WO2014195228A1 - Screening method for a glut9 inhibitor - Google Patents

Screening method for a glut9 inhibitor Download PDF

Info

Publication number
WO2014195228A1
WO2014195228A1 PCT/EP2014/061212 EP2014061212W WO2014195228A1 WO 2014195228 A1 WO2014195228 A1 WO 2014195228A1 EP 2014061212 W EP2014061212 W EP 2014061212W WO 2014195228 A1 WO2014195228 A1 WO 2014195228A1
Authority
WO
WIPO (PCT)
Prior art keywords
glut9
cell
species
kinetics
receptor
Prior art date
Application number
PCT/EP2014/061212
Other languages
French (fr)
Other versions
WO2014195228A8 (en
Inventor
Alan WISE
Andrew James MHYRE
Original Assignee
Tpp Global Development Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Tpp Global Development Limited filed Critical Tpp Global Development Limited
Publication of WO2014195228A1 publication Critical patent/WO2014195228A1/en
Publication of WO2014195228A8 publication Critical patent/WO2014195228A8/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/5308Immunoassay; Biospecific binding assay; Materials therefor for analytes not provided for elsewhere, e.g. nucleic acids, uric acid, worms, mites
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/557Immunoassay; Biospecific binding assay; Materials therefor using kinetic measurement, i.e. time rate of progress of an antigen-antibody interaction
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/566Immunoassay; Biospecific binding assay; Materials therefor using specific carrier or receptor proteins as ligand binding reagents where possible specific carrier or receptor proteins are classified with their target compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/34Genitourinary disorders
    • G01N2800/347Renal failures; Glomerular diseases; Tubulointerstitial diseases, e.g. nephritic syndrome, glomerulonephritis; Renovascular diseases, e.g. renal artery occlusion, nephropathy

