyme approximately 1000-fold higher than reported for ATX substrates [Van Meeteren et al, "Inhibition of autotaxin by lysophosphatidic acid and sphingosine 1-phosphate," Journal of Biological Chemistry, vol. 280, no. 22, pp. 21155-21161, 2005]. The expression of Autotaxin has been detected in brain, ovary, lung, intestine, and kidney. Autotaxin protein has been detected in blood, cerebrospinal, seminal fluid, urine and saliva [Sugiura et al, "Lysophosphatidic acid, a growth factor- like lipid, in the saliva," Journal of Lipid Research, vo l. 43, no. 1 2, pp. 2049-2055, 2002; Tanaka et al, "Prostatic acid phosphatase degrades lysophosphatidic acid in seminal plasma," FEBS Letters, vol. 571, no. 1-3, pp. 197-204, 2004; Masuda et al, "Serum autotaxin measurement in haematological malignancies: a promising marker for follicular lymphoma," British Journal of Haematology, vol. 143, no. 1 , pp. 60-70, 2008; Nakamura et al, "Analysis of serum and urinary lysophospho lipase D/autotaxin in nephrotic syndrome," Clinical Chemistry and Laboratory Medicine, vo l. 46, no. 1 , pp. 150-1 51 , 2008; Nakamura et al, "Autotaxin enzyme immunoassay in human cerebrospinal fluid samples," Clinica Chimica Acta, vol. 405, no. 1 -2, pp. 160-162, 2009].
ATX is the main source of blood LPA (~0.1 μΜ plasma and ~ 1 μΜ serum), but not S IP [Tanaka et al, "Autotaxin stabilizes blood vessels and is required for embryonic vasculature by producing lysophosphatidic acid," Journal of Biological Chemistry, vol. 281 , no. 35, pp. 25822-25830, 2006; Alvarez et al, "Autocrine and paracrine roles of sphingosine- 1 -phosphate," Trends in Endocrinology and Metabolism, vol. 18, no. 8, pp. 300-307, 2007]. In addition to LPC and sphingosylphosphorylcholine, ATX hydrolyzes ATP: however, the affinity to ATP is at least 50-fold lower than for lysophospholipids [Gijsbers et al, "The hydrolysis of lysophospholipids and nucleotides by autotaxin (NPP2) involves a single catalytic site," FEBS Letters, vol. 538, no. 1- 3, pp. 60-64, 2003; Van Meeteren et al, "Regulation and biological activities of the autotaxin- LPA axis," Progress in Lipid Research, vol. 46, no. 2, pp. 145-160, 2007].
The major product of ATX, LPA acts on target cells through specific G-protein-coupled receptors (LPAl /Edg2, LPA2/Edg4, LPA3/Edg7, LPA4/GPR23/P2Y9, LPA5/GPR92, LPA6/P2Y5). LPA receptors are broadly expressed throughout the body (neuronal, peripheral and central; platelets), and up-regulated in pathological conditions including for example fibrosis (renal, liver and lung), cancer (ovarian and breast), and osteoarthritis.
[Aoki et al, "Two pathways for lysophosphatidic acid production," Biochimica et Biophysica Acta, vol. 1781 , no. 9, pp. 513-518, 2008; Okudaira et al, "Biological roles of lysophosphatidic acid signaling through its production by autotaxin," Biochimie, vol. 92, no. 6, pp. 698-706, 2010]. The wide distribution of LPA receptors and the corresponding increase in ATX expression in disease makes ATX an excellent drug target for the treatment of a number of pathologies including fibrosis (eg renal, lung, and liver), cancer (including ovarian cancer, prostate cancer and breast cancer), pain, osteoarthritis, rheumatoid arthritis, multiple sclerosis, atherosclerosis, thrombosis, psoriasis, diabetic neuropathy, neuropathies and inflammatory conditions. Consequently, selective ATX inhibitors have the potential to treat a variety of diseases that involve the LPC/LPA ATX pathway.
The pyrimidine and pyridine derivatives of the present invention are potentially useful in the treatment of a wide range of disorders, particularly pain, acute pain, chronic pain, neuropathic pain, inflammatory pain, osteoarthritis, visceral pain, nociceptive pain including post-surgical pain, and mixed pain types involving the viscera, gastrointestinal tract, cranial structures, musculoskeletal system, spine, urogenital system, cardiovascular system and CNS, including cancer pain, back and orofacial pain. Other conditions that may be treated with the pyrimidine and pyridine derivatives of the present invention include cancer (including ovarian cancer, prostate cancer and breast cancer), atherosclerosis, thrombosis, psoriasis, multiple sclerosis, fibrotic diseases including pulmonary fibrosis, Cirrhosis, endomyocardial fibrosis, mediastinal fibrosis, myelofibrosis, retroperitoneal fibrosis, nephrogenic systemic fibrosis, Crohn's disease, Keloid, old myocardial infarction, scleroderma / systemic sclerosis, atherofibrosis and adhesive capsulitis, neurodegenerative disorders, irritable bowel syndrome, rheumatoid arthritis, neuropathological disorders, functional bowel disorders, inflammatory bowel diseases, pain associated with dysmenorrhea, pelvic pain, cystitis, pancreatitis, migraine, cluster and tension headaches, diabetic neuropathy, peripheral neuropathic pain, sciatica, fibromyalgia, causalgia, and conditions of lower urinary tract dysfunction.
There is a need to provide new ATX inhibitors that are good drug candidates. In particular, preferred compounds should bind potently to the ATX enzyme whilst showing little affinity for other receptors and enzymes which could give rise to unwanted side-effects, and should show functional activity as ATX inhibitors. They should be well absorbed from the gastrointestinal tract, be metabolically stable and possess favourable pharmacokinetic properties. They should be non-toxic and demonstrate few side-effects. Furthermore, the ideal drug candidate will exist in a physical form that is stable, non-hygroscopic and easily formulated. The invention therefore provides a compound of formula I,
Figure imgf000005_0001
wherein
one of X, Y and Z represents N and the other two represent CH, or X=Y-Z represents N=CH-N;
W represents a Ci-6 alkylene group which may be straight or branched;
R1 represents H, phenyl, naphthyi, heteroaryl1 , heterocyclyl1 or C3.6 cycloalkyl;
wherein each phenyl, naphthyi, heteroaryl1, heterocyclyl1 and C3-5 cycloalkyl group is optionally substituted with from 1 to 3 substituents selected from halo,
Figure imgf000005_0002
alkyl optionally substituted with 1-3 halogen atoms, phenyl, C e alkoxy optionally substituted with 1 -3 halogen atoms, cyano, heteroarylla and heterocyclylla;
and wherein each phenyl, naphthyi, heteroaryl1, heterocyclyl1 and C3.6 cycloalkyl group is optionally fused to a 5- or 6-membered heteroaromatic or heterocyclic ring containing from 1 to 3 heteroatoms (selected from N, O and S); and each heteroaryl1, heterocyclyl1 and C3-6 cycloalkyl group is optionally fused to a benzene ring; and when the group is substituted the substitution may occur anywhere on the optionally fused ring system as a who le;
and wherein heterocyclyl1 and heterocyclyl1 a may additionally be substituted with
=0;
m is 1 or 2;
n is 0, 1 or 2;
A is C=0 or S02;
V represents a bond or
Figure imgf000005_0003
alkylene which may be straight or branched;
R2 represents H, -S-(Ci-6 alkyl), -S02N(C,-6 alkyl)2, -C02-(Ci.6 alkyl), -NHCOCH2-phenyl,
-OCH2-phenyl, -S02-phenyl, -NR3R4, phenyl, naphthyi, heteroaryl2, heterocyclyl2 ,
-CO-heterocyclyl2 or C3.6 cycloalkyl;
wherein each phenyl, naphthyi, heteroaryl2, heterocyclyl2 and cycloalkyl group is optionally substituted with from 1 to 3 substituents selected from halo, Ci-6 alkyl optionally substituted with 1-3 halogen atoms, phenyl (optionally substituted with CN or Ci.6 alkyl), phenoxy, benzyloxy, N02, -NHS02(Ci-6 alkyl), -S02(Ci.6 alkyl), pyridylmethyl, C .e alkoxy optionally substituted with 1-3 halogen atoms, cyano, heteroaryl2a and heterocyclyl23;
and wherein each phenyl, naphthyl, heteroaryl2, heterocyclyl2 and cycloalkyl group is optionally fused to a 5- or 6-membered heteroaromatic or heterocyclic ring containing from 1 to 3 heteroatoms (selected from N, O and S); and wherein each heteroaryl2, heterocyclyl2 and C3-6 cycloalkyl group is optionally fused to a benzene ring; and when the group is substituted the substitution may occur anywhere on the optionally fused ring system as a whole;
and wherein heterocyclyl2 and heterocyclyl23 may additionally be substituted with =0;
R represents heterocyclyl2;
R4 represents phenyl-(Ci.6 alkylene)-, wherein the phenyl group is optionally substituted with from 1 to 3 substituents selected from halo, Cj.6 alkyl optionally substituted with 1-3 halogen atoms, Ci-6 alkoxy optionally substituted with 1-3 halogen atoms, and cyano; heteroaryl1, heteroaryl13, heteroaryl2 and heteroaryl23 independently represent a 5- or 6-membered heteroaryl group containing from 1 to 3 heteroatoms (selected from N, O and S); and
heterocyclyl1, heterocyclyl13, heterocyclyl2 and heterocyclyl23 independently represent a 5- or 6-membered heterocyclyl group containing from 1 to 3 heteroatoms (selected from N, O and S);
and pharmaceutically acceptable salts and solvates thereof (also referred to herein as "the compounds of the invention").
In the above definitions, halo means fluoro, chloro, bromo or iodo. Alkyl, alkylene, and alkoxy groups, containing the requisite number of carbon atoms, can be unbranched or branched. Examples of alkyl include methyl, ethyl, n-propyl, i-propyl, n-butyl, i-butyl, sec-butyl and t-butyl. Examples of alkoxy include methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, i-butoxy, sec-butoxy and t-butoxy. Examples of alkylene include methylene,
1.1 - ethylene, 1 ,2-ethyIene, 1 , 1 -propylene, 1 ,2-propylene, 1 ,3-propylene and
2.2- propylene. Examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. "Heteroaryl" is a radical formed from a heteroaromatic ring, for example thienyl, furanyl, pyrrolyl, pyrazolyl, imidazoyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl and tetrazolyl. Specific R2 groups of interest comprising heteroaryl2 include benzothiazolyl (heteroaryl2 = thiazolyl fused to a benzene ring), furyl, indazolyl (heteroaryl2 = pyrazolyl fused to a benzene ring) and indolyl (heteroaryl2 = pyrrolyl fused to a benzene ring).
"Heterocyclyl" is a radical formed from a saturated or partially saturated heterocyclic ring, for example tetrahydrofuranyl, tetrahydrothiophenyl, pyrrolidinyl, tetrahydropyranyl, piperazinyl and imidazolidinyl. Specific R2 groups of interest comprising heterocyclyl2 include isoindolin-l -onyl (heterocyclyl2 = pyrrolidinyl substituted with =0 and fused to a benzene ring), pyrroiidinonyl optionally substituted with pyridinylmethyl (heterocyclyl2 = pyrrolidinyl substituted with =0), piperazinyl optionally substituted with methyl and 2-oxoimidazolinyl optionally substituted with benzonitrile (heterocyclyl2 = imidazolidinyl substituted with =0).
Preferred groups of compounds are those in which:
X=Y-Z represents N=CH-N, and pharmaceutically acceptable salts and solvates thereof;
W is C¾, and pharmaceutically acceptable salts and solvates thereof;
m is 1 , and pharmaceutically acceptable salts and solvates thereof;
n is 1 , and pharmaceutically acceptable salts and solvates thereof;
A is CO, and pharmaceutically acceptable salts and solvates thereof;
R1 is phenyl substituted by F or OCF3, and pharmaceutically acceptable salts and solvates thereof;
V is as defined herein; and
R2 is as defined herein.
In a preferred embodiment V is is selected from a bond or C1.5 alkylene which may be straight or branched.In a particularly preferred embodiment V is C¾ or CH2CH2, and pharmaceutically acceptable salts and solvates thereof; In a preferred embodiment R2 is selected from H, -S-(Ci-6 alkyl), -S02N(Ci-6 alkyl)2, -C02-(C,.6 alkyl), - HCOCH2-phenyl, -OCH2-phenyl, -S02-phenyl, -NR3R4, phenyl, naphthyl, heteroaryl2, heterocyclyl2 -CO-4-methylpiperazin-l-yl or C3.5 cycloatkyl;
Wherein phenyl is optionally fused to tetrahydrofuran and said phenyl or dihydrobenzofuran is optionally substituted by 1 -2 substituents selected from CI, F, OCF3, N02, OCH3, NS02CH3, SO2CH3, OCH2Ph and OPh
Wherein heteroaryl2 is a 5-6 membered heteroaryl group containing from 1 to 2 heteroatoms (selected from N, O and S) said heteroaryl group being optionally fused to a benzene ring and the optionally fused system may be substituted at any position by OPh;
Wherein heterocyclyl2 is a 5 membered heterocyclyl group containing 1-2 N atoms, said heterocyclyl group being optionally fused to a benzene ring and the optionally fused system may be substituted at any position by =0, CH2Ph or CH2-C5NH5
In a particularly preferred embodiment R2 is selected from H, -SCH3, -S02N(CH3)2, -C02-(C,-2 alkyl), -NHCOCH2-phenyl, -OCH2-phenyl, -S02-phenyl, -NR3R4, phenyl, naphthyl, heteroaryl2, heterocyclyl2 , -CO-4-methylpiperazin-l -yl or€3.5 cycloalkyl;
Wherein phenyl is optionally substituted by 1-2 substituents selected from CI, F, OCF3, N02, OCH3, NSO2CH3, SO2CH3, OCH2Ph and OPh; or is optionally fused with tetrahydrofuran to form a 2,3-dihydrobenzofuran moiety.
Wherein heteroaryl2 is selected from furanyl, 1 ,3-benzothiazolyl, pyridinyl substituted by a phenoxy group and an indazolyl;
Wherein heterocyclyl2 is selected from oxoimidazolidinyl, isoindolin-l-one and pyrrolidin-2-one substituted by CH2-pyridine.
In a preferred embodiment R3 is tetrahydropyranyl; and
· R4 is benzyl.
Pharmaceutically acceptable salts of the compounds of formula I include the acid addition and base salts thereof. Suitable acid addition salts are formed from acids which form non-toxic salts. Examples include the acetate, aspartate, benzoate, besylate, bicarbonate/carbonate, bisulphate/sulphate, borate, camsylate, citrate, edisylate, esylate, formate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hibenzate, hydrochloride/chloride, hydrobromide/bromide, hydro iodide/iodide, isethionate, lactate, malate, maleate, malonate, mesylate, methylsulphate, naphthylate, 2-napsylate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/hydrogen phosphate/dihydrogen phosphate, saccharate, stearate, succinate, tartrate, tosylate and trifluoroacetate salts. Suitable base salts are formed from bases which form non-toxic salts. Examples include the aluminium, arginine, benzathine, calcium, choline, diethylamine, diolamine, glycine, lysine, magnesium, meglumine, olamine, potassium, sodium, tromethamine and zinc salts.
Hemisalts of acids and bases may also be formed, for example, hemisulphate and hemicalcium salts.
