US20150374800A1 - Inhibitors of Branched-Chain-Aminotransferase-1 (BCAT1) for the Treatment of Brain Tumors - Google Patents

Inhibitors of Branched-Chain-Aminotransferase-1 (BCAT1) for the Treatment of Brain Tumors Download PDF

Info

Publication number
US20150374800A1
US20150374800A1 US14/752,280 US201514752280A US2015374800A1 US 20150374800 A1 US20150374800 A1 US 20150374800A1 US 201514752280 A US201514752280 A US 201514752280A US 2015374800 A1 US2015374800 A1 US 2015374800A1
Authority
US
United States
Prior art keywords
bcat1
expression
cells
compound
idh
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US14/752,280
Inventor
Bernhard RADLWIMMER
Martje Toenjes
Sebastian BARBUS
Peter Lichter
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Deutsches Krebsforschungszentrum DKFZ
Original Assignee
Deutsches Krebsforschungszentrum DKFZ
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 Deutsches Krebsforschungszentrum DKFZ filed Critical Deutsches Krebsforschungszentrum DKFZ
Priority to US14/752,280 priority Critical patent/US20150374800A1/en
Publication of US20150374800A1 publication Critical patent/US20150374800A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/196Carboxylic acids, e.g. valproic acid having an amino group the amino group being directly attached to a ring, e.g. anthranilic acid, mefenamic acid, diclofenac, chlorambucil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y206/00Transferases transferring nitrogenous groups (2.6)
    • C12Y206/01Transaminases (2.6.1)
    • C12Y206/01042Branched-chain-amino-acid transaminase (2.6.1.42)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/50Physical structure
    • C12N2310/53Physical structure partially self-complementary or closed
    • C12N2310/531Stem-loop; Hairpin