Definitions

  • the present invention relates to a screening method for identifying an agent that is a GLUT9 inhibitor, and/or an agent capable if inhibiting the uptake or release of a species into or from a cell via the GLUT9 receptor.
  • the species comprises a urate anion
  • the method is thus in particular a method for screening for, or identifying, a medicament for treating gout and diseases associated with hyperuricemia.
  • the invention also relates to agents, medicaments, pharmaceutical compositions and methods of treatment containing the gout treatment agents.
  • uric acid The poorly soluble acid, uric acid (UA), is the end-product of purine metabolism in humans and great apes, which have lost hepatic uricase activity during hominid evolution. Loss of uricase activity leads to significantly higher serum UA (sUA) in humans (-240-360 ⁇ , 4-6 mg/dl) than in other mammals (-3-120 ⁇ , ⁇ l -2 mg dl). Abnormally high levels of sUA (>420 ⁇ , 7 mg/dl) is known as hyperuricemia and is the precursor to the deposition of UA crystals in and around the joints, which is the causative factor in the inflammatory arthritic condition gout.
  • gout New therapeutics for the treatment of gout represent an attractive proposition for a number of reasons.
  • the incidence of gout is around 1 -2% of adults in developed countries and is increasing rapidly in developing nations.
  • GLUT9 has been associated with gout and identified as a high capacity urate transporter (Vitart V, Rudan I, Hayward C, Gray NK, Floyd J, Palmer CNA et al. "SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout.” Nature Genetics 2008; 40(4): 437-442). About 70% of daily UA excretion occurs via the kidneys, and in 5-25% of the human population, impaired renal excretion leads to hyperuricemia. Recently, a number of genome-wide association studies led to the identification of a variety of genetic loci linked with variance of sUA levels.
  • the major genetic variant associated with low fractional excretion of UA and susceptibility to gout was found within the SLC2A9 gene, which encodes the GLUT9 transporter.
  • GLUT9 was originally characterised as a hexose transporter.
  • Hereditary mutations in the SLC2A9 gene lead to impaired GLUT9 function and renal hypouricemia, which is characterised by impaired renal reabsorption of UA and subsequent low sUA levels.
  • gout therapies In addition to the blockade of UA production through the use of xanthine oxidase inhibitors such as allopurinol, gout therapies also target the reabsorption of UA in the kidney by blocking UA transporters. Such therapeutics are termed 'uricosurics' and they increase the excretion of UA into the urine. However, existing uricosuric drags such as probenecid and benzbromarone are of limited use due to side effect issues, low potency, and lack of selectivity.
  • GLUT9 inhibition of UA reabsorption by GLUT9 represents an attractive disease-modifying opportunity for gout.
  • novel, selective GLUT9 inhibitors may act as powerful uricosuric agents with enhanced efficacy and safety over existing therapies.
  • a typical assay to screen for inhibitors for such receptors is an uptake assay.
  • transfected cells comprising the necessary receptors are challenged with a potential inhibitor and then the uptake of a relevant transported species is measured (Anzai N, Ichida K, Jutabha P, Kimura T, Babu E, Jin CJ et al. "Plasma Urate Level Is Directly Regulated by a Voltage-driven Urate Efflux Transporter URATv 1 (SLC2A9) in Humans.” Journal of Biological Chemistry 2008; 283(40): 26834-26838).
  • SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout. Nature Genetics 2008; 40(4): 437-442; Bibert S, Hess SK, Firsov D, Thorens B, Geering K, Horisberger JD et al. "Mouse GLUT9: evidences for a urate uniporter.” Am J Physiol Renal Physiol 2009; 297(3): F612-9).
  • oocytes are not suitable for high-throughput screening methods. The cells are larger than regular cells and thus require more space, and are unreliable (as many as 10 cells may be required per test well before a reliable result may be expected).
  • Witkowska et al describe the electrogenic nature of urate uptake by GLUT9. This publication does not link the electrogenic nature of urate uptake with the use of membrane potential-sensitive dyes to provide a kinetic methodology for high-throughput screening for GLUT9 inhibitors.
  • a high-throughput (preferably cell-based) screening assay for identifying GLUT9 inhibitors It is a further aim of the present invention to provide a high-throughput (preferably cell-based) screening method for identifying agents suitable for treating diseases associated with hyperuricemia, which may include but are not limited to: gout, gouty attacks, hypertension, a metabolic syndrome (such as insulin resistance, obesity, dyslipidaemia), atherosclerosis, cardiovascular disease, a kidney disease (such as acute uric acid nephropathy, chronic urate nephropathy, and uric acid nephrolithiasis (urolithiasis)), sarcoidosisis, psoriasis, joint inflammation, arthritis, hyperparathyroidism, plumbism, Lesch-Nylan Syndrome and elley-Seegmiller Syndrome . It is a still further aim of the invention to provide agents, medicaments and pharmaceuticals for treating such diseases, and methods
  • the present invention provides a method for screening for a GLUT9 inhibitor, which method comprises:
  • step (c) determining whether the test compound is a GLUT9 inhibitor from the kinetics measurements taken in step (b).
  • the receptor having a GLUT9 function may be any receptor (or a receptor mimic or analogue) having the same or substantially the same function as GLUT9. It may therefore be any receptor capable of transporting the same species, substantially the same species (such as salts, ions, solvates or the like), or similar species (such as substituted versions of the species) as transported by GLUT9. It thus extends to any receptor encoded by the SCL2A9 gene, including GLUT9, UAQTL2, vURATl and URATvl . Many of these are alternative names for GLUT9, or have only minor differences that do not alter function, such as SNPs in the gene sequence. Thus, this definition extends to any such receptor (or a receptor mimic or analogue) capable of facilitating urate transport across a cell membrane.
  • a test compound comprises any compound, complex, agent or the like which has potential as an inhibitor of GLUT9 (and/or an inhibitor of the receptors as defined above).
  • the test compound may be a small molecule, such as an organic or inorganic compound, a large molecule such as an oligomer or polymer, or a biological molecule such as a peptide, polypeptide, protein, nucleic acid or the like.
  • the compound may be present as a complex, salt, hydrate or other derivative for facilitating solubility, stability, uptake, bioavailability or the like.
  • inhibitor means any compound capable of impairing or detrimentally affecting the GLUT9 function of the receptor in any way (such as preventing, reducing, reversing or diverting the transport of the species across the receptor), including impairing or detrimentally affecting the function of any of the receptors as defined above.
  • the species is any species which is capable of being transported across the GLUT9 receptor. Typically, therefore it is a species which may be transported into or out of a cell (i.e. across the cell membrane) by virtue of the GLUT9 receptor.
  • the species is typically a urate anion, but may be a derivative thereof, such as an organic or inorganic compound, a large molecule such as an oligomer or polymer, or a biological molecule such as a peptide, polypeptide, protein, nucleic acid or the like.
  • the species may be a sugar, such as glucose or fructose, or a sugar derivative, and indeed may be any sugar capable of being transported across the GLUT9 receptor.
  • the species may be present as a complex, salt, hydrate or other derivative for facilitating solubility, stability, uptake, bioavailability or the like.
  • the present inventors have discovered that measurement of kinetics provides an accurate measurement of whether a test compound is a suitable GLUT9 inhibitor. Unlike known uptake assays, which cannot be developed in a manner consistent with high-throughput screening, the kinetics measurements have surprisingly been found to provide reliable information on the throughput of species into and out of the cell, despite the cell uptake data indicating no such transport in many instances.
  • the measurement of kinetics of transport means the successive measurement over time of any parameter which could be related to transport. This could be as few as two measurements, each at a different point in time, but typically involves many such measurements.
  • the kinetics measurements enable a point of comparison, or reference point, to be identified, such that the effect of different inhibitory compounds can more reliably be compared.
  • the parameter being measured is membrane potential (which is related to the transport of species across the cell membrane) that reference point is typically the maximum change in membrane potential (see Figure 4).
  • the change (reduction) in this maximum is indicative of an inhibitory effect on transport across the membrane, and the ability to accurately pinpoint this maximum provides reliability of comparison of inhibitory compounds that could not otherwise be achieved.