For a review on suitable salts, see Handbook of Pharmaceutical Salts: Properties, Selection, and Use by Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002). Pharmaceutically acceptable salts of compounds of formula I may be prepared by one or more of three methods:
(i) by reacting the compound of formula I with the desired acid or base;
(ii) by removing an acid- or base-labile protecting group from a suitable precursor of the compound of formula I or by ring-opening a suitable cyclic precursor, for example, a lactone or lactam, using the desired acid or base; or
(iii) by converting one salt of the compound of formula I to another by reaction with an appropriate acid or base or by means of a suitable ion exchange column.
All three reactions are typically carried out in solution. The resulting salt may precipitate out and be collected by filtration or may be recovered by evaporation of the solvent. The degree of ionisation in the resulting salt may vary from completely ionised to almost non- ionised.
The compounds of the invention may exist in both unsolvated and solvated forms. The term 'solvate' is used herein to describe a molecular complex comprising the compound of the invention and a stoichiometric amount of one or more pharmaceutically acceptable solvent molecules, for example, ethanol. The term 'hydrate' is employed when said solvent is water. Included within the scope of the invention are complexes such as clathrates, drug-host inclusion complexes wherein, in contrast to the aforementioned solvates, the drug and host are present in stoichiometric or non-stoichiometric amounts. Also included are complexes of the drug containing two or more organic and/or inorganic components which may be in stoichiometric or non-stoichiometric amounts. The resulting complexes may be ionised, partially ionised, or non-ionised. For a review of such complexes, see J Pharm Sci, 64 (8), 1269-1288, by Haleblian (August 1975).
When appropriate to the context, references to compounds of formula I below include references to salts, solvates and complexes thereof and to solvates and complexes of salts thereof.
The compounds of the invention include all polymorphs and crystal habits thereof, prodrugs and isomers thereof (including optical, geometric and tautomeric isomers) as hereinafter defined and isotopically-labeled compounds of formula I.
As indicated, so-called 'pro-drugs' of the compounds of formula I are also within the scope of the invention. Thus certain derivatives of compounds of formula I which may have little or no pharmacological activity themselves can, when administered into or onto the body, be converted into compounds of formula I having the desired activity, for example, by hydro lytic cleavage. Such derivatives are referred to as 'prodrugs'. Further information on the use of prodrugs may be found in Pro-drugs as Novel Delivery Systems, Vol. 14, ACS Symposium Series (T. Higuchi and W. Stella) and Bioreversible Carriers in Drug Design, Pergamon Press, 1987 (ed. E. B. Roche, American Pharmaceutical Association).
Prodrugs in accordance with the invention can, for example, be produced by replacing appropriate functionalities present in the compounds of formula I with certain moieties known to those skilled in the art as 'pro-moieties' as described, for example, in Design of Prodrugs by H. Bundgaard (Elsevier, 1985).
Some examples of prodrugs in accordance with the invention include
(i) where the compound of formula I contains a carboxylic acid functionality
(-COOH), an ester thereof, for example, a compound wherein the hydrogen of the carboxylic acid functionality of the compound of formula (I) is replaced by (Ci-Cs)alkyl; (ii) where the compound of formula I contains an alcohol functionality (-OH), an ether thereof, for example, a compound wherein the hydrogen of the alcohol functionality of the compound of formula I is replaced by (Ci-C6)alkanoyloxymethyl; and
(iii) where the compound of formula I contains a primary or secondary amino functionality (-N¾ or -NHR where R≠ H), an amide thereof, for example, a compound wherein, as the case may be, one or both hydrogens of the amino functionality of the compound of formula I is/are replaced by (Ci-Cio)alkanoyl.
Further examples of replacement groups in accordance with the foregoing examples and examples of other prodrug types may be found in the aforementioned references.
Moreover, certain compounds of formula I may themselves act as prodrugs of other compounds of formula I. Also included within the scope of the invention are metabolites of compounds of formula I, that is, compounds formed in vivo upon administration of the drug. Some examples of metabolites in accordance with the invention include
(i) where the compound of formula I contains a methyl group, an hydroxymethyl derivative thereof (-CH3 -> -CH2OH):
(ii) where the compound of formula I contains an alkoxy group, an hydroxy derivative thereof (-OR -> -OH);
(iii) where the compound of formula I contains a tertiary amino group, a secondary amino derivative thereof (-NR'R2 -> -NHR1 or -NHR2);
(iv) where the compound of formula I contains a secondary amino group, a primary derivative thereof (-NHR1 -> -NH2);
(v) where the compound of formula I contains a phenyl moiety, a phenol derivative thereof (-Ph -> -PhOH); and
(vi) where the compound of formula I contains an amide group, a carboxylic acid derivative thereof (-CONH2 -> COOH).
Compounds of formula I containing one or more asymmetric carbon atoms can exist as two or more stereoisomers. Where a compound of formula I contains an alkenyl or alkenylene group, geometric cisltrans (or Z/E) isomers are possible. Where structural isomers are interconvertible via a low energy barrier, tautomeric isomerism ('tautomerism') can occur. This can take the form of proton tautomerism in compounds of formula I containing, for example, an imino, keto, or oxime group, or so-called valence tautomerism in compounds which contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism.
Included within the scope of the present invention are all stereoisomers, geometric isomers and tautomeric forms of the compounds of formula I, including compounds exhibiting more than one type of isomerism, and mixtures of one or more thereof. Also included are acid addition or base salts wherein the counterion is optically active, for example, <i-lactate or /-lysine, or racemic, for example, ^/-tartrate or t#-arginine.
Cisltrans isomers may be separated by conventional techniques well known to those skilled in the art, for example, chromatography and fractional crystallisation. Conventional techniques for the preparation/isolation of individual enantiomers include chiral synthesis from a suitable optically pure precursor or resolution of the racemate (or the racemate of a salt or derivative) using, for example, chiral high pressure liquid chromatography (HPLC). Alternatively, the racemate (or a racemic precursor) may be reacted with a suitable optically active compound, for example, an alcohol, or, in the case where the compound of formula I contains an acidic or basic moiety, a base or acid such as 1-phenylethylamine or tartaric acid. The resulting diastereomeric mixture may be separated by chromatography and/or fractional crystallization and one or both of the diastereoisomers converted to the corresponding pure enantiomer(s) by means well known to a skilled person.
Chiral compounds of the invention (and chiral precursors thereof) may be obtained in enantiomerically-enriched form using chromatography, typically HPLC, on an asymmetric resin with a mobile phase consisting of a hydrocarbon, typically heptane or hexane, containing from 0 to 50% by volume of isopropanol, typically from 2% to 20%, and from 0 to 5% by volume of an alkylamine, typically 0.1 % diethylamine. Concentration of the eluate affords the enriched mixture. Stereoisomeric conglomerates may be separated by conventional techniques known to those skilled in the art - see, for example, Stereochemistry of Organic Compounds by E. L. Eliel and S. H. Wilen (Wiley, New York, 1994). The present invention includes all pharmaceutically acceptable isotopically-labelled compounds of formula I wherein one or more atoms are replaced by atoms having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number which predominates in nature. Examples of isotopes suitable for inclusion in the compounds of the invention include isotopes of hydrogen, such as 2H and 3H, carbon, such as UC, ,3C and 14C, chlorine, such as 36C1, fluorine, such as 18F, iodine, such as 123I and 1251, nitrogen, such as 13N and 15N, oxygen, such as , 50, l 70 and l 80, phosphorus, such as 32P, and sulphur, such as 3 S. Certain isotopically-labelled compounds of formula I, for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies. The radioactive isotopes tritium, i.e. 3H, and carbon-14, i.e. 14C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection. Substitution with heavier isotopes such as deuterium, i.e. 2H, may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances. Substitution with positron emitting isotopes, such as nC, 18F, 150 and l3N, can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
Isotopically- labeled compounds of formula I can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying Examples and Preparations using an appropriate isotopically- labeled reagent in place of the non- labeled reagent previously employed. Pharmaceutically acceptable solvates in accordance with the invention include those wherein the solvent of crystallization may be isotopically substituted, e.g. D2O, de-acetone, d6-DMSO. All of the compounds of the formula (I) can be prepared by the procedures described in the general methods presented below or by the specific methods described in the Examples section and the Preparations section, or by routine modifications thereof. The present invention also encompasses any one or more of these processes for preparing the compounds of formula (I), in addition to any novel intermediates used therein.
Compounds of formula 1 may be prepared by coupling a compound of formula II,
Figure imgf000014_0001
wherein R , W, X, Y, Z, m and n are as defined above,
with a compound of formula III,
Figure imgf000014_0002
wherein A, V and R are as defined above, and L is a leaving group. When A is S02, suitable leaving groups are halogen, such as CI. When A is C=0, suitable leaving groups include OH, and peptide-coupling type procedures may be used, for example using HATU (2-(7-Aza-lH-benzotriazole-l-yl)-l,l,3,3-tetramethyluronium hexafluorophosphate) and DIPEA (diisopropyl ethylamine) in THF at room temperature.
Compounds of formula II may be prepared by a zincate coupling of compounds of formula IV,
Figure imgf000014_0003
wherein X, Y, Z, m and n are as defined above, Hal is a halogen (such as Br or CI) and Pg is a protecting group, with a compound of formula V,
R'-W-ZnBr (V)
wherein R1 and W are as defined above, followed by removal of Pg.
Preferably, the zincate coupling is carried out in the presence of a palladium(O) catalyst, such as Pd2(dba)3 [tris(dibenzylideneacetone)dipalladium(0)] in the presence of P(o-tolyl)3, in THF, at around 40-50°C. Suitable Pg groups include benzyl, which may be removed by hydrogenation in methanol containing aqueous HCI in the presence of Pd/C at a temperature in the range 20°C to the reflux temperature of the solvent, and a pressure of 1-100 atm (lxlO5 - IxlO7 N/m2) hydrogen.
When X=Y-Z represents N=CH-N, compounds of formula IV may be prepared from compounds of formula VI,
Figure imgf000015_0001
wherein m, n and Pg are as defined above,
by the action of PO(Hal)3. Preferably, the reaction is carried out in dichloromethane at room temperature.
Compounds of formulae III, IV and V are readily available or are available using known techniques. Examples of known compounds of formula IV are:
Figure imgf000015_0002
Figure imgf000016_0001
It will be apparent to those skilled in the art that sensitive functional groups may need to be protected and de-protected during synthesis of a compound of the invention. This may be achieved by conventional techniques, for example as described in 'Protective Groups in Organic Synthesis' by Theorora Greene and Peter Wuts (third edition, 1999, John Wiley and Sons). The invention further provides a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, for use as a pharmaceutical.
The invention further provides a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, for use in the treatment of a disorder selected from: pain, acute pain, chronic pain, neuropathic pain, inflammatory pain, visceral pain, nociceptive pain including post-surgical pain, and mixed pain types involving the viscera, gastrointestinal tract, cranial structures, musculoskeletal system, spine, urogenital system, cardiovascular system and CNS, including cancer pain, back and orofacial pain.
The invention further provides a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, for use in the treatment of a disorder selected from: cancer (including ovarian cancer, breast cancer and prostate cancer), atherosclerosis, thrombosis, psoriasis, multiple sclerosis, neurodegenerative disorders, irritable bowel syndrome, osteoarthritis, rheumatoid arthritis, neuropathological disorders, functional bowel disorders, inflammatory bowel diseases, pain associated with , dysmenorrhea, pelvic pain, cystitis, pancreatitis, migraine, cluster and tension headaches, diabetic neuropathy, peripheral neuropathic pain, sciatica, fibromyalgia, causalgia, and conditions of lower urinary tract dysfunction.
The invention further provides a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, for use in the treatment of fibrotic diseases. Such conditions include pulmonary fibrosis, Cirrhosis, endomyocardial fibrosis, mediastinal fibrosis, myelofibrosis, retroperitoneal fibrosis, nephrogenic systemic fibrosis, Crohn's disease, Keloid, old myocardial infarction, scleroderma / systemic sclerosis, atherofibrosis and adhesive capsulitis,