Definitions

  • the present invention provides a compound capable of reducing or inhibiting (a) the biological activity of branched-chain aminotransferase-1 (BCAT1) or (b) the expression of the gen encoding BCAT1 for use in a method of treating a brain tumor.
  • BCAT1 branched-chain aminotransferase-1
  • glioblastomas account for the largest number of human malignant brain tumors. So far, the treatment o gliomas includes neurosurgical techniques (resection o stereotactic procedures), radiation therapy and chemotherapy
  • the current standard of care for, e.g., astrocytic tumor involves surgical tumor resection that can be followed b chemotherapy with the oral alkylating agent temozolamide (TMZ and radiotherapy.
  • TMZ and radiotherapy the oral alkylating agent temozolamide
  • glioblastomas are considered a being incurable as they fail to respond to ionising radiation chemotherapy and surgical resection. In other words, with these therapies only a very limited prolongation of the lifespan of patients can be achieved. This means that despite these therapies, the average life span after the cancer diagnosis is merely 12 to 16 months.
  • the technical problem underlying the present invention is to provide means for the therapy of brain tumors preferably glioblastomas or astrocytic brain tumors, which overcome the disadvantages of the current therapies and improve the survival of patients.
  • BACT1 inhibitors like gabapentin, which have been used so far for example as anticonvulsant drugs represent a novel treatment option for cancer therapy, i.e. an effective therapy for neoplasias in general and in particular for the treatment of brain tumors like astrocytic brain tumors. They potentially act by targeting a molecular pathway that is not targeted by any established chemotherapy.
  • FIG. 1 IDH wt astrocytic gliomas are characterized by hig BCAT1 expression.
  • BCAA Schematic representation of BCAA catabolism.
  • BCAAs branched-chain amino acids
  • BCKAs branched-chain ketoacids
  • BCAT1 b
  • BCAT2 c
  • 7 astrocytic gliomas 41 IDH wt and 29 IDH mut
  • Data are expressed as mean ⁇ s.d. (two-tailed Student's t-test). *, P ⁇ 0.05; ** P, ⁇ 0.001.
  • FIG. 2 BCAT1 shows substrate-dependent expression i glioblastoma cell lines.
  • FIG. 3 Expression levels of the three BCAT1 transcripts ar associated with differential methylation of two alternative promoters.
  • FIG. 4 BCAT1 suppression reduces glutamate release b glioma cells and affects concentrations of membrane fatt acids.
  • FIG. 5 BCAT1 knockdown limits glioblastoma cell invasion potential.
  • FIG. 6 BCAT1 is essential for glioblastoma progression.
  • FIG. 7 BCAT1 transcripts.
  • FIG. 8 Western blot analysis
  • FIG. 9 BCAT1 knockdown
  • BCAT1 knockdown causes apoptosis in a glioblastoma spheroi primary culture.
  • (a) Reduction of proliferation induced b lentiviral transduction of two shRNA constructs (n 3, *** P 0.0001).
  • (b) Cell cycle analysis showing strong increase of the subG1 fraction following BCAT1 knockdown (n 3). The Western blot at the top shows the increased presence of the G1-arrest marker CDKN1B in the knockdown cells.
  • AnnexinV/7AAD assay confirming apoptotic death of spheroi cells with BCAT1 knockdown.
  • FIG. 10 Click-iT EdU assay after 5 mM Gabapentin treatment for 23 h
  • FIG. 11 Click-iT EdU assay after shRNA-mediated knock down
  • the present invention relates to a compound capable of reducing or inhibiting (a) the biological activity o branched-chain-aminotransferase-1 (BCAT1) or (b) the expression of the gene encoding BCAT1 for use in a method of treating a neoplasia.
  • BCAT1 branched-chain-aminotransferase-1
  • Neoplasm is an abnormal mass of tissue as a result of neoplasia.
  • Neoplasia is the abnormal proliferation of cells The growth of the cells exceeds and is uncoordinated with respect to the normal tissues around it. The growth persistant in the same excessive manner, even after cessation of the stimuli. It usually causes a lump or tumor.
  • Neoplasms may be benign, pre-malignant (carcinoma-in-situ) or malignant (cancer).
  • the neoplasms to be treated according to the present invention are those which (over)express BCAT1.
  • the determination of BCAT1 in a neoplasm is an indication to start with a BCAT inhibiting therapy.
  • Neoplasms to be treated ar brain tumors, particularly an astrocytic brain tumor, glioma or glioblastoma, in particular those expressing IDH1 wildtype.
  • BCAAs branched-chain amino acids valine, leucine an isoleucine are essential amino acids that escape live catabolism and are available in the general circulation.
  • BCA metabolism provides an important transport system to move nitrogen throughout the body for the synthesis of non-essential amino acids, including the neurotransmitte glutamate in the central nervous system. Deregulation of the BCAA catabolic pathways frequently results in neura dysfunction.
  • the first step of BCAA catabolism involves the transfer of the alpha-amino group to alpha-ketoglutarat (alpha-KG) by the cytosolic branched-chain amino acid transaminase 1 (BCAT1) or the mitochondrial BCAT2 isoenzyme with glutamate and the respective branched chain ketoaci (BCKA) as products (Ichihara et al., J. Biochem. 59, pp. 160 169, (1966); Taylor et al., J. Biol. Chem. 241, pp. 4396-4405 (1966)).
  • BCAT1 cytosolic branched-chain amino acid transaminase 1
  • BCKA branched chain ketoaci
  • BCAT2 is ubiquitous, whereas expression of BCAT1 is restricted to a small number of tissues including brain, where BCAAs are a major source of nitrogen for the synthesis of the neurotransmitter glutamate.