  • a method for screening for a GLUT9 inhibitor that inhibits uptake or release of a species into or out of a cell which method comprises:
  • the invention also provides method for screening for an agent for treating a disease associated with hyperuricemia which method comprises a method as defined above.
  • the disease associated with hyperuricemia is not especially limited, and may be any such disease. This may include, but is not limited to the following: gout, gouty attacks, hypertension, a metabolic syndrome (such as insulin resistance, obesity, dyslipidaemia), atherosclerosis, cardiovascular disease, a kidney disease (such as acute uric acid nephropathy, chronic urate nephropathy, and uric acid nephrolithiasis (urolithiasis)), sarcoidosisis, psoriasis, joint inflammation, arthritis, hyperparathyroidism, plumbism, Lesch-Nylan Syndrome and Kelley- Seegmiller Syndrome,
  • a metabolic syndrome such as insulin resistance, obesity, dyslipidaemia
  • atherosclerosis cardiovascular disease
  • a kidney disease such as acute uric acid nephropathy, chronic urate n
  • agent for treating a disease associated with hyperaricemia which agent comprises an agent obtainable from a method as defined above.
  • the disease associated with hyperaricemia is not especially limited, and may be any such disease as defined above.
  • compositions for treating a disease associated with hyperaricemia which composition comprises a compound as defined above.
  • the disease associated with hyperaricemia is not especially limited, and may be any such disease, as defined above.
  • a method for treating a disease associated with hyperaricemia comprises administering a agent as defined above and/or a composition as defined above to a patient.
  • the disease associated with hyperuricemia is not especially limited, and may be any such disease, as defined above.
  • Figure 1 shows an overview of FLIPRTM membrane potential dye kit technology (Molecular Devices LLC).
  • Figure 2 shows a Western blot of crude cell lysates from H4 cells transfected to express hGLUT9. High levels of expression are confirmed in clone #10.
  • Figure 3 shows immunocytochemistry demonstrating cell surface expression of hGLUT9 in H4 cells.
  • Figure 4 shows that membrane potential changes are increased by replacement of NaCl with choline chloride in both the dye loading and urate stimulation buffers.
  • Figure 5 shows representative concentration-response curve data for pharmacological reference compounds in the membrane potential assay.
  • Figure 6 shows representative inhibition data for two hit molecules in an orthogonal oocyte uptake assay.
  • the invention relates to a method for screening for a GLUT9 inhibitor, which method comprises:
  • step (c) determining whether the test compound is a GLUT9 inhibitor from the kinetics measurements taken in step (b).
  • the receptor having a GLUT9 function may be any receptor (or a receptor mimic or analogue) having the same or substantially the same function as GLUT9. It may therefore be any receptor capable of transporting the same species, substantially the same species (such as salts, ions, solvates or the like), or similar species (such as substituted versions of the species) as transported by GLUT9. It thus extends to any receptor encoded by the SCL2A9 gene, including GLUT9, UAQTL2, vURATl and URATvl . Many of these are alternative names for GLUT9, or have only minor differences that do not alter function, such as SNPs in the gene sequence. Thus, this definition extends to any such receptor capable of facilitating urate transport across a cell membrane.
  • the test compound (the GLUT9 inhibitor) is not especially limited, and may comprise any compound, complex, agent or the like which has potential as an inhibitor of GLUT9 (and/or an inhibitor of the receptor as defined above).
  • the test compound may be a small molecule, such as an organic or inorganic compound, a large molecule such as an oligomer or polymer, or a biological molecule such as a peptide, polypeptide, protein, nucleic acid or the like.
  • the compound may be present as a complex, salt, hydrate or other derivative for facilitating solubility, stability, uptake, bioavailability or the like.
  • inhibitor means any compound capable of impairing or detrimentally affecting the GLUT9 function of the receptor in any way (such as preventing, reducing, reversing or diverting the transport of the species across the receptor), including impairing or detrimentally affecting the function of any of the receptors as defined above.
  • the species is not especially limited, and may be any species which is capable of being transported across the GLUT9 receptor. Typically, therefore it is a species which may be transported into or out of a cell (i.e. across the cell membrane) by virtue of the GLUT9 receptor.
  • the species is typically a urate anion, but may be a derivative thereof, such as an organic or inorganic compound, a large molecule such as an oligomer or polymer, or a biological molecule such as a peptide, polypeptide, protein, nucleic acid or the like.
  • the species may be a sugar, such as glucose or fructose, or a sugar derivative, and indeed may be any sugar capable of being transported across the GLUT9 receptor.
  • the species may be present as a complex, salt, hydrate or other derivative for facilitating solubility, stability, uptake, bioavailability or the like.
  • the method preferably comprises the following:
  • step (c) determining whether the test compound is a GLUT9 inhibitor from the kinetics measurements taken in step (b).
  • the receptor having the GLUT9 function is a GLUT9 receptor.
  • the receptor may be an artificially modified receptor, a receptor mimic or the like, provided that it retains the GLUT9 receptor function, and in particular its solute carrying function.
  • the kinetics are typically measured across a cell membrane so as to representative of the transport of the species into and out of the cell itself.
  • the system may be an artificial system modelling transport across a cell membrane, and need (in that case) not involve the cell itself.
  • the species is not especially limited, provided that its transport into and out of a cell is capable of being facilitated by the GLUT9 receptor.
  • the species is typically selected from a substituted or unsubstituted uric acid compound, a substituted or unsubstituted urate compound, and a mixture of two or more of the above.
  • the species comprises a substituted or unsubstituted urate anion.
  • the species is uric acid or an anion of uric acid, preferably being a substituted or unsubstituted species with the following structure:
  • the term 'substituent' is not especially limited and may be any functional group or any atom, especially any functional group or atom common in organic chemistry, provided that the ability of the species to be transported by the GLUT9 receptor is not prevented.
  • the substituent may be present at any one or more of the C or N atoms in the uric acid ring system above, provided that at least one N atom is attached to H such that the uric acid retains its ability to form an anion.
  • substituent may have any of the following meanings.
  • the substituent may comprise any organic group and/or one or more atoms from any of groups III A, IV A, VA, VIA or VIIA of the Periodic Table, such as a B, Si, N, P, O, or S atom (e.g. OH, OR, NH 2 , NHR, NR 2 , SH, SR, S0 3 H, PO4H2 etc.) or a halogen atom (e.g. F, CI, Br or I) where R is a lower hydrocarbon (1-6 C atoms) or a higher hydrocarbon (7 C atoms or more, e.g. 7-40 C atoms).
  • groups III A, IV A, VA, VIA or VIIA of the Periodic Table such as a B, Si, N, P, O, or S atom (e.g. OH, OR, NH 2 , NHR, NR 2 , SH, SR, S0 3 H, PO4H2 etc.) or
  • the organic group preferably comprises a hydrocarbon group.
  • the hydrocarbon group may comprise a straight chain, a branched chain or a cyclic group. Independently, the hydrocarbon group may comprise an aliphatic or an aromatic group. Also independently, the hydrocarbon group may comprise a saturated or unsaturated group.
  • the hydrocarbon when the hydrocarbon comprises an unsaturated group, it may comprise one or more alkene functionalities and/or one or more alkyne functionalities. When the hydrocarbon comprises a straight or branched chain group, it may comprise one or more primary, secondary and/or tertiary alkyl groups. When the hydrocarbon comprises a cyclic group it may comprise an aromatic ring, an aliphatic ring, a heterocyclic group, and/or fused ring derivatives of these groups.
  • the cyclic group may thus comprise a benzene, naphthalene, anthracene, indene, fluorene, pyridine, quinoline, thiophene, benzothiophene, furan, benzofuran, pyrrole, indole, imidazole, thiazole, and/or an oxazole group, as well as regioisomers of the above groups.
  • the number of carbon atoms in the hydrocarbon group is not especially limited, but preferably the hydrocarbon group comprises from 1-40 C atoms.
  • the hydrocarbon group may thus be a lower hydrocarbon (1-6 C atoms) or a higher hydrocarbon (7 C atoms or more, e.g. 7-40 C atoms).
  • the lower hydrocarbon group may be a methyl, ethyl, propyl, butyl, pentyl or hexyl group or regioisomers of these, such as isopropyl, isobutyl, tert-butyl, etc.
  • the number of atoms in the ring of the cyclic group is not especially limited, but preferably the ring of the cyclic group comprises from 3-10 atoms, such as 3, 4, 5, 6 or 7 atoms.