The invention further provides a pharmaceutical formulation comprising a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable adjuvant, diluent or carrier. The pharmaceutical formulation may further comprise one or more additional active agents for the treatment of a disorder mentioned above. The invention further provides a pharmaceutical kit comprising a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, and one or more additional active agents, as a combined preparation for separate, simultaneous or sequential administration in the treatment of a disorder mentioned above.
The invention further provides a method of treatment of a disorder mentioned above in a mammal (especially a human), comprising administration of a therapeutically effective amount of a compound of formula I, as defined above, or a pharmaceutically acceptable salt or solvate thereof, to a mammal in need of such treatment.
Compounds of the invention intended for pharmaceutical use may be administered as crystalline or amorphous products. They may be obtained, for example, as solid plugs, powders, or films by methods such as precipitation, crystallization, freeze drying, spray drying, or evaporative drying. Microwave or radio frequency drying may be used for this purpose.
They may be administered alone or in combination with one or more other compounds of the invention or in combination with one or more other drugs (or as any combination thereof). Generally, they will be administered as a formulation in association with one or more pharmaceutically acceptable excipients. The term 'excipient' is used herein to describe any ingredient other than the compound(s) of the invention. The choice of excipient will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
Pharmaceutical compositions suitable for the delivery of compounds of the present invention and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company, 1995).
ORAL ADMINISTRATION The compounds of the invention may be administered orally. Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, or buccal or sublingual administration may be employed by which the compound enters the blood stream directly from the mouth.
Formulations suitable for oral administration include solid formulations such as tablets, capsules containing particulates, liquids, or powders, lozenges (including
liquid- filled), chews, multi- and nano-particulates, gels, solid solution, liposome, films, ovules, sprays and liquid formulations.
Liquid formulations include suspensions, solutions, syrups and elixirs. Such formulations may be employed as fillers in soft or hard capsules and typically comprise a carrier, for example, water, ethanol, polyethylene glycol, propylene glycol, methylcellulose, or a suitable oil, and one or more emulsifying agents and/or suspending agents. Liquid formulations may also be prepared by the reconstitution of a solid, for example, from a sachet.
The compounds of the invention may also be used in fast-dissolving, fast-disintegrating dosage forms such as those described in Expert Opinion in Therapeutic Patents, Π. (6), 981-986, by Liang and Chen (2001).
For tablet dosage forms, depending on dose, the drug may make up from 1 weight % to 80 weight % of the dosage form, more typically from 5 weight % to 60 weight % of the dosage form. In addition to the drug, tablets generally contain a disintegrant. Examples of disintegrants include sodium starch glycolate, sodium carboxymethyl cellulose, calcium carboxymethyl cellulose, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, methyl cellulose, microcrystalline cellulose, lower alkyl-substituted hydroxypropyl cellulose, starch, pregelatinised starch and sodium alginate. Generally, the disintegrant will comprise from 1 weight % to 25 weight %, preferably from 5 weight % to 20 weight % of the dosage form. Binders are generally used to impart cohesive qualities to a tablet formulation. Suitable binders include microcrystalline cellulose, gelatin, sugars, polyethylene glycol, natural and synthetic gums, polyvinylpyrrolidone, pregelatinised starch, hydroxypropyl cellulose and hydroxypropyl methylcellulose. Tablets may also contain diluents, such as lactose (monohydrate, spray-dried monohydrate, anhydrous and the like), mannitol, xylitol, dextrose, sucrose, sorbitol, microcrystalline cellulose, starch and dibasic calcium phosphate dihydrate.
Tablets may also optionally comprise surface active agents, such as sodium lauryl sulfate and polysorbate 80, and glidants such as silicon dioxide and talc. When present, surface active agents may comprise from 0.2 weight % to 5 weight % of the tablet, and glidants may comprise from 0.2 weight % to 1 weight % of the tablet.
Tablets also generally contain lubricants such as magnesium stearate, calcium stearate, zinc stearate, sodium stearyl fumarate, and mixtures of magnesium stearate with sodium lauryl sulphate. Lubricants generally comprise from 0.25 weight % to 10 weight %, preferably from 0.5 weight % to 3 weight % of the tablet.
Other possible ingredients include anti-oxidants, colourants, flavouring agents, preservatives and taste-masking agents.
Exemplary tablets contain up to about 80% drug, from about 10 weight % to about 90 weight % binder, from about 0 weight % to about 85 weight % diluent, from about 2 weight % to about 10 weight % disintegrant, and from about 0.25 weight % to about 10 weight % lubricant.
Tablet blends may be compressed directly or by roller to form tablets. Tablet blends or portions of blends may alternatively be wet-, dry-, or melt-granulated, melt congealed, or extruded before tabletting. The final formulation may comprise one or more layers and may be coated or uncoated; it may even be encapsulated. The formulation of tablets is discussed in Pharmaceutical Dosage Forms: Tablets. Vol. 1, by H. Lieberman and L. Lachman (Marcel Dekker, New York, 1980).
Consumable oral films for human or veterinary use are typically pliable water-soluble or water-swellable thin film dosage forms which may be rapidly dissolving or mucoadhesive and typically comprise a compound of formula I, a film-forming polymer, a binder, a solvent, a humectant, a plasticiser, a stabiliser or emulsifier, a viscosity-modifying agent and a solvent. Some components of the formulation may perform more than one function.
The compound of the invention may be water-soluble or insoluble. A water-soluble compound typically comprises from 1 weight % to 80 weight %, more typically from 20 weight % to 50 weight %, of the solutes. Less soluble compounds may comprise a greater proportion of the composition, typically up to 88 weight % of the solutes. Alternatively, the compound of the invention may be in the form of multiparticulate beads.
The film-forming polymer may be selected from natural polysaccharides, proteins, or synthetic hydrocolloids and is typically present in the range 0.01 to 99 weight %, more typically in the range 30 to 80 weight %.
Other possible ingredients include anti-oxidants, colorants, flavourings and flavour enhancers, preservatives, salivary stimulating agents, cooling agents, co-solvents (including oils), emollients, bulking agents, anti-foaming agents, surfactants and taste-masking agents.
Films in accordance with the invention are typically prepared by evaporative drying of thin aqueous films coated onto a peelable backing support or paper. This may be done in a drying oven or tunnel, typically a combined coater dryer, or by freeze-drying or vacuuming. Solid formulations for oral administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
Suitable modified release formulations for the purposes of the invention are described in US Patent No. 6,106,864. Details of other suitable release technologies such as high energy dispersions and osmotic and coated particles are to be found in Pharmaceutical Technology On-line. 25(2), 1-14, by Verma et al (2001). The use of chewing gum to achieve controlled release is described in WO 00/35298.
PARENTERAL ADMINISTRATION
The compounds of the invention may also be administered directly into the blood stream, into muscle, or into an internal organ. Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular and subcutaneous. Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
Parenteral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (preferably to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water.
The preparation of parenteral formulations under sterile conditions, for example, by lyophilisation, may readily be accomplished using standard pharmaceutical techniques well known to those skilled in the art. The solubility of compounds of the invention used in the preparation of parenteral solutions may be increased by the use of appropriate formulation techniques, such as the incorporation of solubility-enhancing agents.
Formulations for parenteral administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release. Thus compounds of the invention may be formulated as a solid, semi-solid, or thixotropic liquid for administration as an implanted depot providing modified release of the active compound. Examples of such formulations include drug-coated stents and poly(i#-lactic-coglycolic)acid (PGLA) microspheres.
TOPICAL ADMINISTRATION
The compounds of the invention may also be administered topically to the skin or mucosa, that is, dermally or transdermal ly. Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used. Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol. Penetration enhancers may be incorporated - see, for example, J Pharm Sci, 88 (10), 955-958, by Finnin and Morgan (October 1999).
Other means of topical administration include delivery by electroporation, iontophoresis, phonophoresis, sonophoresis and microneedle or needle-free (e.g. Powderject™, Bioject™, etc.) injection.
Formulations for topical administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release. INHALED TNTRANASAL ADMINISTRATION
The compounds of the invention can also be administered intranasally or by inhalation, typically in the form of a dry powder (either alone, as a mixture, for example, in a dry blend with lactose, or as a mixed component particle, for example, mixed with phospholipids, such as phosphatidylcholine) from a dry powder inhaler or as an aerosol spray from a pressurised container, pump, spray, atomiser (preferably an atomiser using electrohydrodynamics to produce a fine mist), or nebuliser, with or without the use of a suitable propellant, such as 1,1 ,1,2-tetrafluoroethane or 1 , 1, 1,2,3,3,3-heptafluoropropane. For intranasal use, the powder may comprise a bioadhesive agent, for example, chitosan or cyclodextrin.
The pressurised container, pump, spray, atomizer, or nebuliser contains a solution or suspension of the compound(s) of the invention comprising, for example, ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilising, or extending release of the active, a propellant(s) as solvent and an optional surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.
Prior to use in a dry powder or suspension formulation, the drug product is micronised to a size suitable for delivery by inhalation (typically less than 5 microns). This may be achieved by any appropriate comminuting method, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenisation, or spray drying.
Capsules (made, for example, from gelatin or hydroxypropylmethylcellulose), blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the compound of the invention, a suitable powder base such as lactose or starch and a performance modifier such as /-leucine, mannitol, or magnesium stearate. The lactose may be anhydrous or in the form of the monohydrate, preferably the latter. Other suitable excipients include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose and trehalose. A suitable solution formulation for use in an atomiser using electrohydrodynamics to produce a fine mist may contain from ^g to 20mg of the compound of the invention per actuation and the actuation volume may vary from Ι μΐ to ΙΟΟμΙ. A typical formulation may comprise a compound of formula I, propylene glycol, sterile water, ethanol and sodium chloride. Alternative solvents which may be used instead of propylene glycol include glycerol and polyethylene glycol.
Suitable flavours, such as menthol and Ievomenthol, or sweeteners, such as saccharin or saccharin sodium, may be added to those formulations of the invention intended for inhaled/intranasal administration.
Formulations for inhaled/intranasal administration may be formulated to be immediate and/or modified release using, for example, PGLA. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
In the case of dry powder inhalers and aerosols, the dosage unit is determined by means of a valve which delivers a metered amount. Units in accordance with the invention are typically arranged to administer a metered dose or "puff containing from 1 to 10,000 μg of the compound of the invention. The overall daily dose will typically be in the range ^g to 10 mg which may be administered in a single dose or, more usually, as divided doses throughout the day.
RECTAL/TNTRA VAGINAL ADMINISTRATION
The compounds of the invention may be administered rectally or vaginally, for example, in the form of a suppository, pessary, or enema. Cocoa butter is a traditional suppository base, but various alternatives may be used as appropriate. Formulations for rectal/vaginal administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted and programmed release.
OCULAR/AURAL ADMINISTRATION
The compounds of the invention may also be administered directly to the eye or ear, typically in the form of drops of a micronised suspension or solution in isotonic, pH-adjusted, sterile saline. Other formulations suitable for ocular and aural administration include ointments, biodegradable (e.g. absorbable gel sponges, collagen) and non-biodegradable (e.g. silicone) implants, wafers, lenses and particulate or vesicular systems, such as niosomes or liposomes. A polymer such as crossed-linked polyacrylic acid, polyvinylalcohol, hyaluronic acid, a cellulosic polymer, for example, hydroxypropylmethylcellulose, hydroxyethylcellulose, or methyl cellulose, or a heteropoly saccharide polymer, for example, gelan gum, may be incorporated together with a preservative, such as benzalkonium chloride. Such formulations may also be delivered by iontophoresis.
Formulations for ocular/aural administration may be formulated to be immediate and/or modified release. Modified release formulations include delayed-, sustained-, pulsed-, controlled-, targeted, or programmed release.
OTHER TECHNOLOGIES
The compounds of the invention may be combined with soluble macromolecular entities, such as cyclodextrin and suitable derivatives thereof or polyethylene gly col-containing polymers, in order to improve their solubility, dissolution rate, taste-masking, bioavailability and/or stability for use in any of the aforementioned modes of administration. Drug-cyclodextrin complexes, for example, are found to be generally useful for most dosage forms and administration routes. Both inclusion and non- inclusion complexes may be used. As an alternative to direct complexation with the drug, the cyclodextrin may be used as an auxiliary additive, i.e. as a carrier, diluent, or solubiliser. Most commonly used for these purposes are alpha-, beta- and gamma-cyclodextrins, examples of which may be found in International Patent Applications Nos. WO 91/1 1172, WO 94/02518 and WO 98/55148.
KIT-OF-PARTS
Inasmuch as it may desirable to administer a combination of active compounds, for example, for the purpose of treating a particular disease or condition, it is within the scope of the present invention that two or more pharmaceutical compositions, at least one of which contains a compound in accordance with the invention, may conveniently be combined in the form of a kit suitable for coadministration of the compositions.
Thus the kit of the invention comprises two or more separate pharmaceutical compositions, at least one of which contains a compound of formula I in accordance with the invention, and means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet. An example of such a kit is the familiar blister pack used for the packaging of tablets, capsules and the like.
The kit of the invention is particularly suitable for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another. To assist compliance, the kit typically comprises directions for administration and may be provided with a so-called memory aid.
Examples of additional active agents which may be part of a formulation or pharmaceutical kit according to the present invention, particularly for the treatment of pain or pain-related disorders, include: a Navl .7 channel modulator, such as a compound disclosed in WO 2009/012242; a Navl .3 modulator (e.g. as disclosed in WO2008/1 18758); or a Navl.8 modulator (e.g. as disclosed in WO 2008/135826, more particularly N-[6-Amino-5-(2-chIoro-5-methoxyphenyl)pyridin-2-yl]-l-methyl-lH-pyrazole-5 -carboxamide);
an inhibitor of nerve growth factor signaling, such as: an agent that binds to NGF and inhibits NGF biological activity and/or downstream pathway(s) mediated by NGF signaling (e.g. tanezumab), a TrkA antagonist or a p75 antagonist;
a compound which increases the levels of endocannabinoid, such as a compound with fatty acid amid hydrolase inhibitory (FAAH) activity, in particular those disclosed in WO 2008/047229 (e.g.
N-pyridazin-3-yl-4-(3-{[5-(trifluoromethyI)pyridine-2-yl]oxy}benzylidene)piperid ene- 1 -carboxamide);
an opioid analgesic, e.g. morphine, heroin, hydromorphone, oxymorphone, levorphanol, levallorphan, methadone, meperidine, fentanyl, cocaine, codeine, dihydrocodeine, oxycodone, hydrocodone, propoxyphene, nalmefene, nalorphine, naloxone, naltrexone, buprenorphine, butorphanol, nalbuphine or pentazocine; a nonsteroidal antiinflammatory drug (NSAID), e.g. aspirin, diclofenac, diflusinal, etodolac, fenbufen, fenoprofen, flufenisal, flurbiprofen, ibuprofen, indomethacin, ketoprofen, ketorolac, meclofenamic acid, mefenamic acid, meloxicam, nabumetone, naproxen, nimesulide, nitroflurbiprofen, olsalazine, oxaprozin, phenylbutazone, piroxicam, sulfasalazine, sulindac, tolmetin or zomepirac;
a barbiturate sedative, e.g. amobarbital, aprobarbital, butabarbital, butabital, mephobarbital, metharbital, methohexital, pentobarbital, phenobartital, secobarbital, talbutal, theamylal or thiopental;
a benzodiazepine having a sedative action, e.g. chlordiazepoxide, clorazepate, diazepam, flurazepam, lorazepam, oxazepam, temazepam or triazolam;
an Hi antagonist having a sedative action, e.g. diphenhydramine, pyrilamine, promethazine, chlorpheniramine or chlorcyclizine; a sedative such as glutethimide, meprobamate, methaqualone or dichloralphenazone;
a skeletal muscle relaxant, e.g. baclofen, carisoprodol, chlorzoxazone, cyclobenzaprine, methocarbamol or orphrenadine;
an NMDA receptor antagonist, e.g. dextromethorphan ((+)-3-hydroxy-N-methylmorphinan) or its metabolite dextrorphan ((+)-3-hydroxy-N-methylmorphinan), ketamine, memantine, pyrroloquinoline quinine, cis-4-(phosphonomethyl)-2-piperidinecarboxylic acid, budipine, EN-3231 (MorphiDex®, a combination formulation of morphine and dextromethorphan), topiramate, neramexane or perzinfotel including an NR2B antagonist, e.g. ifenprodil, traxoprodil or
(-)-(R)-6-{2-[4-(3-fluorophenyl)-4-hydroxy-l-piperidinyl]-l-hydroxyethyl-3,4-di hydro-2(lH)-quinolinone;
an alpha-adrenergic, e.g. doxazosin, tamsulosin, clonidine, guanfacine, dexmetatomidine, modafinil, or
4- amino-6,7-dimethoxy-2-(5-methane-sulfonamido-l,2,3,4-tetrahydroisoquinol-2- yl)-5-(2-pyridyl) quinazoline;
a tricyclic antidepressant, e.g. desipramine, imipramine, amitriptyline or nortriptyline;
an anticonvulsant, e.g. carbamazepine, lamotrigine, topiratmate or valproate; a tachykinin (NK) antagonist, particularly an NK-3, NK-2 or NK- antagonist, e.g. (aR,9R)-7-[3,5-bis(trifluoromethyl)benzyl]-8,9,10,l -tetrahydro-9-methyl-5-(4-m ethylphenyl)-7H-[l,4]diazocino[2,l-g][l,7]-naphthyridine-6-13-dione (TAK-637),
5- [[(2R,3S)-2-[(lR)-l-[3,5-bis(trifluoromethyl)phenyl]ethoxy-3-(4-fluorophenyl)- 4-moφholinyl]-methyl]- ,2-dihydro-3H-l,2,4-triazol-3-one (MK-869), aprepitant, lanepitant, dapitant or 3-[[2-methoxy-5-(trifluoromethoxy)phenyl]-methylamino]-2-phenylpiperidine (2S,3S);
a muscarinic antagonist, e.g oxybutynin, tolterodine, propiverine, tropsium chloride, darifenacin, solifenacin, temiverine and ipratropium; • a COX-2 selective inhibitor, e.g. celecoxib, rofecoxib, parecoxib, valdecoxib, deracoxib, etoricoxib, or lumiracoxib;
• a coal-tar analgesic, in particular paracetamol;
• a neuroleptic such as droperidol, chlorpromazine, haloperidol, perphenazine, thioridazine, mesoridazine, trifluoperazine, fluphenazine, clozapine, olanzapine, risperidone, ziprasidone, quetiapine, sertindole, aripiprazole, sonepiprazole, blonanserin, iloperidone, perospirone, raclopride, zotepine, bifeprunox, asenapine, lurasidone, amisulpride, balaperidone, palindore, eplivanserin, osanetant, rimonabant, meclinertant, Miraxion® or sarizotan;
· a vanilloid receptor agonist (e.g. resinferatoxin) or antagonist (e.g. capsazepine);
• a beta-adrenergic such as propranolol;
• a local anaesthetic such as mexiletine;
• a corticosteroid such as dexamethasone;
• a 5-HT receptor agonist or antagonist, particularly a 5-HTIB/ID agonist such as eletriptan, sumatriptan, naratriptan, zolmitriptan or rizatriptan;
• a 5-HT2A receptor antagonist such as R(+)-alpha-(2,3-dimethoxy-phenyl)-l -[2-(4-fluorophenylethyl)]-4-piperidinemeth anol (MDL- 100907);
• a 5-HT3 antagonist, such as ondansetron
· a cholinergic (nicotinic) analgesic, such as ispronicline (TC-1734), (E)-N-methyl-4-(3-pyridinyl)-3-buten-l-amine (RJR-2403), (R)-5-(2-azetidinylmethoxy)-2-chloropyridine (ABT-594) or nicotine;
• Tramadol®;
• a PDEV inhibitor, such as 5-[2-ethoxy-5-(4-methyl-l-piperazinyl-sulphonyl)phenyl]-l-methyl-3-n-propyl-l ,
6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one (sildenafil), (6R, 12aR)-2,3,6,7, 12,12a-hexahydro-2-methyl-6-(3,4-methylenedioxyphenyl)-pyr azino[2',l':6,l]-pyrido[3,4-b]indole-l,4-dione (IC-351 or tadalafil), 2-[2-ethoxy-5-(4-ethyl-piperazin- 1 -yl- 1 -sulphonyl)-phenyl]-5-methy l-7-propyl-3 H-imidazo[5,l-fJ[l,2,4]triazin-4-one (vardenafil), 5-(5-acetyl-2-butoxy-3-pyridinyl)-3-ethyl-2-(l-ethyl-3-azetidinyl)-2,6-dihydro-7H -pyrazolo[4,3-iflpyrimidin-7-one,
5-(5-acetyl-2-propoxy-3-pyridinyl)-3-ethyl-2-(l-isopropyl-3-azetidinyl)-2,6-dihyd ro-7H-pyrazolo[4,3-c/]pyrimidin-7-one,
5-[2-ethoxy-5-(4-ethylpiperazin- l-ylsulphonyl)pyridin-3-yl]-3-ethyl-2-[2-methox yethyl]-2,6-dihydro-7H-pyrazolo[4,3-d]pyrimidin-7-one,
4-[(3-chloro-4-methoxybenzyl)amino]-2-[(2S)-2-(hydroxyrnethyl)pyrrolidin- l-yl] -N-(pyrimidin-2-ylmethyl)pyrimidine-5-carboxamide,
3-(l-methyl-7-oxo-3-propyl-6,7-dihydro- lH-pyrazolo[4,3-d]pyrimidin-5-yl)-N-[2 -(l-methylpyrrolidin-2-yl)ethyl]-4-propoxybenzenesulfonamide;
an alpha-2-delta ligand such as gabapentin, pregabalin, 3-methylgabapentin, ( l a,3a,5a)(3-amino-methyl-bicyclo[3.2.0]hept-3-yl)-acetic acid, (3S,5R)-3-aminomethyl-5-methyl-heptanoic acid, (3S,5R)-3-amino-5-methyl-heptanoic acid, (3S,5R)-3-amino-5-methyl-octanoic acid, (2S,4S)-4-(3-chlorophenoxy)proline, (2S,4S)-4-(3-fluorobenzyl)-proline, [(lR,5R,6S)-6-(aminomethyl)bicyclo[3.2.0]hept-6-yl]acetic acid, 3-(l-ammomethyl-cyclohexylmethyl)-4H-[l,2,4]oxadiazol-5-one,
C-[l -(lH-tetrazol-5-ylmethyl)-cycloheptyl]-methylamine,
(3S,4S)-(l-aminomethyl-3,4-dimethyl-cyclopentyl)-acetic acid, (3 S,5R)-3-aminomethyl-5-methyl-octanoic acid, (3S,5R)-3-amino-5-methyl-nonanoic acid, (3S,5R)-3-amino-5-methyl-octanoic acid, (3R,4R,5R)-3-amino-4,5-dimethyl-heptanoic acid and
(3R,4R,5R)-3-amino-4,5-dimethyl-octanoic acid;
metabotropic glutamate subtype 1 receptor (mGluRl) antagonist;
a serotonin reuptake inhibitor such as sertraline, sertraline metabolite demethylsertraline, fluoxetine, norfluoxetine (fluoxetine desmethyl metabolite), fluvoxamine, paroxetine, citalopram, citalopram metabolite desmethylcitalopram, escitalopram, d,l- fenfluramine, femoxetine, ifoxetine, cyanodothiepin, litoxetine, dapoxetine, nefazodone, cericlamine and trazodone;
a noradrenaline (norepinephrine) reuptake inhibitor, such as maprotiline, lofepramine, mirtazepine, oxaprotiline, fezolamine, tomoxetine, mianserin, buproprion, buproprion metabolite hydroxybuproprion, nomifensine and viloxazine (Vivalan®), especially a selective noradrenaline reuptake inhibitor such as reboxetine, in particular (S,S)-reboxetine;
• a dual serotonin-noradrenaline reuptake inhibitor, such as venlafaxine, venlafaxine metabolite O-desmethylvenlafaxine, clomipramine, clomipramine metabolite desrnethylclomipramine, duloxetine, milnacipran and imipramine;
• an inducible nitric oxide synthase (iNOS) inhibitor such as S-[2-[(l -iminoethyl)amino]ethyl]-L-homocysteine,
S-[2-[( l-iminoethyl)-amino]ethyl]-4,4-dioxo-L-cysteine,
S-[2-[(] -iminoethyl)amino]ethyl]-2-methyl-L-cysteine,
(2S,5Z)-2-amino-2-methyl-7-[(l -iminoethyl)amino]-5-heptenoic acid,
2-[[(IR,3S)-3-amino-4- hydroxy- l -(5-thiazolyl)-butyl]thio]-5-chloro-3-pyridinecarbonitrile;
2-[[(lR,3S)-3-amino-4-hydroxy-l-(5-thiazolyl)butyl]thio]-4-chlorobenzonitrile, (2S,4R)-2-amino-4-[[2-chloro-5-(trifluoromethyl)phenyI]thio]-5-thiazolebutanol, 2- [[( 1 R,3 S)-3-amino-4-hydroxy- 1 -(5-thiazo ly 1) buty l]thio]-6-(trifluoromethy l)-3 pyridinecarbonitrile, 2-[[(l R,3S)-3- amino-4-hydroxy- 1
-(5-thiazo lyl)butyl]thio]-5-chlorobenzonitrile,
N-[4-[2-(3-chlorobenzylamino)ethyl]phenyl]thiophene-2-carboxamidine, or guanidinoethyldisulfide;
· an acetylcholinesterase inhibitor such as donepezil;
• a prostaglandin E2 subtype 4 (EP4) antagonist such as N-[({2-[4-(2-ethyl-4,6-dimethyl-lH-imidazo[4,5-c]pyridin- l-yl)phenyl]ethyl}ami no)-carbonyl]-4-methylbenzenesulfonamide or 4-[(lS)-l -({[5-chloro-2-(3-fluorophenoxy)pyridin-3-yl]carbonyl}amino)ethyl]ben zoic acid;
• a microsomal prostaglandin E synthase type 1 (mPGES-1) inhibitor;
• a leukotriene B4 antagonist, such as l -(3-biphenyl-4-ylmethyl-4-hydroxy-chroman-7-yl)-cyclopentanecarboxylic acid (CP- 105696), 5-[2-(2-Carboxyethyl)-3-[6-(4-methoxyphenyl)-5E- hexenyl]oxyphenoxy]-valeric acid (ΟΝΟ-4057) or DPC- 1870; and • a 5-lipoxygenase inhibitor, such as zileuton, 6-[(3-fluoro-5-[4-methoxy-3,4,5,6-tetrahydro-2H-pyran-4-yl])phenoxy-methyl]-l- methyl-2-quinolone (ZD-2138), or 2,3,5-trimethyl-6-(3-pyridylmethyl),l,4-benzoquinone (CV-6504).
Examples of additional active agents which may be part of a formulation or pharmaceutical kit according to the present invention, particularly for the treatment of cancer, include:
• an anti-tumour platinum-based compound such as cisplatin or carboplatin;
• an anti-estrogen or selective estrogen receptor modulator such as tamoxifen, afimoxifene, arzoxifene, bazedoxifene, lasofoxifene or nafoxidene; and
• a tyrosine kinase inhibitor such as afatinib, imatinib, gefitinib, sorafenib, sunitinib, vandetanib, crizotinib or lapatinib.
DOSAGE
For administration to human patients, the total daily dose of the compounds of the invention is typically in the range 0.5 mg to 3000 mg depending, of course, on the mode of administration. For example, oral administration may require a total daily dose of from 3 mg to 3000 mg, while an intravenous dose may only require from 0.5 mg to 500 mg. The total daily dose may be administered in single or divided doses and may, at the physician's discretion, fall outside of the typical range given herein.
These dosages are based on an average human subject having a weight of about 60kg to 70kg. The physician will readily be able to determine doses for subjects whose weight falls outside this range, such as infants and the elderly.
For the avoidance of doubt, references herein to "treatment" include references to curative, palliative and prophylactic treatment. Biological Activity
The biological activity of the compounds of the invention may be measured using the assay described below.
FS-3 Assay
Autotaxin (ATX) converts Lysophosphatidyl choline (LPC) to Lysophosphatidic acid (LP A) via its Lysophosphodiesterase (lysoPLD) activity. FS-3 is an LPC analogue that is conjugated with both a fluorophore and a quencher. In its native state the quencher interferes with the fluorophore's fluorescence. Once autotaxin cleaves FS-3, the fluorophore becomes liberated from the quencher, resulting in increased fluorescence. Thus increase in fluorescence is a measure of ATX activity. Any compound that inhibits the activity of Autotaxin will show less fluorescence read in presence of FS-3, which could be measured by fluorimetric analysis. See US Patent 7,989,663.