  • the groups comprising heteroatoms described above, as well as any of the other groups defined above, may comprise one or more heteroatoms from any of groups 1IIA, IVA, VA, VIA or V1IA of the Periodic Table, such as a B, Si, N, P, O, or S atom or a halogen atom (e.g. F, CI, Br or I).
  • groups 1IIA, IVA, VA, VIA or V1IA of the Periodic Table such as a B, Si, N, P, O, or S atom or a halogen atom (e.g. F, CI, Br or I).
  • the substiruent may comprise one or more of any of the common functional groups in organic chemistry, such as hydroxy groups, carboxylic acid groups, ester groups, ether groups, aldehyde groups, ketone groups, amine groups, amide groups, imine groups, thiol groups, thioether groups, sulphate groups, sulphonic acid groups, and phosphate groups etc.
  • the substituent may also comprise derivatives of these groups, such as carboxylic acid anhydrydes and carboxylic acid halides.
  • any substituent may comprise a combination of two or more of the substituents and/or functional groups defined above.
  • the cell is not especially limited, provided that it comprises a receptor with a GLUT9 function. However, in typical embodiments it is a cell capable of being used in a high-throughput cell-based assay or method, such as HEK-293 or CHO cells. It is further typical that the cell is a cell endogenously expressing GLUT9 (such as RPTEC (Renal Proximal Tubule Epithelial Cells)), or is a cell that has been transfected such that it comprises a GLUT9 receptor at its surface. In some embodiments the cell comprises an H4 cell. When transfection has been employed, typically the cell is one that has been transfected with a plasmid containing hGLUT9. Transfection is beneficial, since it may give rise to a cell overexpressing GLUT9, which may improve the efficiency of the assay.
  • RPTEC Renal Proximal Tubule Epithelial Cells
  • the kinetics measured in accordance with the present method are not especially limited, provided that they provide a measure of uptake of the species.
  • the measurement of kinetics of transport comprises a measurement of flux.
  • the species when the species is ionic, such as a urate anion, the ionic nature of the species is advantageous allowing the measurement of kinetics via changes in membrane potential.
  • the dye comprises a membrane permeable dye. It is further typical that the dye comprises a fluorescent dye.
  • the measurement of kinetics is performed using a Fluorescent Imaging Plate Reader (FLIPR ) technique.
  • FLIPR Fluorescent Imaging Plate Reader
  • the measurement of kinetics is carried out in a buffer medium.
  • the buffer medium is not especially limited, provided that it is not detrimental to the assay.
  • the buffer medium comprises choline chloride.
  • the choline chloride is present in the buffer in a concentration of from 50 mM to 200 mM, preferably from 75 mM to 175 mM, preferably from 100 mM to 150 mM and most preferably about 125 mM.
  • the buffer medium does not comprise NaCI.
  • the buffer medium further comprises one or more of: KC1, MgS0 4 , and KHP0 4 .
  • the KC1 is present in a concentration of from 2-8 mM, preferably from 3-7 mM, preferably from 4-6 mM and most preferably about 4.8 mM.
  • the MgS0 4 is present in a concentration of from 0.2-2.2 mM, preferably from 0.5-2 mM, preferably from 1-1.5 mM and most preferably about 1.2 mM.
  • the KHPO 4 is present in a concentration of from 0.2-2.2 mM, preferably from 0.5-2 mM, preferably from 1-1.5 mM and most preferably about 1.2 mM.
  • the pH of the buffer is not especially limited, provided that it is not detrimental to the assay.
  • the buffer medium pH is from 6.5 to 8.5, preferably from 7.0 to 7.7, preferably from 7.2-7.5, preferably from 7.3-7.5 and most preferably about 7.4.
  • test compound may be determined to be effective.
  • the test compound is determined to be a GLUT9 inhibitor by comparing the kinetics measurements obtained in step (b) with kinetics measurements obtained for one or more known GLUT9 inhibitors. Any such GLUT9 inhibitors may be employed, but typical GLUT9 inhibitors are selected from one or more of benzbromarone, indomethacin and oxypurinol.
  • the present invention further provides a method for screening for a GLUT9 inhibitor that inhibits uptake or release of a species into or from a cell, which method comprises:
  • the species is typically one as already defined above.
  • the uptake assay (step b) if the species is radiolabelled, preferably 14 C labelled.
  • the cell for the uptake assay comprises an oocyte. Oocytes have been found to be useful in a cell-based assay to confirm cell uptake, although are not suitable for a high-throughput cell-based assay. Therefore, in the present methods, if a small number of inhibitors are identified in a high-throughput method of the invention, this small number may be tested in a cross-check for cell uptake using the oocyte system.
  • the cell such as the oocyte
  • the cell is a cell that has been injected with GLUT9 mRNA transcript such that it comprises a GLUT9 receptor at its surface.
  • the present screening methods are suitable for screening for an agent for treating a disease associated with hyperuricemia.
  • the disease associated with hyperuricemia may be any disease as defined above, such as gout, gouty attacks, hypertension, a metabolic syndrome (such as insulin resistance, obesity, dyslipidaemia), atherosclerosis, cardiovascular disease, a kidney disease (such as acute uric acid nephropathy, chronic urate nephropathy, and uric acid nephrolithiasis (urolithiasis)), sarcoidosis, psoriasis, joint inflammation, arthritis, hyperparathyroidism, plumbism, Lesch-Nylan Syndrome and Kelley-Seegmiller Syndrome.
  • a metabolic syndrome such as insulin resistance, obesity, dyslipidaemia
  • atherosclerosis cardiovascular disease
  • a kidney disease such as acute uric acid nephropathy, chronic urate nephropathy, and uric acid nephrolithiasis (urolithia
  • test compound identified as a GLUT9 inhibitor is considered a potential agent for treating the disease in accordance with the present invention.
  • the invention provides such agents, pharmaceutical compositions for treating such a disease comprising such agents, and methods for treating such a disease, comprising administering such agents as and/or compositions to a patient.
  • the patient is not especially limited, and may be a human or an animal, although is typically a human.
  • a cell line stably expressing human GLUT9 was first generated.
  • Human H4 cells were transfected with the pReceiver-Lvl 05 plasmid containing hGLUT9 with Lipofectamine 2000.
  • hGLUT9-overexpressing polyclonal populations were isolated under purinomycin selective pressure (1 ⁇ / ⁇ !) and maintained in Dulbecco's modified Eagle's medium supplemented with 1 p- /ml purinomycin + 10% foetal bovine serum (FBS).
  • Monoclonal cell lines were then isolated and high hGLUT9-expressing clones selected following confirmation of protein expression by Western blotting (see Figure 2) and cell-surface expression by immunocytochemistry (see Figure 3).
  • H4/hGLUT9 cells (clone #10) were seeded in black, clear bottom tissue culture treated 384- well plates at l xlO 4 cells/well in DMEM containing 10% FBS and grown for 18-22 hrs in a tissue culture incubator maintained at 37°C with 5% C0 2 .
  • the media was removed from the cell assay plates and the cells loaded with 20 ⁇ /well membrane potential-sensitive blue dye (Molecular Devices LLC) dissolved at 1.2x in Assay Buffer (25 mM HEPES buffer containing 125 mM choline chloride, 4.8 mM KC1, 1.2 mM MgS0 4 , and 1.2 mM KHP0 4 at pH 7.4).
  • oocytes were injected with 50 ng of human GLUT9 mRNA transcript or with an equivalent volume of 3 ⁇ 40 and then maintained in Barth's buffer for 2 days at 18°C before use.
  • uptake buffer (10 mM HEPES, pH 7.5, 100 mM NaCl, 2 mM KC1, 1 mM CaCl 2 , MgCl 2 ) containing 30-52 ⁇ [ 14 C] -urate for 1 h.
  • Oocytes were extensively washed with ice-cold buffer.
  • oocytes were individually transferred to scintillation tubes and solubilized with 200 ⁇ of 10% (w/v) SDS. Samples were counted after addition of 2 ml scintillation fluid. Data are expressed as rate of urate uptake in picomoles per oocyte per hour calculated from the counted dpm. For urate uptake inhibition studies, compounds (at 10, 30 and 80 ⁇ final test concentrations) and oocytes were pre-incubated in uptake buffer for 30 min prior to the addition of [ ! C] -urate.
  • Small molecule inhibitors of hGLUT9 were identified by screening a diverse library of 121 ,000 samples at a final testing concentration of 10 ⁇ solubilized in 100% DMSO. 'Hits' were selected based on a triaging strategy.
  • the control inhibitors, benzbromarone, indomethacin and oxypurinol, were routinely tested on quality control plates located within each batch process in a 10-point concentration response format to monitor the overall performance of the assay.
  • hit molecules were also screened in a counter-screen assay which measured the flux of chloride anions across the plasma membrane.
  • Table 1 shows representative ⁇ 3 ⁇ 4 ⁇ data for inhibition of urate uptake in the membrane potential assay by 2 hit compounds.
  • Table 1 - Representative pICSO data for hit compound inhibition of urate uptake in H4 GLUT9 cells using the membrane potential assay.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Microbiology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