Autotaxin Inhibitor Screening Kits are available from Echelon Biosciences, Logan, UT, USA [http://echelon-inc.comA accessed 6 October 2011]. Using the methods of Gierse et al ["A novel autotaxin inhibitor reduces lysophosphatidic acid levels in plasma and the site of inflammation", Journal of Pharmacology and Experimental Therapeutics, vol 334(1), 310-317 (2010), see in particular page 312 lines 4-14], the potency of the compounds of the invention as inhibitors of human autotaxin enzyme may be measured (as IC50 values).
The FS-3 assay to identify ATX inhibitors was preformed as follows: 3 μΐ of standard inhibitor (referred to as PF-8380 in Gierse et al above) and test compounds were added to an assay plate. To each assay well, containing test compounds or standard, 24 μΐ of human Autotaxin enzyme (2 nM) was added. The assay plate was then centrifuged at 1000 rpm for 1 minute and allowed to incubate at 37°C for 30 minutes. Following the incubation period each plate was read in a fluorescence plate reader (Spectra Max M5: excitation: 494nm and emission: 520nm) and IC50 values were derived from inhibition of FS-3 fluorescence (as described above).
The invention is illustrated by the following Examples. Experimental details for Example 2 are provided below. The other compounds in Table 1 below were prepared using analogous methods. Table 1
Example Structure IUPAC NAME
number
1 6- [(3 ,4-dichloropheny l)acetyl]-4-(4-fluor obenzyl)-5,6,7,8-tetrahydropyrido[4,3-d]p yrimidine
2 6-[3-(l,3-benzothiazol-2-yl)propanoyl]-4- (4-fluorobenzyl)-5,6,7,8-tetrahydropyrido [4,3-d]pyrimidine
3 6-(cyclopropylacetyl)-4-(4-fluorobenzyl)- 5 , 6, 7, 8-tetrahydropyrido [4, 3 -d] pyr imidin e
^^^^
4 6- [3 -(3-chloropheny l)propanoy 1] -4-(4-flu orobenzyl)-5,6,7,8-tetrahydropyrido[4,3-d
]pyrimidine
5 6-[(3,4-difluorophenyl)acetyl]-4-(4-fluoro benzyl)-5,6,7,8-tetrahydropyrido[4,3-d]py rimidine
6 6-[(3-chlorophenyl)acetyl]-4-(4-fluoroben zyl)-5,6,7,8-tetrahydropyrido[4,3-d]pyrim idine
7 4-(4-fluorobenzyl)-6-[3-(methylthio)prop anoyl]-5,6,7,8-tetrahydropyrido[4,3-d]pyr imidine
Figure imgf000036_0001
16 6-[(3,5-difluorophenyl)acetyl]-4-(4-fluoro benzyl)-5,6,7,8-tetrahydropyrido[4,3-d]py rimidine
17 4-(4-fluorobenzyl)-6-[(3-nitrophenyl)acet yl]-5,6,7,8-tetrahydropyrido[4,3-d]pyrimi dine
18 6-(2,3-dihydro-l-benzofuran-5-ylacetyl)- 4-(4-fluorobenzyl)-5,6,7,8-tetrahydropyri do[4,3-d]pyrimidine
19 2-[4-(4-fluorobenzyl)-7,8-dihydropyrido[
4,3-d]pyrimidin-6(5H)-yl]-N,N-dimethyl- 2-oxoethanesulfonamide
20 4-(4-fluorobenzyl)-6-[(3-phenoxyphenyl) acetyl]-5,6,7,8-tetrahydropyrido[4,3-d]py rimidine
21 N-{2-[4-(4-fluorobenzyl)-7,8-dihydropyri do[4,3-d]pyrimidin-6(5H)-yl]-2-oxoethyl
}-2-phenylacetamide
22 4-(4-fluorobenzyl)-6-[3-(4-fluorophenyl) propanoyl] -5 , 6, 7, 8-tetrahydropyr ido [4,3- d]pyrimidine
23 4-(4-fluorobenzyl)-6-[3-(3-fluorophenyl) propanoyl] -5 , 6, 7, 8-tetrahydropyr ido [4,3 - d]pyrimidine methyl
6-[4-(4-fluorobenzyl)-7,8-dihydropyrido[
4,3-d]pyrimidin-6(5H)-yl]-6-oxohexanoat e
^^^^
6-[(2,3-difluorophenyl)acetyl]-4-(4-fluoro benzyl)-5 , 6,7, 8-tetrahydropyr ido [4,3-d] py rimidine
Figure imgf000038_0001
4-(4-fluorobenzyl)-6-[(4-phenoxyphenyl) acetyl]-5,6,7,8-tetrahydropyrido[4,3-d]py rimidine
4-(4-fluorobenzy l)-6- { [3-(trifluoromethy 1 )phenyl]acetyl}-5,6,7,8-tetrahydropyrido[ 4,3-d]pyrimidine
4-(4-fluorobenzyl)-6-(2H-indazol-2-ylace tyl)-5,6,7,8-tetrahydropyrido[4,3-d]pyrim idine
4-(4-fluorobenzyl)-6-(3,5,5-trimethylhexa noyl)-5,6,7,8-tetrahydropyrido[4,3-d]pyri m idine
Figure imgf000038_0002
ethyl
7-[4-(4-fluorobenzyl)-7,8-dihydropyrido[
4,3-d]pyrimidin-6(5H)-yl]-7-oxoheptanoa te
4-(4-fluorobenzy l)-6- [(4-methoxy phenyl) acetyl]-5,6,7,8-tetrahydropyrido[4,3-d]py rimidine 32 4-(4-fluorobenzyl)-6-[(phenylsulfonyl)ac etyl]-5,6,7,8-tetrahydropyrido[4,3-d]pyri midine
33 6-(3-cyclopentylpropanoyl)-4-[4-(trifluor omethoxy)benzyl]-5,6,7,8-tetra ydropyri do[4,3-d]pyrimidine
34 6-[(benzyloxy)acetyl]-4-(4-fluorobenzyl)-
5,6,7,8-tetrahydropyrido[4,3-d]pyrimidin e
35 6-(biphenyl-4-yIacetyl)-4-(4-fluorobenzyl
)-5,6,7,8-tetrahydropyrido[4,3-d]pyrimidi ne
36 4-(4-fluorobenzy l)-6- { [4-(trifluoromethy 1
)pheny 1] acetyl } - 5 ,6, 7, 8-tetrahydropyr ido [ 4,3-d]pyrimidine
37 6- [(3 -methoxypheny l)acetyl]-4- [4-(trifluo romethoxy)benzyl]-5,6,7,8-tetrahydropyri do[4,3-d]pyrimidine
38 4-(4-fluorobenzyl)-6-[(3-methoxyphenyl) acetyl] -5 , 6, 7, 8-tetrahydropyr ido [4, 3 -d]py rimidine
39 6- { [3-(benzyloxy)phenyl]acetyl}-4-(4-flu orobenzyl)-5,6,7,8-tetrahydropyrido[4,3-d ]pyrimidine N-benzyl-N-{2-[4-(4-fluorobenzyl)-7,8-di hydropyrido[4,3-d]pyrimidin-6(5H)-yl]-2 -oxoethy 1 } tetrahydro-2H-pyran-4-am ine
4-(4-fluorobenzyl)-6-[3-(3-methoxypheny l)propanoyl]-5,6,7,8-tetrahydropyrido[4,3 -d]pyrimidine
4-(4-fluorobenzy l)-6-( 1 H-indazo 1- 1 -ylace tyl)-5,6,7,8-tetrahydropyrido[4,3-d]pyrim idine
ethyl
7-(4-benzyl-7,8-dihydropyrido[4,3-d]pyri midin-6(5H)-yI)-7-oxoheptanoate
4-(4-fluorobenzyl)-6-[(2-methoxyphenyl) acetyl]-5,6,7,8-tetrahydropyrido[4,3-d]py
'V ? rimidine
4-(4-fluorobenzyl)-6-(lH-indol-3-ylacetyl )-5,6,7,8-tetrahydropyrido[4,3-d]pyrimidi ne
N-(4-{2-[4-(4-fluorobenzyl)-7,8-dihydrop yrido[4,3-d]pyrimidin-6(5H)-yl]-2-oxoeth yl}phenyl)methanesulfonamide
4-(4-fluorobenzyl)-6-{[3-(methylsulfonyl )phenyl]acetyl}-5,6,7,8-tetrahydropyrido[ 4,3-d]pyrimidine
Figure imgf000040_0001
48 6-[(2,5-dimethoxyphenyl)acetyl]-4-(4-flu orobenzyl)-5,6,7,8-tetrahydropyrido[4,3-d
]pyrimidine
49 4-(4-fiuorobenzyl)-6-[(6-phenoxypyridin- 2-yl)carbonyl]-5,6,7,8-tetrahydropyrido[4 ,3-d]pyrimidine
50 2-{3-[4-(4-fluorobenzyl)-7,8-dihydropyri do[4,3-d]pyrimidin-6(5H)-yl]-3-oxopropy l}isoindolin-l-one
51 4-{[4-(4-fIuorobenzyl)-7,8-dihydropyrido
[4,3-d]pyrimidin-6(5H)-yl]carbonyl}-l-(p yridin-2-ylmethyl)pyrrolidin-2-one
52 6-[3,3-dimethyl-5-(4-methylpiperazin-l-y l)-5-oxopentanoyl]-4-(4-fluorobenzyl)-5,6 ,7,8-tetrahydropyrido[4,3-d]pyriinidine
Figure imgf000041_0001
53 4-(3-{2-[4-(4-fluorobenzyl)-7,8-dihydrop yrido[4,3-d]pyrimidin-6(5H)-yl]-2-oxoeth yl} -2-oxoimidazo lidin- 1 -yl)benzonitrile
Figure imgf000041_0002
All starting materials are available commercially, are described in the literature or are available using known techniques. All temperatures are in °C. Flash column chromatography was carried out using Merck silica gel 60 (9385). Thin layer chromatography (TLC) was carried out on Merck silica gel 60 plates (5729). "Rf" represents the distance travelled by a compound divided by the distance travelled by the solvent front on a TLC plate. Melting points were determined using a Gallenkamp MPD350 apparatus and are uncorrected. NMR was carried out using a Varian-Unity Inova 400 MHz NMR spectrometer or a Varian Mercury 400 MHz NMR spectrometer. Mass spectroscopy was carried out using a Finnigan Navigator single quadrupole electrospray mass spectrometer or a Finnigan aQa APCI mass spectrometer. The following abbreviations and definitions may be used below:
APCI atmospheric pressure chemical ionisation
CDCI3 deuterated chloroform
CD3OD deuterated methanol
DMSO deuterated dimethyl sulphoxide
δ chemical shift
d Doublet
DAD Diode array detector
DIPEA Diisopropyl ethylamine
ESCI electrospray chemical ionisation
EtOAc Ethyl acetate
h hours
HATU 2-(7-Aza-l H-benzotriazole-l -yl)- l , l ,3,3-tetramethyluronium
hexafluorophosphate
HPLC high pressure liquid chromatography
LRMS low resolution mass spectrum
LCMS Liquid chromatography mass spectrum
M molar
m multiplet
mg milligrams
MHz mega hertz
min minutes
mL millilitres
μΐ, microlitres
mmol milli moles
mol moles
MS mass spectrometry
NMR nuclear magnetic resonance q quartet
Rt retention time
s singlet
t triplet
TFA trifluoroacetic acid
THF tetrahydrofuran
SFC Supercritritcal fluid chromatography
Where compounds have been analysed by LCMS a Waters Acquity LCMS system has been used with:
Mass Spectrometer Model: Waters ZQ
ionization Mode: API-ES
Polarity: Positive
Detectors: UV - Acquity PDA; Varian ELSD (evaporative light scattering detector)
Chromatography system
A: 0.1 % formic acid in water
B: 0.1 % formic acid in acetonitrile
Column: Acquity CSH CI 8 50 x 2.1 mm with 1.7 micron particle size
Gradient: 95-5% A over 1.12 min, 0.43 min hold, 1 mL/min
UV: 210 nm - 225 nm PDA
Temperature: 50 °C
Example 2
6-[3-(l,3-benzothiazol-2-yl)propanoyl]-4-(4-fluorobenzyl)-5,6,7,8-tetrahydropyrido- [4,3-d] py rimidine
Figure imgf000043_0001
A solution of 3-(l,3-benzothiazol-2-yl)propanoic acid (682 mg, 3.29 mmol), HATU (1.65 g, 4.34 mmol) and DIPEA (1.6 ml, 9.87 mmol) in THF (10 ml) was stirred at room temperature for 10 min. Then the reaction mixture was cooled to 0°C, the title compound from Preparation 4 (800 mg, 3.29 mmol) was added and stirred at room temperature for 1 h. After completion of the reaction (monitored by TLC), the solvent was evaporated under reduced pressure. The crude product was diluted with EtOAc and washed with water and brine solution. The organic layer was dried over anhydrous Na2SC»4 and concentrated. The crude product was purified by Prep- TLC to get 680 mg of the title compound (48%). Rf: 0.3 (100% EtOAc) UV active.
'H NMR (DMSO-d6, 400 MHz) δ: 8.9 (s, 1 H), 8.0 (s, 1H), 7.9 (dd, 5H), 7.5 (m, 1H), 7.4 (m, IH), 7.3 (m, 2H), 7.1(m, 2H), 4.7 (d, 2H), 4.1 (d, 2H), 3.8 (m, 2H), 3.4 (d, 2H), 3.1 (t, lH), 3.0 (m, 2H), 2.8 (t, IH).
LCMS: 97.9 % (m/z= 433.0 [M+H]+). Additional characterizing data:
Example number NMR (300 MHz) LCMS
20 (DMSO) 5: 8.9 (s, IH) , 6.7-7.4 (m, m/z= 454.0 [M+H]+
14H) , 4.7 (s, 2H), 4.1 (d, 2H) , 3.9
(m, 2H), 3.7 (m, 2H) 2.7 (m, 2H) ,
2.6 (m, 2H) , 1.2 (s, 4H ) .
21 ( DMSO ) 6: 8.9 (s, IH) , 8.2 (m, IH) , m/z= 419.0 [M+H]+
7.3 (m, 7H), 7.1 (m, 2H) , 4.7 (s,
2H) , 4.1 (d, 4H), 3.7 (m, 2H) , 3.5
( s , 2 H ) 2 . 9 ( t , 3H ) , 1 . 3 ( s , 2H ) .
28 ( DMSO ) 6: 8.9 (s, IH) , 8.3 (s, IH) , m/z= 402.0 [M+H]+
7.7 (m, IH), 7.6 (m, IH) , 7.3 (m,
IH) 7.2 (m, 2H), 7.1 (m, 3H) , 5.6
(m, 2H), 4.8 (d, 2H) 4.1 (d, 2H) ,
3.9 (d, 2H), 3.0 (d, IH) , 2.9 (d,
IH ) .
40 (CDC13) δ: 9.0 (s, IH) , 7.3 (s, 2H), m/z= 474.9 [M+H]+
7.2 (s, 5H), 6.9 (m, 2H) , 4.9 (s,
IH) , 4.6 (s, IH) , 4.0 (d, 4H) , 3.7
(s, 3H), 3.5 (s, 3H), 3.3 (d, 2H) , 2.8 (s, 1H), 2.5 (s, 2H), 1.7 (s,
4H) .
Preparations
Preparation 1
6-Benz i-5,6,7,8-tetrah dropyrido[4,3-rf]pyrimidin-4 3H)-
Figure imgf000045_0001
Sodium methylate (43 g, 0.8 mol) was added to a mixture of imidoformamide acetate (39.85 g, 0.383 mol) and ethyl l-benzyl-4-oxopiperidine-3-carboxylate (110 g, 0.37 mol) in absolute ethanol (250 mL). The reaction mixture was refluxed for 3-4 h, during which time the course of the reaction was monitored by TLC (Merck UV-254; chloroform/methanol 4:1 ). The mixture was then filtered, and the filtrate was evaporated. Water (-400 mL) was added to the residue, and the obtained aqueous solution was acidified to pH 7-8. The product was then extracted with chloroform, and the extract was dried over MgS04 and evaporated to give the title compound in 55% (50.7 g) yield. The product was then recrystallized from ethylacetate.
Preparation 2
6-Benz l-4-bromo-5,6,7,8-tetrahydropyrido[4,3-rf]pyriniidine
Figure imgf000045_0002
POBr3 (8.92 g, 21 x 1.5mmol) was added to a slurry of the title compound from Preparation 1 (5 g, 21 mmol) in dichloroethane (100 mL). The mixture was stirred at room temperature for 1 h and then refluxed and stirred for a further 1 h, during which time the course of the reaction was monitored by TLC (Merck UV-254; chloroform/methanol 10:1; samples were treated with saturated Na2C(¾ solution). The reaction mixture was cooled to room temperature, treated with saturated Na2CC>3 solution, stirred for 10 min and treated with ultrasound for 10 min. The organic layer was separated, and the aqueous layer was extracted with chloroform. The organic layers were combined, washed with water, dried over MgS04 and evaporated to give the product in 79% (5 g) yield.
Preparation 3
-BenzyI-4-(4-fluorobenzyl)-5,6,7,8-tetrahydropyrido[4,3-<fIpyrimidine
Figure imgf000046_0001
A 1 -L three-neck flask equipped with an addition funnel and returning condenser was heated, vacuumed, then filled with argon. Zinc (73 g) was added, and the flask was heated, vacuumed and filled with argon once again. Then absolute THF (150 mL) and dibromoethane (9.6 mL) were added. The mixture was heated at 65-70 °C for 30 min, then trimethylsilyl chloride (2.9 mL) was added, and the mixture was stirred for another 30 min. A solution of l-(bromomethyl)-4-fluorobenzene (90 g) in THF (150 mL) was slowly added dropwise (45 min). The temperature was maintained at -40 °C. After 30 min of stirring, almost no starting material could be seen on the TLC. The title compound from Preparation 2 (98.5 g) was added, then d2(dba)3 [tris(dibenzylideneacetone)dipalladium(0)] (1 1.7g) and P(o-tolyl)3 (5 g) were added and flushed into the mixture with THF (35 mL). The reaction was exothermic. The reaction mixture was stirred at 50 °C for 1 h. Ethyl acetate and water were added, and the organic layer was separated and dried over magnesium sulfate. Product was purified on silica gel (hexane/chloroform -» chloroform — > chloroform/methanol) to give 60 g of the title compound. Preparation 4
4-(4-Fluorobenzyl)-5,6,7,8-tetrahydropyrido[4,3-d]pyrimidine
Figure imgf000047_0001
The title compound from Preparation 3 (60 g) was hydrogenated in methanol containing aqueous hydrochloric acid in the presence of Pd/C (25 g) at room temperature and 1 atm (lxlO5 N/m2) pressure of hydrogen to give 20 g of the title compound as its HC1 salt. 'H NMR (300MHz, D6-DMSO) δ: 2.95 (m, 2H), 3.40 (m, 2H), 4.13 (s, 2H), 4.26 (s, 2H), 5.80 (br s, 1H), 7.12 (m, 2H), 7.25 (m, 2H), 8.95 (s, 1 H), 9.65 (br s, 1H). Example 54
The compounds of the examples were tested in the Autotaxin FS3 assay described above, with the following results:
Example number ICso
1 7.20 nM
2 2.59 nM
3 251 nM
4 18.7 nM
5 19.5 nM
6 34.5 nM
7 433 nM
8 153 nM
9 327 nM 60.7 nM
124 nM
11.8 nM
7.01 nM
13.7 nM
261 nM
48.0 nM
32.7 nM
34.0 nM
196 nM
3.57 nM
28.3 nM
92.2 nM
91.9 nM
167 nM
96.1 nM
6.00 nM
33.8 nM
60.6 nM
229 nM
89.8 nM
155 nM
141 nM
84.6 nM 259 nM
31.9 nM
100 nM
38.1 nM
>321 nM
14.5 nM
16.7 nM
216 nM
225 nM
41 1 nM
474 nM
382 nM
168 nM
276 nM
345 nM
129 nM
302 nM
449 nM
332 nM
318 nM