Provided is a method for screening for a GLUT9 inhibitor, which method comprises: (a) contacting a receptor having a GLUT9 function with a test compound; (b) measuring the kinetics of transport of a species across the receptor, which species is one whose transport is facilitated by GLUT9; and (c) determining whether the test compound is a GLUT9 inhibitor from the kinetics measurements taken in step (b).

Description

SCREENING METHOD FOR A GLUT9 INHIBITOR
The present invention relates to a screening method for identifying an agent that is a GLUT9 inhibitor, and/or an agent capable if inhibiting the uptake or release of a species into or from a cell via the GLUT9 receptor. Typically the species comprises a urate anion, and the method is thus in particular a method for screening for, or identifying, a medicament for treating gout and diseases associated with hyperuricemia. The invention also relates to agents, medicaments, pharmaceutical compositions and methods of treatment containing the gout treatment agents.
The poorly soluble acid, uric acid (UA), is the end-product of purine metabolism in humans and great apes, which have lost hepatic uricase activity during hominid evolution. Loss of uricase activity leads to significantly higher serum UA (sUA) in humans (-240-360 μΜ, 4-6 mg/dl) than in other mammals (-3-120 μΜ, < l -2 mg dl). Abnormally high levels of sUA (>420 μΜ, 7 mg/dl) is known as hyperuricemia and is the precursor to the deposition of UA crystals in and around the joints, which is the causative factor in the inflammatory arthritic condition gout.
New therapeutics for the treatment of gout represent an attractive proposition for a number of reasons. The incidence of gout is around 1 -2% of adults in developed countries and is increasing rapidly in developing nations. With few new drags approved in the last 40 years, and with up to 60% of patients intolerant or unresponsive to the current first-line therapy (allopurinol), gout is a disease with significant unmet medical need.
GLUT9 has been associated with gout and identified as a high capacity urate transporter (Vitart V, Rudan I, Hayward C, Gray NK, Floyd J, Palmer CNA et al. "SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout." Nature Genetics 2008; 40(4): 437-442). About 70% of daily UA excretion occurs via the kidneys, and in 5-25% of the human population, impaired renal excretion leads to hyperuricemia. Recently, a number of genome-wide association studies led to the identification of a variety of genetic loci linked with variance of sUA levels. The major genetic variant associated with low fractional excretion of UA and susceptibility to gout was found within the SLC2A9 gene, which encodes the GLUT9 transporter. GLUT9 was originally characterised as a hexose transporter. However, following the association of various SNPs in the promoter and intron regions of SLC2A9 with hyperuricemia, a number of studies have demonstrated that GLUT9 is in fact a high capacity UA transporter expressed in the renal proximal tubule. Hereditary mutations in the SLC2A9 gene lead to impaired GLUT9 function and renal hypouricemia, which is characterised by impaired renal reabsorption of UA and subsequent low sUA levels.
In addition to the blockade of UA production through the use of xanthine oxidase inhibitors such as allopurinol, gout therapies also target the reabsorption of UA in the kidney by blocking UA transporters. Such therapeutics are termed 'uricosurics' and they increase the excretion of UA into the urine. However, existing uricosuric drags such as probenecid and benzbromarone are of limited use due to side effect issues, low potency, and lack of selectivity.
Hence, inhibition of UA reabsorption by GLUT9 represents an attractive disease-modifying opportunity for gout. The identification of novel, selective GLUT9 inhibitors may act as powerful uricosuric agents with enhanced efficacy and safety over existing therapies.
However, despite the potential benefits of a selective GLUT9 inhibitor, screening for such inhibitors has proven problematic. A typical assay to screen for inhibitors for such receptors is an uptake assay. In an uptake assay, transfected cells comprising the necessary receptors are challenged with a potential inhibitor and then the uptake of a relevant transported species is measured (Anzai N, Ichida K, Jutabha P, Kimura T, Babu E, Jin CJ et al. "Plasma Urate Level Is Directly Regulated by a Voltage-driven Urate Efflux Transporter URATv 1 (SLC2A9) in Humans." Journal of Biological Chemistry 2008; 283(40): 26834-26838). Typically radiolabeled species are employed to facilitate measurement. However, such uptake assays for GLUT9 in cell systems other than Xenopus laevis oocytes injected to express GLUT9 have not been unequivocally demonstrated. The reasons for this are not yet clearly understood. Consequently, no commercial high-throughput cell uptake assay for GLUT9 has been developed. Cell uptake assays using oocytes comprising the GLUT9 receptor have shown some effectiveness on a small scale (Anzai N, Ichida K, Jutabha P, Kimura T, Babu E, Jin CJ et al. "Plasma Urate Level Is Directly Regulated by a Voltage- driven Urate Efflux Transporter URATvl (SLC2A9) in Humans." Journal of Biological Chemistry 2008; 283(40): 26834-26838; Caulfield MJ, Munroe PB, O'Neill D, Witkowska K, Charchar FJ, Doblado M et al. "SLC2A9 is a high-capacity urate transporter in humans." PLoS Med 2008; 5(10): el 97; Vitart V, Rudan I, Hayward C, Gray NK, Floyd J, Palmer CNA et al. "SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout." Nature Genetics 2008; 40(4): 437-442; Bibert S, Hess SK, Firsov D, Thorens B, Geering K, Horisberger JD et al. "Mouse GLUT9: evidences for a urate uniporter." Am J Physiol Renal Physiol 2009; 297(3): F612-9). However, oocytes are not suitable for high-throughput screening methods. The cells are larger than regular cells and thus require more space, and are unreliable (as many as 10 cells may be required per test well before a reliable result may be expected). This is both because the cells are large, and also because viability may not be immediately apparent. Consequently tests need to be undertaken on both viable and unviable cells, which can significantly reduce efficiency. Thus, there is still a need for a high-throughput cell-based screening assay for identifying GLUT9 inhibitors.
There is a body of literature related to the imaging and use of fluorescent probes to measure activity of biological targets. In US 6,727,071 Cellomics describe an optical imaging system for analysing cells containing fluorescently labelled molecules. In US 2009/226931 Corning describe a label-free biosensor assay system for use in cell -based compound screening. However, methodologies related to changes in membrane potential, or the application of methodologies for screening for GLUT9 inhibitors are not studied, or considered, in any of these references.
There is also established literature investigating the role of GLUT9 as a uric acid transporter. Mueckler and Thorens (Molecular Aspects of Medicine, 2013, 34, 121-138) provide a review of the whole GLUT family of membrane transporters. In WO 2009/063200, Wright et al. describe the identification of GLUT9 as a uric acid transporter. JP 2009294057 from J. Pharma. describes an expression system for measuring uric acid flux across cells comprising expression of the uric acid transporters URATl and GLUT9. However, none of these applications refer to the electrogenic nature of uric acid transport by GLUT9 nor do they allude to the use of membrane potential sensitive technologies for screening for GLUT9 inhibitors.
Witkowska et al (Biochemistry and Cell Biology, 201 1, 89, 283) describe the electrogenic nature of urate uptake by GLUT9. This publication does not link the electrogenic nature of urate uptake with the use of membrane potential-sensitive dyes to provide a kinetic methodology for high-throughput screening for GLUT9 inhibitors.
Accordingly, it is an aim of the present invention to provide a high-throughput (preferably cell-based) screening assay for identifying GLUT9 inhibitors. It is a further aim of the present invention to provide a high-throughput (preferably cell-based) screening method for identifying agents suitable for treating diseases associated with hyperuricemia, which may include but are not limited to: gout, gouty attacks, hypertension, a metabolic syndrome (such as insulin resistance, obesity, dyslipidaemia), atherosclerosis, cardiovascular disease, a kidney disease (such as acute uric acid nephropathy, chronic urate nephropathy, and uric acid nephrolithiasis (urolithiasis)), sarcoidosisis, psoriasis, joint inflammation, arthritis, hyperparathyroidism, plumbism, Lesch-Nylan Syndrome and elley-Seegmiller Syndrome . It is a still further aim of the invention to provide agents, medicaments and pharmaceuticals for treating such diseases, and methods for treating such diseases using the agents, medicaments and/or pharmaceuticals.
Accordingly, the present invention provides a method for screening for a GLUT9 inhibitor, which method comprises:
(a) contacting a receptor having a GLUT9 function with a test compound;
(b) measuring the kinetics of transport of a species across the receptor, which species is one whose transport is facilitated by GLUT9; and
(c) determining whether the test compound is a GLUT9 inhibitor from the kinetics measurements taken in step (b).
In the present context, the receptor having a GLUT9 function may be any receptor (or a receptor mimic or analogue) having the same or substantially the same function as GLUT9. It may therefore be any receptor capable of transporting the same species, substantially the same species (such as salts, ions, solvates or the like), or similar species (such as substituted versions of the species) as transported by GLUT9. It thus extends to any receptor encoded by the SCL2A9 gene, including GLUT9, UAQTL2, vURATl and URATvl . Many of these are alternative names for GLUT9, or have only minor differences that do not alter function, such as SNPs in the gene sequence. Thus, this definition extends to any such receptor (or a receptor mimic or analogue) capable of facilitating urate transport across a cell membrane.
In the present context, a test compound (the potential GLUT9 inhibitor) comprises any compound, complex, agent or the like which has potential as an inhibitor of GLUT9 (and/or an inhibitor of the receptors as defined above). Thus, the test compound may be a small molecule, such as an organic or inorganic compound, a large molecule such as an oligomer or polymer, or a biological molecule such as a peptide, polypeptide, protein, nucleic acid or the like. The compound may be present as a complex, salt, hydrate or other derivative for facilitating solubility, stability, uptake, bioavailability or the like. In this context, inhibitor means any compound capable of impairing or detrimentally affecting the GLUT9 function of the receptor in any way (such as preventing, reducing, reversing or diverting the transport of the species across the receptor), including impairing or detrimentally affecting the function of any of the receptors as defined above.
In the present context the species is any species which is capable of being transported across the GLUT9 receptor. Typically, therefore it is a species which may be transported into or out of a cell (i.e. across the cell membrane) by virtue of the GLUT9 receptor. Thus, the species is typically a urate anion, but may be a derivative thereof, such as an organic or inorganic compound, a large molecule such as an oligomer or polymer, or a biological molecule such as a peptide, polypeptide, protein, nucleic acid or the like. In particular the species may be a sugar, such as glucose or fructose, or a sugar derivative, and indeed may be any sugar capable of being transported across the GLUT9 receptor. The species may be present as a complex, salt, hydrate or other derivative for facilitating solubility, stability, uptake, bioavailability or the like.