Claims

Claims:
1. A compound of formula I,
Figure imgf000050_0001
wherein
one of X, Y and Z represents N and the other two represent CH, or X=Y-Z represents N=CH-N;
W represents a C .e alkylene group which may be straight or branched;
R1 represents H, phenyl, naphthyl, heteroaryl', heterocyclyl1 or C3-6 cycloalkyl;
wherein each phenyl, naphthyl, heteroaryl1, heterocyclyl1 and C3.6 cycloalkyl group is optionally substituted with from 1 to 3 substituents selected from halo, Ci_s alkyl optionally substituted with 1 -3 halogen atoms, phenyl, C e alkoxy optionally substituted with 1 -3 halogen atoms, cyano, heteroaryl'3 and heterocyclyl'3;
and wherein each phenyl, naphthyl, heteroaryl1, heterocyclyl1 and C3-6 cycloalkyl group is optionally fused to a 5- or 6-membered heteroaromatic or heterocyclic ring containing from 1 to 3 heteroatoms (selected from N, O and S); and each heteroaryl1, heterocyclyl1 and C3.6 cycloalkyl group is optionally fused to a benzene ring; and when the group is substituted the substitution may occur anywhere on the optionally fused ring system as a whole;
and wherein heterocyclyl1 and heterocyclyl13 may additionally be substituted with
=0;
m is 1 or 2;
n is 0, 1 or 2;
A is C=0 or S02;
V represents a bond or C i.g alkylene which may be straight or branched;
R2 represents H, -S-(d-6 alkyl), -S02N(C,-6 alkyl)2, -C02-(Ci.6 alkyl), -NHCOCH2-phenyl, -OCH2-phenyl, -S02-phenyl, -NR3R4, phenyl, naphthyl, heteroaryl2, heterocyclyl2, -CO-heterocyclyl2 or C _ cycloalkyl; wherein each phenyl, naphthyl, heteroaryl2, heterocyclyl2 and C3. cycloalkyl group is optionally substituted with from 1 to 3 substituents selected from halo, Ci-6 alkyl optionally substituted with 1-3 halogen atoms, phenyl (optionally substituted with CN or Ci-6 alkyl), phenoxy, benzyloxy, N02, -NHS02(C,.6 alkyl), -S02(C1-6 alkyl), pyridylmethyl, Ci_6 alkoxy optionally substituted with 1-3 halogen atoms, cyano, heteroaryl23 and heterocyclyl23;
and wherein each phenyl, naphthyl, heteroaryl2, heterocyclyl2 and C3-6 cycloalkyl group is optionally fused to a 5- or 6-membered heteroaromatic or heterocyclic ring containing from 1 to 3 heteroatoms (selected from N, O and S); and wherein each heteroaryl2, heterocyclyl2 and C3.6 cycloalkyl group is optionally fused to a benzene ring; and when the group is substituted the substitution may occur anywhere on the optionally fused ring system as a whole;
and wherein heterocyclyl2 and heterocyclyl23 may additionally be substituted with =0;
R3 represents heterocyclyl2;
R4 represents phenyl-(Ci_6 alkylene)-, wherein the phenyl group is optionally substituted with from 1 to 3 substituents selected from halo, Ci-6 alkyl optionally substituted with 1 -3 halogen atoms, Ci-e alkoxy optionally substituted with 1-3 halogen atoms, and cyano; heteroaryl1, heteroaryl13, heteroaryl2 and heteroaryl23 independently represent a 5- or 6-membered heteroaryl group containing from 1 to 3 heteroatoms (selected from N, O and S); and
heterocyclyl1, heterocyclyl13, heterocyclyl2 and heterocyclyl23 independently represent a 5- or 6-membered heterocyclyl group containing from 1 to 3 heteroatoms (selected from N, O and S);
and pharmaceutically acceptable salts and solvates thereof.
2. A compound as claimed in claim 1 , wherein X=Y-Z represents N=CH-N, and pharmaceutically acceptable salts and solvates thereof.
3. A compound as claimed in claim 1 or 2, wherein W is C¾, and pharmaceutically acceptable salts and solvates thereof.
4. A compound as claimed in any one of the preceding claims, wherein m is 1, and pharmaceutically acceptable salts and solvates thereof.
5. A compound as claimed in any one of the preceding claims, wherein n is 1 , and pharmaceutically acceptable salts and solvates thereof.
6. A compound as claimed in any one of the preceding claims, wherein A is CO, and pharmaceutically acceptable salts and solvates thereof.
7. A compound as claimed in any one of the preceding claims, wherein R1 is phenyl substituted by F or OCF3, and pharmaceutically acceptable salts and solvates thereof.
8. A compound as claimed in any one of the preceding claims, wherein V is selected from a bond or C alkylene which may be straight or branched, and pharmaceutically acceptable salts and solvates thereof.
9. A compound as claimed in any one of the preceding claims wherein R2 is selected from H, -S-(C1-6 alkyl), -S02N(Ci-6 alkyl)2, -C02-(C,-6 alkyl), -NHCOCH2-phenyl, -OCH2-phenyl, -S02-phenyl, -NR3R4, phenyl, naphthyl, heteroaryl2, heterocyclyl2,
-CO-4-methylpiperazin- l-yl or C3.5 cycloalkyl;
Wherein phenyl is optionally fused to tetrahydrofuran and said phenyl or dihydrobenzofuran is optionally substituted by 1-2 substituents selected from CI, F, OCF3,
N02, OCH3, NS02CH3, S02CH3, OCH2Ph and OPh
Wherein heteroaryl2 is a 5-6 membered heteroaryl group containing from 1 to 2 heteroatoms (selected from N, O and S) said heteroaryl group being optionally fused to a benzene ring and the optionally fused system may be substituted at any position by OPh;
Wherein heterocyclyl2 is a 5 membered heterocyclyl group containing 1 -2 N atoms, said heterocyclyl group being optionally fused to a benzene ring and the optionally fused system may be substituted at any position by =0, CH2Ph or CH2-C5NH5
and pharmaceutically acceptable salts and solvates thereof.
10. A compound as claimed in any one of the preceding claims, wherein R2 is selected from H, -SCH3, -S02N(CH3)2, -C02-(C1-2 alkyl), -NHCOCH2-phenyl, -OCH2-phenyl, -S02-phenyl, -NR3R4, phenyl, naphthyl, heteroaryl2, heterocyclyl2 , -CO-4-methylpiperazin- 1 -yl or C3.5 cycloalkyl;
Wherein phenyl is optionally substituted by 1-2 substituents selected from CI, F, OCF3, N02, OCH3, NSO2CH3, S02CH3, OCH2Ph and OPh; or is optionally fused with tetrahydrofuran to form a 2,3-dihydrobenzofuran moiety.
Wherein heteroaryl2 is selected from furanyl, 1 ,3-benzothiazolyl, pyridinyl substituted by a phenoxy group and an indazolyl;
Wherein heterocyclyl2 is selected from oxoimidazolidinyl, isoindolin-l-one and pyrrolidin-2-one substituted by CH2-pyridine.
1 1. A compound as claimed in any one of the preceding claims, wherein R3 is tetrahydropyranyl and R4 is benzyl and pharmaceutically acceptable salts and solvates thereof.
12. A compound of formula I, as defined in any one of the preceding claims, or a pharmaceutically acceptable salt or solvate thereof, for use as a pharmaceutical.
13. A compound of formula I, as defined in any one of the preceding claims, or a pharmaceutically acceptable salt or solvate thereof, for use in the treatment of a disorder selected from: pain, acute pain, chronic pain, neuropathic pain, inflammatory pain, osteoarthritis, visceral pain, nociceptive pain including post-surgical pain, and mixed pain types involving the viscera, gastrointestinal tract, cranial structures, musculoskeletal system, spine, urogenital system, cardiovascular system and CNS, including cancer pain, back and orofacial pain.
14. A compound of formula I, as defined in any one of the preceding claims, or a pharmaceutically acceptable salt or solvate thereof, for use in the treatment of a disorder selected from: cancer (including ovarian cancer, breast cancer and prostate cancer), atherosclerosis, thrombosis, psoriasis, multiple sclerosis, neurodegenerative disorders, irritable bowel syndrome, rheumatoid arthritis, neuropathological disorders, functional bowel disorders, inflammatory bowel diseases, pain associated with dysmenorrhea, pelvic pain, cystitis, pancreatitis, migraine, cluster and tension headaches, diabetic neuropathy, peripheral neuropathic pain, sciatica, fibromyalgia, eausalgia, and conditions of lower urinary tract dysfunction.
15. A compound of formula I, as defined in any one of the preceding claims, or a pharmaceutically acceptable salt or solvate thereof, for use in the treatment of fibrotic diseases including pulmonary fibrosis, Cirrhosis, endomyocardial fibrosis, mediastinal fibrosis, myelofibrosis, retroperitoneal fibrosis, nephrogenic systemic fibrosis, Crohn's disease, Keloid, old myocardial infarction, scleroderma / systemic sclerosis, atherofibrosis and adhesive capsulitis.
16. A pharmaceutical formulation comprising a compound of formula I, as defined in any one of the preceding claims, or a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable adjuvant, diluent or carrier.
17. A pharmaceutical formulation as claimed in claim 16, further comprising one or more additional active agents for the treatment of a disorder defined in claim 13, 14 or claim 15.
18. A pharmaceutical kit comprising a compound of formula I, as defined in any one of the preceding claims, or a pharmaceutically acceptable salt or solvate thereof, and one or more additional active agents, as a combined preparation for separate, simultaneous or sequential administration in the treatment of a disorder defined in claim 13, 14 or claim 15.
19. A method of treatment of a disorder defined in claim 13, 14 or claim 15 in a mammal, comprising administration of a therapeutically effective amount of a compound of formula I, as defined in any one of the preceding claims, or a pharmaceutically acceptable salt or solvate thereof, to a mammal in need of such treatment.
20. A process for the production of a compound of formula I, as defined in any one of the preceding claims, or a pharmaceutically acceptable salt or solvate thereof, which comprises coupling a compound of formula II,
Figure imgf000054_0001
wherein R , W, X, Y, Z, m and n are as defined in claim 1,
with a compound of formula III,
Figure imgf000054_0002
wherein A, V and R2 are as defined in claim 1, and L is a leaving group.
PCT/IB2012/002073 2011-10-13 2012-10-15 Pyrimidine and pyridine derivatives useful in therapy WO2013054185A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201161546647P 2011-10-13 2011-10-13
US61/546,647 2011-10-13

Publications (1)

Publication Number Publication Date
WO2013054185A1 true WO2013054185A1 (en) 2013-04-18

Family

ID=47297324

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2012/002073 WO2013054185A1 (en) 2011-10-13 2012-10-15 Pyrimidine and pyridine derivatives useful in therapy

Country Status (1)

Country Link
WO (1) WO2013054185A1 (en)

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014110000A1 (en) * 2013-01-11 2014-07-17 Eli Lilly And Company Pyrido- or pyrrolo-fused pyrimidine derivatives as autotaxin inhibitors for treating pain
WO2015078803A1 (en) * 2013-11-26 2015-06-04 F. Hoffmann-La Roche Ag NEW OCTAHYDRO-CYCLOBUTA [1,2-c;3,4-c']DIPYRROL-2-YL
US9051320B1 (en) 2014-08-18 2015-06-09 Pharmakea, Inc. Methods for the treatment of metabolic disorders by a selective small molecule autotaxin inhibitor
US9334261B2 (en) 2013-11-22 2016-05-10 Pharmakea, Inc. Autotaxin inhibitor compounds
WO2016197009A1 (en) 2015-06-05 2016-12-08 Vertex Pharmaceuticals Incorporated Triazoles for the treatment of demyelinating diseases
WO2017037146A1 (en) * 2015-09-04 2017-03-09 F. Hoffmann-La Roche Ag Phenoxymethyl derivatives
US9714240B2 (en) 2013-09-17 2017-07-25 Pharmakea, Inc. Vinyl autotaxin inhibitor compounds
US9802944B2 (en) 2014-03-26 2017-10-31 Hoffmann-La Roche Inc. Bicyclic compounds as autotaxin (ATX) and lysophosphatidic acid (LPA) production inhibitors
US9850203B2 (en) 2013-09-26 2017-12-26 Pharmakea, Inc. Autotaxin inhibitor compounds
US9926318B2 (en) 2013-11-22 2018-03-27 Pharmakea, Inc. Tetracyclic autotaxin inhibitors
US9951026B2 (en) 2013-09-17 2018-04-24 Pharmakea, Inc. Heterocyclic vinyl autotaxin inhibitor compounds
WO2018106643A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Heterocyclic azoles for the treatment of demyelinating diseases
WO2018106646A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Aminotriazoles for the treatment of demyelinating diseases
WO2018106641A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Pyrazoles for the treatment of demyelinating diseases
US10106559B2 (en) 2013-06-25 2018-10-23 Bristol-Myers Squibb Company Substituted tetrahydrocarbazole and carbazole carboxamide compounds
US10138230B2 (en) 2015-02-04 2018-11-27 Cancer Research Technology Limited Autotaxin inhibitors
US10632104B2 (en) 2015-05-27 2020-04-28 Sabre Therapeutics Llc Autotaxin inhibitors and uses thereof
US10633384B2 (en) 2012-06-13 2020-04-28 Hoffmann-La Roche Inc. Diazaspirocycloalkane and azaspirocycloalkane
US10647719B2 (en) 2015-09-24 2020-05-12 Hoffmann-La Roche Inc. Bicyclic compounds as dual ATX/CA inhibitors
US10654846B2 (en) 2015-02-06 2020-05-19 Cancer Research Technology Limited Autotaxin inhibitory compounds
US10669268B2 (en) 2012-09-25 2020-06-02 Hoffmann-La Roche Inc. Bicyclic derivatives
US10669285B2 (en) 2014-03-26 2020-06-02 Hoffmann-La Roche Inc. Condensed [1,4] diazepine compounds as autotaxin (ATX) and lysophosphatidic acid (LPA) production inhibitors
US10676446B2 (en) 2015-04-10 2020-06-09 Hoffmann-La Roche Inc. Bicyclic quinazolinone derivatives
US10738053B2 (en) 2015-09-24 2020-08-11 Hoffmann-La Roche Inc. Bicyclic compounds as dual ATX/CA inhibitors
US10787459B2 (en) 2015-09-24 2020-09-29 Hoffmann-La Roche Inc. Bicyclic compounds as ATX inhibitors
US10800786B2 (en) 2015-09-24 2020-10-13 Hoffman-La Roche Inc. Bicyclic compounds as ATX inhibitors
US10882857B2 (en) 2017-03-16 2021-01-05 Hoffmann-La Roche Inc. Bicyclic compounds as ATX inhibitors
US10913745B2 (en) 2013-03-12 2021-02-09 Hoffmann-La Roche Inc. Octahydro-pyrrolo[3,4-c]-pyrrole derivatives and analogs thereof as autotaxin inhibitors
US10961242B2 (en) 2017-05-17 2021-03-30 Legochem Biosciences, Inc. Compounds as autotaxin inhibitors and pharmaceutical compositions comprising the same
US11059794B2 (en) 2017-03-16 2021-07-13 Hoffmann-La Roche Inc. Heterocyclic compounds useful as dual ATX/CA inhibitors
WO2022022569A1 (en) * 2020-07-28 2022-02-03 武汉人福创新药物研发中心有限公司 Solid form of nitrogen-containing heterocyclic compound, and pharmaceutical composition and use thereof
US11548883B2 (en) 2017-05-17 2023-01-10 Legochem Biosciences, Inc. Compounds as autotaxin inhibitors and pharmaceutical compositions comprising the same

Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991011172A1 (en) 1990-01-23 1991-08-08 The University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
WO1994002518A1 (en) 1992-07-27 1994-02-03 The University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
WO1998055148A1 (en) 1997-06-05 1998-12-10 Janssen Pharmaceutica N.V. Pharmaceutical compositions comprising cyclodextrins
WO2000035298A1 (en) 1996-11-27 2000-06-22 Wm. Wrigley Jr. Company Chewing gum containing medicament active agents
US6106864A (en) 1995-09-15 2000-08-22 Pfizer Inc. Pharmaceutical formulations containing darifenacin
WO2008047229A2 (en) 2006-10-18 2008-04-24 Pfizer Products Inc. Biaryl ether urea compounds
WO2008118758A1 (en) 2007-03-23 2008-10-02 Icagen, Inc. Inhibitors of ion channels
WO2008135826A2 (en) 2007-05-03 2008-11-13 Pfizer Limited 2 -pyridine carboxamide derivatives as sodium channel modulators
WO2009012242A2 (en) 2007-07-13 2009-01-22 Icagen, Inc. Sodium channel inhibitors
WO2010060532A1 (en) * 2008-11-28 2010-06-03 Merck Patent Gmbh Benzonaphtyridine compounds used as inhibitors of autotaxin
WO2010063352A1 (en) * 2008-12-01 2010-06-10 Merck Patent Gmbh 2, 5-diamino-substituted pyrido [4, 3-d] pyrimidines as autotaxin inhibitors against cancer
US7989663B2 (en) 2002-12-10 2011-08-02 Echelon Biosciences Incorporated Compounds and methods of use thereof for assaying lysophospholipase D activity
WO2011116867A1 (en) * 2010-03-26 2011-09-29 Merck Patent Gmbh Benzonaphthyridinamines as autotaxin inhibitors

Patent Citations (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1991011172A1 (en) 1990-01-23 1991-08-08 The University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
WO1994002518A1 (en) 1992-07-27 1994-02-03 The University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US6106864A (en) 1995-09-15 2000-08-22 Pfizer Inc. Pharmaceutical formulations containing darifenacin
WO2000035298A1 (en) 1996-11-27 2000-06-22 Wm. Wrigley Jr. Company Chewing gum containing medicament active agents
WO1998055148A1 (en) 1997-06-05 1998-12-10 Janssen Pharmaceutica N.V. Pharmaceutical compositions comprising cyclodextrins
US7989663B2 (en) 2002-12-10 2011-08-02 Echelon Biosciences Incorporated Compounds and methods of use thereof for assaying lysophospholipase D activity
WO2008047229A2 (en) 2006-10-18 2008-04-24 Pfizer Products Inc. Biaryl ether urea compounds
WO2008118758A1 (en) 2007-03-23 2008-10-02 Icagen, Inc. Inhibitors of ion channels
WO2008135826A2 (en) 2007-05-03 2008-11-13 Pfizer Limited 2 -pyridine carboxamide derivatives as sodium channel modulators
WO2009012242A2 (en) 2007-07-13 2009-01-22 Icagen, Inc. Sodium channel inhibitors
WO2010060532A1 (en) * 2008-11-28 2010-06-03 Merck Patent Gmbh Benzonaphtyridine compounds used as inhibitors of autotaxin
WO2010063352A1 (en) * 2008-12-01 2010-06-10 Merck Patent Gmbh 2, 5-diamino-substituted pyrido [4, 3-d] pyrimidines as autotaxin inhibitors against cancer
WO2011116867A1 (en) * 2010-03-26 2011-09-29 Merck Patent Gmbh Benzonaphthyridinamines as autotaxin inhibitors

Non-Patent Citations (31)