The present inventors have discovered that measurement of kinetics provides an accurate measurement of whether a test compound is a suitable GLUT9 inhibitor. Unlike known uptake assays, which cannot be developed in a manner consistent with high-throughput screening, the kinetics measurements have surprisingly been found to provide reliable information on the throughput of species into and out of the cell, despite the cell uptake data indicating no such transport in many instances. In the present context, the measurement of kinetics of transport means the successive measurement over time of any parameter which could be related to transport. This could be as few as two measurements, each at a different point in time, but typically involves many such measurements. Typically, the kinetics measurements enable a point of comparison, or reference point, to be identified, such that the effect of different inhibitory compounds can more reliably be compared. For example, if the parameter being measured is membrane potential (which is related to the transport of species across the cell membrane) that reference point is typically the maximum change in membrane potential (see Figure 4). The change (reduction) in this maximum is indicative of an inhibitory effect on transport across the membrane, and the ability to accurately pinpoint this maximum provides reliability of comparison of inhibitory compounds that could not otherwise be achieved.
Further provided by the invention is a method for screening for a GLUT9 inhibitor that inhibits uptake or release of a species into or out of a cell, which method comprises:
(a) screening a test compound for a GLUT9 inhibitor according to any of the methods described above; and
(b) performing an assay to determine whether the test compound inhibits the uptake or release of the species into or out of the cell, which species is one whose transport is facilitated by GLUT9.
The invention also provides method for screening for an agent for treating a disease associated with hyperuricemia which method comprises a method as defined above. The disease associated with hyperuricemia is not especially limited, and may be any such disease. This may include, but is not limited to the following: gout, gouty attacks, hypertension, a metabolic syndrome (such as insulin resistance, obesity, dyslipidaemia), atherosclerosis, cardiovascular disease, a kidney disease (such as acute uric acid nephropathy, chronic urate nephropathy, and uric acid nephrolithiasis (urolithiasis)), sarcoidosisis, psoriasis, joint inflammation, arthritis, hyperparathyroidism, plumbism, Lesch-Nylan Syndrome and Kelley- Seegmiller Syndrome,
Further provided is an agent for treating a disease associated with hyperaricemia, which agent comprises an agent obtainable from a method as defined above. The disease associated with hyperaricemia is not especially limited, and may be any such disease as defined above.
Still further provided is a pharmaceutical composition for treating a disease associated with hyperaricemia, which composition comprises a compound as defined above. The disease associated with hyperaricemia is not especially limited, and may be any such disease, as defined above.
Yet further provided is a method for treating a disease associated with hyperaricemia, which method comprises administering a agent as defined above and/or a composition as defined above to a patient. The disease associated with hyperuricemia is not especially limited, and may be any such disease, as defined above.
The invention will now be explained in more detail, by way of example only, with reference to the following Figures.
Figure 1 shows an overview of FLIPR™ membrane potential dye kit technology (Molecular Devices LLC).
Figure 2 shows a Western blot of crude cell lysates from H4 cells transfected to express hGLUT9. High levels of expression are confirmed in clone #10.
Figure 3 shows immunocytochemistry demonstrating cell surface expression of hGLUT9 in H4 cells.
Figure 4 shows that membrane potential changes are increased by replacement of NaCl with choline chloride in both the dye loading and urate stimulation buffers. Figure 5 shows representative concentration-response curve data for pharmacological reference compounds in the membrane potential assay.
Figure 6 shows representative inhibition data for two hit molecules in an orthogonal oocyte uptake assay.
The invention will now be described in more detail.
As has been described, the invention relates to a method for screening for a GLUT9 inhibitor, which method comprises:
(a) contacting a receptor having a GLUT9 function with a test compound;
(b) measuring the kinetics of transport of a species across the receptor, which species is one whose transport is facilitated by GLUT9; and
(c) determining whether the test compound is a GLUT9 inhibitor from the kinetics measurements taken in step (b).
In the present context, the receptor having a GLUT9 function may be any receptor (or a receptor mimic or analogue) having the same or substantially the same function as GLUT9. It may therefore be any receptor capable of transporting the same species, substantially the same species (such as salts, ions, solvates or the like), or similar species (such as substituted versions of the species) as transported by GLUT9. It thus extends to any receptor encoded by the SCL2A9 gene, including GLUT9, UAQTL2, vURATl and URATvl . Many of these are alternative names for GLUT9, or have only minor differences that do not alter function, such as SNPs in the gene sequence. Thus, this definition extends to any such receptor capable of facilitating urate transport across a cell membrane.
The test compound (the GLUT9 inhibitor) is not especially limited, and may comprise any compound, complex, agent or the like which has potential as an inhibitor of GLUT9 (and/or an inhibitor of the receptor as defined above). Thus, the test compound may be a small molecule, such as an organic or inorganic compound, a large molecule such as an oligomer or polymer, or a biological molecule such as a peptide, polypeptide, protein, nucleic acid or the like. The compound may be present as a complex, salt, hydrate or other derivative for facilitating solubility, stability, uptake, bioavailability or the like. In this context, inhibitor means any compound capable of impairing or detrimentally affecting the GLUT9 function of the receptor in any way (such as preventing, reducing, reversing or diverting the transport of the species across the receptor), including impairing or detrimentally affecting the function of any of the receptors as defined above.
The species is not especially limited, and may be any species which is capable of being transported across the GLUT9 receptor. Typically, therefore it is a species which may be transported into or out of a cell (i.e. across the cell membrane) by virtue of the GLUT9 receptor. Thus, the species is typically a urate anion, but may be a derivative thereof, such as an organic or inorganic compound, a large molecule such as an oligomer or polymer, or a biological molecule such as a peptide, polypeptide, protein, nucleic acid or the like. In particular the species may be a sugar, such as glucose or fructose, or a sugar derivative, and indeed may be any sugar capable of being transported across the GLUT9 receptor. The species may be present as a complex, salt, hydrate or other derivative for facilitating solubility, stability, uptake, bioavailability or the like.
In a typical embodiment, the method preferably comprises the following:
(a) contacting a cell comprising a GLUT9 receptor with a test compound;
(b) measuring the kinetics of transport of a species across a membrane of the cell, which species is one whose transport across the membrane is facilitated by GLUT9; and
(c) determining whether the test compound is a GLUT9 inhibitor from the kinetics measurements taken in step (b).
Thus, in these typical embodiments, the receptor having the GLUT9 function is a GLUT9 receptor. In other embodiments, the receptor may be an artificially modified receptor, a receptor mimic or the like, provided that it retains the GLUT9 receptor function, and in particular its solute carrying function. In addition, the kinetics are typically measured across a cell membrane so as to representative of the transport of the species into and out of the cell itself. However, in other embodiments, the system may be an artificial system modelling transport across a cell membrane, and need (in that case) not involve the cell itself. The species is not especially limited, provided that its transport into and out of a cell is capable of being facilitated by the GLUT9 receptor. The species is typically selected from a substituted or unsubstituted uric acid compound, a substituted or unsubstituted urate compound, and a mixture of two or more of the above. In the more preferred embodiments, the species comprises a substituted or unsubstituted urate anion.
Thus, typically the species is uric acid or an anion of uric acid, preferably being a substituted or unsubstituted species with the following structure:
Figure imgf000011_0001
In all of the embodiments mentioned in connection with this invention, both above and in the following, the term 'substituent' is not especially limited and may be any functional group or any atom, especially any functional group or atom common in organic chemistry, provided that the ability of the species to be transported by the GLUT9 receptor is not prevented.
The substituent may be present at any one or more of the C or N atoms in the uric acid ring system above, provided that at least one N atom is attached to H such that the uric acid retains its ability to form an anion.
Thus, substituent may have any of the following meanings. The substituent may comprise any organic group and/or one or more atoms from any of groups III A, IV A, VA, VIA or VIIA of the Periodic Table, such as a B, Si, N, P, O, or S atom (e.g. OH, OR, NH2, NHR, NR2, SH, SR, S03H, PO4H2 etc.) or a halogen atom (e.g. F, CI, Br or I) where R is a lower hydrocarbon (1-6 C atoms) or a higher hydrocarbon (7 C atoms or more, e.g. 7-40 C atoms).
When the substiruent comprises an organic group, the organic group preferably comprises a hydrocarbon group. The hydrocarbon group may comprise a straight chain, a branched chain or a cyclic group. Independently, the hydrocarbon group may comprise an aliphatic or an aromatic group. Also independently, the hydrocarbon group may comprise a saturated or unsaturated group.
When the hydrocarbon comprises an unsaturated group, it may comprise one or more alkene functionalities and/or one or more alkyne functionalities. When the hydrocarbon comprises a straight or branched chain group, it may comprise one or more primary, secondary and/or tertiary alkyl groups. When the hydrocarbon comprises a cyclic group it may comprise an aromatic ring, an aliphatic ring, a heterocyclic group, and/or fused ring derivatives of these groups. The cyclic group may thus comprise a benzene, naphthalene, anthracene, indene, fluorene, pyridine, quinoline, thiophene, benzothiophene, furan, benzofuran, pyrrole, indole, imidazole, thiazole, and/or an oxazole group, as well as regioisomers of the above groups.
The number of carbon atoms in the hydrocarbon group is not especially limited, but preferably the hydrocarbon group comprises from 1-40 C atoms. The hydrocarbon group may thus be a lower hydrocarbon (1-6 C atoms) or a higher hydrocarbon (7 C atoms or more, e.g. 7-40 C atoms). The lower hydrocarbon group may be a methyl, ethyl, propyl, butyl, pentyl or hexyl group or regioisomers of these, such as isopropyl, isobutyl, tert-butyl, etc. The number of atoms in the ring of the cyclic group is not especially limited, but preferably the ring of the cyclic group comprises from 3-10 atoms, such as 3, 4, 5, 6 or 7 atoms.
The groups comprising heteroatoms described above, as well as any of the other groups defined above, may comprise one or more heteroatoms from any of groups 1IIA, IVA, VA, VIA or V1IA of the Periodic Table, such as a B, Si, N, P, O, or S atom or a halogen atom (e.g. F, CI, Br or I). Thus the substiruent may comprise one or more of any of the common functional groups in organic chemistry, such as hydroxy groups, carboxylic acid groups, ester groups, ether groups, aldehyde groups, ketone groups, amine groups, amide groups, imine groups, thiol groups, thioether groups, sulphate groups, sulphonic acid groups, and phosphate groups etc. The substituent may also comprise derivatives of these groups, such as carboxylic acid anhydrydes and carboxylic acid halides.
In addition, any substituent may comprise a combination of two or more of the substituents and/or functional groups defined above.
The cell is not especially limited, provided that it comprises a receptor with a GLUT9 function. However, in typical embodiments it is a cell capable of being used in a high-throughput cell-based assay or method, such as HEK-293 or CHO cells. It is further typical that the cell is a cell endogenously expressing GLUT9 (such as RPTEC (Renal Proximal Tubule Epithelial Cells)), or is a cell that has been transfected such that it comprises a GLUT9 receptor at its surface. In some embodiments the cell comprises an H4 cell. When transfection has been employed, typically the cell is one that has been transfected with a plasmid containing hGLUT9. Transfection is beneficial, since it may give rise to a cell overexpressing GLUT9, which may improve the efficiency of the assay.
The kinetics measured in accordance with the present method are not especially limited, provided that they provide a measure of uptake of the species. Typically, however, the measurement of kinetics of transport comprises a measurement of flux. In some embodiments, when the species is ionic, such as a urate anion, the ionic nature of the species is advantageous allowing the measurement of kinetics via changes in membrane potential. In such embodiments, it is especially beneficial to employ a dye to facilitate the measurement. Typically the dye comprises a membrane permeable dye. It is further typical that the dye comprises a fluorescent dye. In especially favourable embodiments, the measurement of kinetics is performed using a Fluorescent Imaging Plate Reader (FLIPR ) technique.
In typical embodiments, especially those employing cells, the measurement of kinetics is carried out in a buffer medium. The buffer medium is not especially limited, provided that it is not detrimental to the assay. Typically the buffer medium comprises choline chloride. In such embodiments, the choline chloride is present in the buffer in a concentration of from 50 mM to 200 mM, preferably from 75 mM to 175 mM, preferably from 100 mM to 150 mM and most preferably about 125 mM. In further preferred embodiments, the buffer medium does not comprise NaCI. In other embodiments, the buffer medium further comprises one or more of: KC1, MgS04, and KHP04. Typically, the KC1 is present in a concentration of from 2-8 mM, preferably from 3-7 mM, preferably from 4-6 mM and most preferably about 4.8 mM. Typically the MgS04 is present in a concentration of from 0.2-2.2 mM, preferably from 0.5-2 mM, preferably from 1-1.5 mM and most preferably about 1.2 mM. Typically the KHPO4 is present in a concentration of from 0.2-2.2 mM, preferably from 0.5-2 mM, preferably from 1-1.5 mM and most preferably about 1.2 mM. The pH of the buffer is not especially limited, provided that it is not detrimental to the assay. Advantageously, the buffer medium pH is from 6.5 to 8.5, preferably from 7.0 to 7.7, preferably from 7.2-7.5, preferably from 7.3-7.5 and most preferably about 7.4.
It is not especially limited how the test compound may be determined to be effective. However, typically the test compound is determined to be a GLUT9 inhibitor by comparing the kinetics measurements obtained in step (b) with kinetics measurements obtained for one or more known GLUT9 inhibitors. Any such GLUT9 inhibitors may be employed, but typical GLUT9 inhibitors are selected from one or more of benzbromarone, indomethacin and oxypurinol.
As has been mentioned, the present invention further provides a method for screening for a GLUT9 inhibitor that inhibits uptake or release of a species into or from a cell, which method comprises:
(a) screening a test compound for a GLUT9 inhibitor according to any of the methods described above; and
(b) performing an assay to determine whether the test compound inhibits the uptake or release of the species into or from the cell, which species is one whose transport is facilitated by GLUT9.
In this method, the species is typically one as already defined above. However, in this method it is advantageous for the uptake assay (step b) if the species is radiolabelled, preferably 14C labelled. In typical embodiments, the cell for the uptake assay comprises an oocyte. Oocytes have been found to be useful in a cell-based assay to confirm cell uptake, although are not suitable for a high-throughput cell-based assay. Therefore, in the present methods, if a small number of inhibitors are identified in a high-throughput method of the invention, this small number may be tested in a cross-check for cell uptake using the oocyte system.
Typically the cell, such as the oocyte, is a cell that has been injected with GLUT9 mRNA transcript such that it comprises a GLUT9 receptor at its surface.
As has been mentioned, the present screening methods are suitable for screening for an agent for treating a disease associated with hyperuricemia. The disease associated with hyperuricemia may be any disease as defined above, such as gout, gouty attacks, hypertension, a metabolic syndrome (such as insulin resistance, obesity, dyslipidaemia), atherosclerosis, cardiovascular disease, a kidney disease (such as acute uric acid nephropathy, chronic urate nephropathy, and uric acid nephrolithiasis (urolithiasis)), sarcoidosis, psoriasis, joint inflammation, arthritis, hyperparathyroidism, plumbism, Lesch-Nylan Syndrome and Kelley-Seegmiller Syndrome. Any test compound identified as a GLUT9 inhibitor is considered a potential agent for treating the disease in accordance with the present invention. Thus the invention provides such agents, pharmaceutical compositions for treating such a disease comprising such agents, and methods for treating such a disease, comprising administering such agents as and/or compositions to a patient. The patient is not especially limited, and may be a human or an animal, although is typically a human.
The invention will now be described in more detail, by way of example only, with reference to the following specific embodiments.
EXAMPLES
In order to identify inhibitors of GLUT9, a functional cell-based assay was developed that measures changes in membrane potential in response to transport of urate anions across the plasma membrane. For this purpose the Fluorescent Imaging Plate Reader (FLIPR™) is a routinely used platform for real-time measurement of ion flux across cell membranes with appropriate fluorescent dyes. A subset of such membrane-permeable dyes rapidly redistribute across the membrane in response to changing membrane potential. Hence, a fluorescent read-out provides a measure of changes to membrane potential following addition of a suitable electrogenic agent such as an anion. Consequently, this assay provides the opportunity to measure the effects of test agents which modulate ion flux across the membrane. An overview of this technology is depicted in Figure 1.
Methods
Stable cell line generation
To support assay development, a cell line stably expressing human GLUT9 was first generated. Human H4 cells were transfected with the pReceiver-Lvl 05 plasmid containing hGLUT9 with Lipofectamine 2000. hGLUT9-overexpressing polyclonal populations were isolated under purinomycin selective pressure (1 μ§/ηι!) and maintained in Dulbecco's modified Eagle's medium supplemented with 1 p- /ml purinomycin + 10% foetal bovine serum (FBS). Monoclonal cell lines were then isolated and high hGLUT9-expressing clones selected following confirmation of protein expression by Western blotting (see Figure 2) and cell-surface expression by immunocytochemistry (see Figure 3).
H4/hGLUT9 cells (clone #10) were seeded in black, clear bottom tissue culture treated 384- well plates at l xlO4 cells/well in DMEM containing 10% FBS and grown for 18-22 hrs in a tissue culture incubator maintained at 37°C with 5% C02. In order to measure membrane potential, the media was removed from the cell assay plates and the cells loaded with 20 μΐ/well membrane potential-sensitive blue dye (Molecular Devices LLC) dissolved at 1.2x in Assay Buffer (25 mM HEPES buffer containing 125 mM choline chloride, 4.8 mM KC1, 1.2 mM MgS04, and 1.2 mM KHP04 at pH 7.4). After a 30 min pre-incubation at room temperature, the assay plates were delivered by robotic arm to the FLIPR™ tetra for real-time imaging of the stimulation with 2.5 mM urate (final) prepared in assay buffer. During assay development, it was found that replacement of NaCl in the assay buffer during both dye loading and urate stimulation phases significantly improved the assay window and also that of urate solubility (see Figure 4). In order to pharmacologically validate the assay a number of small molecule agents previously shown to inhibit urate-mediated uptake via GLUT9 were tested. In these instances, compounds were added to the cells simultaneously with the blue dye and incubated for 30 min prior to the addition of urate. Example data using reference compounds is shown in Figure 5. In all cases, IC50 values generated were in agreement and rank order with literature reports.
Expression of GLUT9 and [14CJ -urate uptake in Xenop s laevis oocytes
Adult female X. laevis oocytes were injected with 50 ng of human GLUT9 mRNA transcript or with an equivalent volume of ¾0 and then maintained in Barth's buffer for 2 days at 18°C before use. To determine urate uptake, oocytes were incubated at 18°C in uptake buffer (10 mM HEPES, pH 7.5, 100 mM NaCl, 2 mM KC1, 1 mM CaCl2, MgCl2) containing 30-52 μΜ [14C] -urate for 1 h. Oocytes were extensively washed with ice-cold buffer. After washing, oocytes were individually transferred to scintillation tubes and solubilized with 200 μΐ of 10% (w/v) SDS. Samples were counted after addition of 2 ml scintillation fluid. Data are expressed as rate of urate uptake in picomoles per oocyte per hour calculated from the counted dpm. For urate uptake inhibition studies, compounds (at 10, 30 and 80 μΜ final test concentrations) and oocytes were pre-incubated in uptake buffer for 30 min prior to the addition of [! C] -urate.
High-throughput Screening and Hit Identification
Small molecule inhibitors of hGLUT9 were identified by screening a diverse library of 121 ,000 samples at a final testing concentration of 10 μΜ solubilized in 100% DMSO. 'Hits' were selected based on a triaging strategy. The control inhibitors, benzbromarone, indomethacin and oxypurinol, were routinely tested on quality control plates located within each batch process in a 10-point concentration response format to monitor the overall performance of the assay. In order to remove 'false positive' or 'technology artefact' hits, hit molecules were also screened in a counter-screen assay which measured the flux of chloride anions across the plasma membrane. Table 1 shows representative Κ¾ο data for inhibition of urate uptake in the membrane potential assay by 2 hit compounds. Table 1 - Representative pICSO data for hit compound inhibition of urate uptake in H4 GLUT9 cells using the membrane potential assay.
Figure imgf000018_0001
Hit confirmation
Selected hit compounds were then profiled in the orthogonal in vitro assay system measuring the inhibition of [ l4C] -urate uptake in X. laevis oocytes injected to express human GLUT9. Figure 7 shows representative inhibition data for 2 hit molecules.
The data clearly show that not only does the assay provide a suitable assay for a GLUT9 inhibitor, which had not been possible previously, but that the assay is predictive for inhibiting urate uptake in cells, and hence a viable screening method for gout treatments.