* Cited by examiner, † Cited by third party
Title
"Bioreversible Carriers in Drug Design", 1987, PERGAMON PRESS
"Remington's Pharmaceutical Sciences", 1995, MACK PUBLISHING COMPANY
A.L. PARRILL ET AL.: "Autotaxin inhibitors: a perspective on initial medicinal chemistry efforts.", EXPERT OPINION ON THERAPEUTIC PATENTS, vol. 20, no. 12, 2010 - December 2010 (2010-12-01), Informa, UK, pages 1619 - 1625, XP002690642, ISSN: 1354-3776 *
ALVAREZ ET AL.: "Autocrine and paracrine roles of sphingosine-1-phosphate", TRENDS IN ENDOCRINOLOGY AND METABOLISM, vol. 18, no. 8, 2007, pages 300 - 307, XP022289713, DOI: doi:10.1016/j.tem.2007.07.005
AOKI ET AL.: "Two pathways for lysophosphatidic acid production", BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1781, no. 9, 2008, pages 513 - 518, XP024339627, DOI: doi:10.1016/j.bbalip.2008.06.005
E. L. ELIEL; S. H. WILEN: "Stereochemistry of Organic Compounds", 1994, WILEY
FINNIN; MORGAN, J PHARM SCI, vol. 88, no. 10, October 1999 (1999-10-01), pages 955 - 958
GIERSE ET AL.: "A novel autotaxin inhibitor reduces lysophosphatidic acid levels in plasma and the site of inflammation", JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 334, no. 1, 2010, pages 310 - 317, XP002666664, DOI: doi:10.1124/jpet.110.165845
GIJSBERS ET AL.: "The hydrolysis of lysophospholipids and nucleotides by autotaxin (NPP2) involves a single catalytic site", FEBS LETTERS, vol. 538, no. 1- 3, 2003, pages 60 - 64, XP004842825, DOI: doi:10.1016/S0014-5793(03)00133-9
H. BUNDGAARD: "Design of Prodrugs", 1985, ELSEVIER
H. LIEBERMAN; L. LACHMAN: "Pharmaceutical Dosage Forms: Tablets", vol. 1, 1980, MARCEL DEKKER
HALEBLIAN, J PHARM SCI, vol. 64, no. 8, August 1975 (1975-08-01), pages 1269 - 1288
JANSEN ET AL.: "Proteolytic maturation and activation of autotaxin (NPP2), a secreted metastasis-enhancing lysophospholipase D", JOURNAL OF CELL SCIENCE, vol. 118, no. 14, 2005, pages 3081 - 3089
LIANG; CHEN, EXPERT OPINION IN THERAPEUTIC PATENTS, vol. 11, no. 6, 2001, pages 981 - 986
MASUDA ET AL.: "Serum autotaxin measurement in haematological malignancies: a promising marker for follicular lymphoma", BRITISH JOURNAL OF HAEMATOLOGY, vol. 143, no. 1, 2008, pages 60 - 70, XP002536839, DOI: doi:10.1111/J.1365-2141.2008.07325.X
NAKAMURA ET AL.: "Analysis of serum and urinary lysophospholipase D/autotaxin in nephrotic syndrome", CLINICAL CHEMISTRY AND LABORATORY MEDICINE, vol. 46, no. 1, 2008, pages 150 - 151
NAKAMURA ET AL.: "Autotaxin enzyme immunoassay in human cerebrospinal fluid samples", CLINICA CHIMICA ACTA, vol. 405, no. 1-2, 2009, pages 160 - 162, XP026159584, DOI: doi:10.1016/j.cca.2009.04.025
NAKANAGA ET AL.: "Autotaxin-An LPA producing enzyme with diverse functions", JOURNAL OFBIO CHEMISTRY, vol. 148, no. 1, 2010, pages 13 - 24, XP002666667, DOI: doi:10.1093/JB/MVQ052
OKUDAIRA ET AL.: "Biological roles of lysophosphatidic acid signaling through its production by autotaxin", BIOCHIMIE, vol. 92, no. 6, 2010, pages 698 - 706
STAHL; WERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCH
STEFAN ET AL.: "NPP-type ectophosphodiesterases: unity in diversity", TRENDS IN BIOCHEMICAL SCIENCES, vol. 30, no. 10, 2005, pages 542 - 550, XP005090072, DOI: doi:10.1016/j.tibs.2005.08.005
STRACKE: "Autotaxin is an N-linked glycoprotein but the sugar moieties are not needed for its stimulation of cellular motility", MELANOMA RESEARCH, vol. 5, no. 4, 1995, pages 203 - 209
SUGIURA ET AL.: "Lysophosphatidic acid, a growth factor- like lipid, in the saliva", JOURNAL OF LIPID RESEARCH, vol. 43, no. 12, 2002, pages 2049 - 2055
T. HIGUCHI; W. STELLA: "ACS Symposium Series", vol. 14, article "Pro-drugs as Novel Delivery Systems"
TANAKA ET AL.: "Autotaxin stabilizes blood vessels and is required for embryonic vasculature by producing lysophosphatidic acid", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 281, no. 35, 2006, pages 25822 - 25830, XP002692832, DOI: doi:10.1074/jbc.M605142200
TANAKA ET AL.: "Prostatic acid phosphatase degrades lysophosphatidic acid in seminal plasma", FEBS LETTERS, vol. 571, no. 1-3, 2004, pages 197 - 204, XP004523337, DOI: doi:10.1016/j.febslet.2004.06.083
THEORORA GREENE; PETER WUTS: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY AND SONS
VAN MEETEREN ET AL.: "Inhibition of autotaxin by lysophosphatidic acid and sphingosine 1-phosphate", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 280, no. 22, 2005, pages 21155 - 21161
VAN MEETEREN ET AL.: "Regulation and biological activities of the autotaxin-LPA axis", PROGRESS IN LIPID RESEARCH, vol. 46, no. 2, 2007, pages 145 - 160, XP022051590, DOI: doi:10.1016/j.plipres.2007.02.001
VERMA ET AL., PHARMACEUTICAL TECHNOLOGY, vol. 25, no. 2, 2001, pages 1 - 14
YUELLING ET AL.: "Autotaxin (ATX): a multifunctional and multi-modular protein possessing enzymatic lysoPLD activity and matricellular properties", BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1781, no. 9, 2008, pages 525 - 530, XP024339629, DOI: doi:10.1016/j.bbalip.2008.04.009

Cited By (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10633384B2 (en) 2012-06-13 2020-04-28 Hoffmann-La Roche Inc. Diazaspirocycloalkane and azaspirocycloalkane
US10669268B2 (en) 2012-09-25 2020-06-02 Hoffmann-La Roche Inc. Bicyclic derivatives
US8969555B2 (en) 2013-01-11 2015-03-03 Eli Lilly And Company Bicyclic pyrimidine compounds
WO2014110000A1 (en) * 2013-01-11 2014-07-17 Eli Lilly And Company Pyrido- or pyrrolo-fused pyrimidine derivatives as autotaxin inhibitors for treating pain
JP2016505010A (en) * 2013-01-11 2016-02-18 イーライ リリー アンド カンパニー Bicyclic pyrimidine compounds
EA025106B1 (en) * 2013-01-11 2016-11-30 Эли Лилли Энд Компани Bicyclic pyrimidine compounds
US10913745B2 (en) 2013-03-12 2021-02-09 Hoffmann-La Roche Inc. Octahydro-pyrrolo[3,4-c]-pyrrole derivatives and analogs thereof as autotaxin inhibitors
US10435415B2 (en) 2013-06-25 2019-10-08 Bristol-Myers Squibb Company Substituted tetrahydrocarbazole and carbazole carboxamide compounds
US10106559B2 (en) 2013-06-25 2018-10-23 Bristol-Myers Squibb Company Substituted tetrahydrocarbazole and carbazole carboxamide compounds
US9714240B2 (en) 2013-09-17 2017-07-25 Pharmakea, Inc. Vinyl autotaxin inhibitor compounds
US9951026B2 (en) 2013-09-17 2018-04-24 Pharmakea, Inc. Heterocyclic vinyl autotaxin inhibitor compounds
US9850203B2 (en) 2013-09-26 2017-12-26 Pharmakea, Inc. Autotaxin inhibitor compounds
US9468628B2 (en) 2013-11-22 2016-10-18 Pharmakea, Inc Autotaxin inhibitor compounds
US10688081B2 (en) 2013-11-22 2020-06-23 Sabre Therapeutics Llc Autotaxin inhibitor compounds
US11344533B2 (en) 2013-11-22 2022-05-31 Sabre Therapeutics Llc Autotaxin inhibitor compounds
US9334261B2 (en) 2013-11-22 2016-05-10 Pharmakea, Inc. Autotaxin inhibitor compounds
US9926318B2 (en) 2013-11-22 2018-03-27 Pharmakea, Inc. Tetracyclic autotaxin inhibitors
US11779568B2 (en) 2013-11-22 2023-10-10 Sabre Therapeutics Llc Autotaxin inhibitor compounds
US9999615B2 (en) 2013-11-22 2018-06-19 Pharmakea, Inc. Autotaxin inhibitor compounds
AU2014356583B2 (en) * 2013-11-26 2019-02-28 F. Hoffmann-La Roche Ag New octahydro-cyclobuta (1,2-c;3,4-c')dipyrrol-2-yl
US10849881B2 (en) 2013-11-26 2020-12-01 Hoffmann-La Roche Inc. Octahydro-cyclobuta[1,2-c;3,4-c′]dipyrrol-2-yl
JP2016538303A (en) * 2013-11-26 2016-12-08 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Novel octahydro-cyclobuta [1,2-c; 3,4-c '] dipyrrol-2-yl
CN105764905A (en) * 2013-11-26 2016-07-13 豪夫迈·罗氏有限公司 New octahydro-cyclobuta [1,2-c;3,4-c']dipyrrol-2-yl
CN105764905B (en) * 2013-11-26 2019-06-07 豪夫迈·罗氏有限公司 New octahydro-cyclobutadiene simultaneously [1,2-c;3,4-c '] -2 base of two pyrroles
WO2015078803A1 (en) * 2013-11-26 2015-06-04 F. Hoffmann-La Roche Ag NEW OCTAHYDRO-CYCLOBUTA [1,2-c;3,4-c']DIPYRROL-2-YL
US10669285B2 (en) 2014-03-26 2020-06-02 Hoffmann-La Roche Inc. Condensed [1,4] diazepine compounds as autotaxin (ATX) and lysophosphatidic acid (LPA) production inhibitors
US11098048B2 (en) 2014-03-26 2021-08-24 Hoffmann-La Roche Inc. Bicyclic compounds as autotaxin (ATX) and lysophosphatidic acid (LPA) production inhibitors
US9802944B2 (en) 2014-03-26 2017-10-31 Hoffmann-La Roche Inc. Bicyclic compounds as autotaxin (ATX) and lysophosphatidic acid (LPA) production inhibitors
US10654857B2 (en) 2014-03-26 2020-05-19 Hoffman-La Roche Inc. Bicyclic compounds as autotaxin (ATX) and lysophosphatidic acid (LPA) production inhibitors
US9051320B1 (en) 2014-08-18 2015-06-09 Pharmakea, Inc. Methods for the treatment of metabolic disorders by a selective small molecule autotaxin inhibitor
US10428061B2 (en) 2015-02-04 2019-10-01 Cancer Research Technology Limited Autotaxin inhibitors
US10138230B2 (en) 2015-02-04 2018-11-27 Cancer Research Technology Limited Autotaxin inhibitors
US10654846B2 (en) 2015-02-06 2020-05-19 Cancer Research Technology Limited Autotaxin inhibitory compounds
US11453666B2 (en) 2015-02-06 2022-09-27 Cancer Research Technology Limited Autotaxin inhibitory compounds
US10676446B2 (en) 2015-04-10 2020-06-09 Hoffmann-La Roche Inc. Bicyclic quinazolinone derivatives
US10632104B2 (en) 2015-05-27 2020-04-28 Sabre Therapeutics Llc Autotaxin inhibitors and uses thereof
WO2016197009A1 (en) 2015-06-05 2016-12-08 Vertex Pharmaceuticals Incorporated Triazoles for the treatment of demyelinating diseases
IL256548A (en) * 2015-09-04 2018-02-28 Hoffmann La Roche Phenoxymethyl derivatives
CN108026077A (en) * 2015-09-04 2018-05-11 豪夫迈·罗氏有限公司 Phenoxymethyl derivative
US11352330B2 (en) 2015-09-04 2022-06-07 Hoffmann-La Roche Inc. Phenoxymethyl derivatives
CN108026077B (en) * 2015-09-04 2021-11-05 豪夫迈·罗氏有限公司 Phenoxymethyl derivatives
WO2017037146A1 (en) * 2015-09-04 2017-03-09 F. Hoffmann-La Roche Ag Phenoxymethyl derivatives
US10640472B2 (en) 2015-09-04 2020-05-05 Hoffman-La Roche Inc. Phenoxymethyl derivatives
RU2746481C1 (en) * 2015-09-04 2021-04-14 Ф. Хоффманн-Ля Рош Аг Phenoxymethyl derivatives
US10800786B2 (en) 2015-09-24 2020-10-13 Hoffman-La Roche Inc. Bicyclic compounds as ATX inhibitors
US10889588B2 (en) 2015-09-24 2021-01-12 Hoffmann-La Roche Inc. Bicyclic compounds as dual ATX/CA inhibitors
US10647719B2 (en) 2015-09-24 2020-05-12 Hoffmann-La Roche Inc. Bicyclic compounds as dual ATX/CA inhibitors
US10787459B2 (en) 2015-09-24 2020-09-29 Hoffmann-La Roche Inc. Bicyclic compounds as ATX inhibitors
US10738053B2 (en) 2015-09-24 2020-08-11 Hoffmann-La Roche Inc. Bicyclic compounds as dual ATX/CA inhibitors
WO2018106646A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Aminotriazoles for the treatment of demyelinating diseases
WO2018106641A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Pyrazoles for the treatment of demyelinating diseases
WO2018106643A1 (en) 2016-12-06 2018-06-14 Vertex Pharmaceuticals Incorporated Heterocyclic azoles for the treatment of demyelinating diseases
US10882857B2 (en) 2017-03-16 2021-01-05 Hoffmann-La Roche Inc. Bicyclic compounds as ATX inhibitors
US11059794B2 (en) 2017-03-16 2021-07-13 Hoffmann-La Roche Inc. Heterocyclic compounds useful as dual ATX/CA inhibitors
US11673888B2 (en) 2017-03-16 2023-06-13 Hoffmann-La Roche Inc. Bicyclic compounds as ATX inhibitors
US11548883B2 (en) 2017-05-17 2023-01-10 Legochem Biosciences, Inc. Compounds as autotaxin inhibitors and pharmaceutical compositions comprising the same
US10961242B2 (en) 2017-05-17 2021-03-30 Legochem Biosciences, Inc. Compounds as autotaxin inhibitors and pharmaceutical compositions comprising the same
WO2022022569A1 (en) * 2020-07-28 2022-02-03 武汉人福创新药物研发中心有限公司 Solid form of nitrogen-containing heterocyclic compound, and pharmaceutical composition and use thereof

Similar Documents

Publication Publication Date Title
EP2771325B1 (en) Pyridazine derivatives useful in therapy
WO2013054185A1 (en) Pyrimidine and pyridine derivatives useful in therapy
US7649004B2 (en) Pyridine derivatives
US9085517B2 (en) Sulfonamide derivatives
US7566739B2 (en) Substituted N-sulfonylaminophenylethyl-2-phenoxyacetamide compounds as VR1 receptor antagonists
EP1861400B1 (en) Crystal forms of an imidazole derivative
ES2368460T3 (en) SUBSTITUTED BICICLOCARBOXYAMIDE PHENYLMETIL COMPOUNDS.
WO2007129188A1 (en) Cyclopropanecarboxamide compound
US20100240652A1 (en) Pyridine Derivatives as Sodium Channel Modulators
EP2903989A1 (en) Pyrrolo[2,3-d]pyrimidine tropomyosin-related kinase inhibitors
US20150250785A1 (en) Tropomyosin-Related Kinase Inhibitors
US20080207665A1 (en) Alpha-(Aryl-or Heteroaryl-Methyl)-Beta-Piperidinopropanoic Acid Compounds as Orl-1-Receptor Antagonists
US20090227680A1 (en) Amino Acid Derivatives

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: 12797968

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: 12797968

Country of ref document: EP

Kind code of ref document: A1