Claims

CLAIMS:
1. A method for screening for a GLUT9 inhibitor, which method comprises:
(a) contacting a receptor having a GLUT9 function with a test compound;
(b) measuring the kinetics of transport of a species across the receptor, which species is one whose transport is facilitated by GLUT9; and
(c) determining whether the test compound is a GLUT9 inhibitor from the kinetics measurements taken in step (b).
2. A method according to claim 1, which method comprises:
(a) contacting a cell comprising a GLUT9 receptor with a test compound;
(b) measuring the kinetics of transport of a species across a membrane of the cell, which species is one whose transport across the membrane is facilitated by GLUT9; and
(c) determining whether the test compound is a GLUT9 inhibitor from the kinetics measurements taken in step (b).
3. A method according to claim 1 or claim 2, wherein the species is selected from a substituted or unsubstituted uric acid compound, a substituted or unsubstituted urate compound, and a mixture of two or more of the above.
4. A method according to claim 3, wherein the species comprises a substituted or unsubstituted urate anion.
5. A method according to any of claims 2-4, wherein the cell is a cell endogenously expressing GLUT9, or is a cell that has been transfected such that it comprises a GLUT9 receptor at its surface.
6. A method according to claim 5, wherein the cell comprises an H4 cell.
7. A method according to claim 5 or claim 6 wherein the cell is one that has been transfected with a plasmid containing hGLUT9.
8. A method according to any of claims 5-7 wherein the cell is a cell overexpressing GLUT9.
9. A method according to any preceding claim, wherein the measurement of kinetics of transport comprises a measurement of flux.
10. A method according to any preceding claim, wherein the measurement of kinetics comprises measurement of changes in membrane potential.
1 1. A method according to claim 8 or claim 9, wherein a dye is employed to facilitate the measurement of kinetics.
12. A method according to claim 1 1 , wherein the dye comprises a membrane permeable dye.
13. A method according to claim 1 1 or claim 12, wherein the dye comprises a fluorescent dye.
14. A method according to any of claims 11-13, wherein the measurement of kinetics is performed using a Fluorescent Imaging Plate Reader technique.
15. A method according to any preceding claim, wherein the measurement of kinetics is carried out in a buffer medium.
16. A method according to claim 15, wherein the buffer medium comprises choline chloride.
17. A method according claim 16, wherein the choline chloride is present in the buffer in a concentration of from 50 mM to 200 mM, preferably from 75 mM to 175 mM, preferably from 100 mM to 150 mM and most preferably about 125 mM.
18. A method according to any of claims 15-17 wherein the buffer medium does not comprise NaCL
19. A method according to any of claims 15-18 wherein the buffer medium further comprises one or more of: KC1, MgS0 , and KHPO4.
20. A method according to claim 19, wherein the KC1 is present in a concentration of from 2-8 mM, preferably from 3-7 mM, preferably from 4-6 mM and most preferably about 4.8 mM.
21. A method according to claim 19 or claim 20, wherein the MgS04 is present in a concentration of from 0.2-2.2 mM, preferably from 0.5-2 mM, preferably from 1 -1.5 mM and most preferably about 1.2 mM.
22. A method according to any of claims 19-21 , wherein the HPO4 is present in a concentration of from 0.2-2.2 mM, preferably from 0.5-2 mM, preferably from 1-1.5 mM and most preferably about 1.2 mM.
23. A method according to any of claims 15-22, wherein the buffer medium pH is from 6.5 to 8.5, preferably from 7.0 to 7.7, preferably from 7.2-7.5, preferably from 7.3-7.5 and most preferably about 7.4.
24. A method according to any preceding claim, wherein the test compound is determined to be a GLUT9 inhibitor by comparing the kinetics measurements obtained in step (b) with kinetics measurements obtained for one or more known GLUT9 inhibitors.
25. A method according to claim 24, wherein the one or more known GLUT9 inhibitors are selected from one or more of benzbromarone, indomethacin and oxypurinol.
26. A method for screening for a GLUT9 inhibitor that inhibits uptake or release of a species into or from a cell, which method comprises:
(a) screening a test compound for a GLUT9 inhibitor according to any of claims 1 -25; and
(b) performing an assay to determine whether the test compound inhibits the uptake or release of the species into or from the cell, which species is one whose transport is facilitated by GLUT9.
27. A method according to claim 26, wherein the species is selected from a substituted or unsubstituted uric acid compound, a substituted or unsubstituted urate compound, and a mixture of two or more of the above.
28. A method according to claim 27, wherein the species comprises a substituted or unsubstituted urate anion.
29. A method according to any of claims 26-28, wherein the species is radiolabeled, preferably 14C labelled.
30. A method according to any of claims 26-29, wherein the cell comprises an oocyte.
31. A method according to any of claims 26-30, wherein the cell is a cell that has been injected with GLUT9 mRNA transcript such that it comprises a GLUT9 receptor at its surface.
32. A method for screening for an agent for treating a disease associated with hyperuricemia, which method comprises a method as defined in any of claims 1-31.
33. A method according to claim 32, wherein the disease comprises one or more diseases selected from: gout, gouty attacks, hypertension, a metabolic syndrome (such as insulin resistance, obesity, dyslipidaemia), atherosclerosis, cardiovascular disease, a kidney disease (such as acute uric acid nephropathy, chronic urate nephropathy, and uric acid nephrolithiasis (urolithiasis)), sarcoidosis, psoriasis, joint inflammation, arthritis, hyperparathyroidism, plumbism, Lesch-Nylan Syndrome and Kelley-Seegmiller Syndrome.
34. An agent for treating a disease associated with hyperuricemia, which agent comprises an agent obtainable from a method as defined in claim 32 or claim 33.
35. A pharmaceutical composition for treating a disease associated with hyperuricemia, which composition comprises a compound as defined in claim 34.
36. A method for treating a disease associated with hyperuricemia, which method comprises administering an agent as defined in claim 34 and/or a composition as defined in claim 35 to a patient.
37. A method according to claim 36, wherein the patient is human.
PCT/EP2014/061212 2013-06-03 2014-05-29 Screening method for a glut9 inhibitor WO2014195228A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB201309876A GB201309876D0 (en) 2013-06-03 2013-06-03 Screening method
GB1309876.9 2013-06-03

Publications (2)

Publication Number Publication Date
WO2014195228A1 true WO2014195228A1 (en) 2014-12-11
WO2014195228A8 WO2014195228A8 (en) 2015-03-12

Family

ID=48805658

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2014/061212 WO2014195228A1 (en) 2013-06-03 2014-05-29 Screening method for a glut9 inhibitor

Country Status (2)

Country Link
GB (1) GB201309876D0 (en)
WO (1) WO2014195228A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009063200A2 (en) * 2007-11-15 2009-05-22 Medical Research Council Solute carrier
JP2009294057A (en) * 2008-06-04 2009-12-17 J-Pharma Co Ltd Renal urate transporter

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009063200A2 (en) * 2007-11-15 2009-05-22 Medical Research Council Solute carrier
JP2009294057A (en) * 2008-06-04 2009-12-17 J-Pharma Co Ltd Renal urate transporter

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
A MASBERNARD ET AL: "Ten years' experience with benzbromarone in the management of gout and hyperuricaemia", SOUTH AFRICAN MEDICAL JOURNAL = SUID-AFRIKAANSE TYDSKRIF VIR GENEESKUNDE, 9 May 1981 (1981-05-09), SOUTH AFRICA, pages 701, XP055136813, Retrieved from the Internet <URL:http://www.ncbi.nlm.nih.gov/pubmed/7221794> [retrieved on 20140827] *
K. WITKOWSKA ET AL: "Human SLC2A9a and SLC2A9b isoforms mediate electrogenic transport of urate with different characteristics in the presence of hexoses", AMERICAN JOURNAL OF PHYSIOLOGY. RENAL PHYSIOLOGY, vol. 303, no. 4, 30 May 2012 (2012-05-30), pages F527 - F539, XP055136586, ISSN: 1931-857X, DOI: 10.1152/ajprenal.00134.2012 *
N. ANZAI ET AL: "Plasma Urate Level Is Directly Regulated by a Voltage-driven Urate Efflux Transporter URATv1 (SLC2A9) in Humans", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 283, no. 40, 13 August 2008 (2008-08-13), pages 26834 - 26838, XP055136574, ISSN: 0021-9258, DOI: 10.1074/jbc.C800156200 *
S. BIBERT ET AL: "Mouse GLUT9: evidences for a urate uniporter", AJP: RENAL PHYSIOLOGY, vol. 297, no. 3, 8 July 2009 (2009-07-08), pages F612 - F619, XP055136576, ISSN: 0363-6127, DOI: 10.1152/ajprenal.00139.2009 *

Also Published As

Publication number Publication date
GB201309876D0 (en) 2013-07-17
WO2014195228A8 (en) 2015-03-12

Similar Documents

Publication Publication Date Title
Goytain et al. Identification and characterization of a novel mammalian Mg 2+ transporter with channel-like properties
US20210393557A1 (en) Regulators of the Endoplasmic Reticulum Proteostasis Network
Avasthi et al. Stages of ciliogenesis and regulation of ciliary length
Butterworth et al. Regulation of the epithelial sodium channel by membrane trafficking
JP6025758B2 (en) Regulatory factors of NFAT
Pramod et al. SLC6 transporters: structure, function, regulation, disease association and therapeutics
Nies et al. Expression of organic cation transporters OCT1 (SLC22A1) and OCT3 (SLC22A3) is affected by genetic factors and cholestasis in human liver
Iftinca et al. Regulation of T-type calcium channels by Rho-associated kinase
Gupta-Agarwal et al. NMDA receptor-and ERK-dependent histone methylation changes in the lateral amygdala bidirectionally regulate fear memory formation
Hwang et al. Methylsulfonylnitrobenzoates, a new class of irreversible inhibitors of the interaction of the thyroid hormone receptor and its obligate coactivators that functionally antagonizes thyroid hormone
US20210069175A1 (en) Compositions and Methods for Identifying and Treating Dystonia Disorders
Arora et al. Stabilizing rescued surface-localized ΔF508 CFTR by potentiation of its interaction with Na+/H+ exchanger regulatory factor 1
Ferreira et al. Polycystins and molecular basis of autosomal dominant polycystic kidney disease
Garner et al. ΔMEKK3: ER* activation induces a p38α/β2-dependent cell cycle arrest at the G2 checkpoint
WO2003068983A1 (en) Use of the axl receptor for diagnosis and treatment of renal disease
Mycielska et al. Expression of Na+‐dependent citrate transport in a strongly metastatic human prostate cancer PC‐3M cell line: regulation by voltage‐gated Na+ channel activity
CA2753897A1 (en) Methods for modulating metabolic and circadian rhythms
Kuhns et al. Urate transport in health and disease
JP2011529564A (en) Fluorescence-based assay for detecting sodium / calcium exchanger (NCX) “reverse mode” modulating compounds
O’Neill et al. Regulation of the calcium release channel from skeletal muscle by suramin and the disulfonated stilbene derivatives DIDS, DBDS, and DNDS
WO2014195228A1 (en) Screening method for a glut9 inhibitor
EP2919009B1 (en) LRRC8 proteins and protein complexes and methods for identification of channel modulators
Zhang et al. Age-associated decline in RAB-10 efficacy impairs intestinal barrier integrity
Gerarduzzi et al. Prostaglandin E2‐Dependent Blockade of Actomyosin and Stress Fibre Formation Is Mediated Through S1379 Phosphorylation of ROCK2
US9541546B2 (en) Method of promoting excitatory synapse formation with an anti-Ephexin5 phospho-Y361 antibody

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: 14730106

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: 14730106

Country of ref document: EP

Kind code of ref document: A1