  • BCKAs are catabolized further to acetyl-Co and succinyl-CoA, which enter the tricarboxylic acid (TCA cycle ( FIG. 1 a ).
  • NADH and FADH2 which are generated as a by product of BCKA catabolism, are used to transfer reducing equivalents to complex III of the respiratory chain for AT production.
  • IDH1 isocitrat dehydrogenase 1
  • WHO World Health Organization
  • IDH2 also have been detected albeit at a lower frequency of 5-10% (Bales, Bales et al. Acta Neuropath. 116, pp. 597-602 (2008); Yan et al., N. Eng J. Med. 360, pp. 765-773 (2009)).
  • IDH mutations play a central role in glioma pathogenesis (Parsons et al, Science 321, pp 1807-1812 (2008)) and have been shown to constitute a ke classifier distinguishing two major glioma subgroups that were identified initially based on RNA expression and DN methylation patterns. It has been revealed that brain tumor showing an IDH1 mutation (IDH mut ) have a better prognosis that those where IDH1 is not mutated (IDH wt ) (Hartmann et al., Act Neuropathol. 120, pp. 707-718 (2010)).
  • BCAT1 staining may help to distinguish IDH wt from IDH m gliomas; however, BCAT1 staining offers the added advantage c also distinguishing IDH wt primary glioblastoma from the clos to 10% of IDH mut astrocytomas with non-IDH1-R132H mutations Most importantly, the present invention shows that BCAT1 an BCAA metabolism provide the basis for the development of nove metabolism-based approaches in glioma therapy.
  • the data of the present invention show that availability of large amounts of glucose and glutamine, the two nutrients considered to be most important for supporting malignant cell growth, is not sufficient to support sustained fast growth of IDH wt glioblastoma.
  • BCAT1 overexpression is a highly characteristic feature of glioblastoma, in particular IDH wt glioblastoma, and essential for their aggressive clinical behavior.
  • BCAT1 expression and BCAA catabolism are promising markers for the diagnostic and prognostic assessment of gliomas and serve a novel therapeutic targets.
  • the present invention represents the first example of silencing of a metabolic gen that is central to glioma pathomechanism by IDH1 mutation associated aberrant DNA methylation.
  • the reduction, silencing or inhibition of the biological activity can be effected by direct interaction or binding of compound to BCAT1 or by indirect interaction, e.g., b interacting with a compound that is associated with the biological activity of BCAT1.
  • the reduction or inhibition of the biological activity can also be achieved by the application of altered, e.g., inactive forms of BCAT1 preferably in excess.
  • Suitable compounds reducing, silencing o inhibiting the biological activity of BCAT1 or the expression of the gene encoding BCAT1 with the aim to get a therapeutic effect are:
  • Plasmids, vectors or natural/synthetic/mutated viruses oligonucleotides of various types of modification e.g. PTO LNA, 2′F-ANA, protein-nucleotide complexes, RNA i , siRNA o mikro mi RNA, shRNA, Methylmethoxy-, Phosphoroamidates, PNA Morpholino, Phosphoramidate, Cyclohexen (CeNA), gap-meres ribozymes, aptamers, CpG-oligos, DNA-zymes, riboswitches, o lipids or lipid containing molecules; (b) peptides, peptide complexes, including all types of linkers, (c) small molecules; (d) antibodies and their derivatives, especially chimeras Fab-fragments, Fc-fragments, or (e) carriers, liposomes, nanoparticles, complexes, or an other delivery systems containing the above named construct
  • a pharmaceutical composition such compounds as described above are combined with a pharmaceuticall acceptable carrier.
  • “Pharmaceutically acceptable” is meant to encompass any carrier, which does not interfere with the effectiveness of the biological activity of the activ ingredient and that is not toxic to the host to which it is administered.
  • suitable pharmaceutical carriers are well known in the art and include phosphate buffered salin solutions, water, emulsions, such as oil/water emulsions various types of wetting agents, sterile solutions etc.
  • Suc carriers can be formulated by conventional methods and the active compound can be administered to the subject at an effective dose.
  • an “effective dose” refers to an amount of the activ ingredient that is sufficient to affect the course and the severity of the neoplasia, leading to the reduction of remission of such a pathology.
  • An “effective dose” useful for treating and/or preventing neoplasias may be determined using methods known to one skilled in the art (see for example Fingl et al., The Pharmocological Basis of Therapeutics Goodman and Gilman, eds. Macmillan Publishing Co., New York pp. 1-46 ((1975)).
  • Administration of the suitable compositions may be effected b different ways, e.g. by intravenous, intraperitoneal subcutaneous, intramuscular, topical or intraderma administration.
  • the route of administration of course depends on the kind of therapy and the kind of compound contained in the pharmaceutical composition.
  • the dosage regimen will be determined by the attending physician and other clinical factors. As is well known in the medical arts dosages for any one patient depends on many factors, including the patient's size, body surface area, age, sex, th particular compound to be administered, time and route o administration, the kind of therapy, general health and of the drugs being administered concurrently.
  • the compound useful for reducing or inhibiting the expression of the gene encoding BCAT1 is an antisense oligonucleotide shRNA or siRNA specific for BCAT1.
  • the compound useful for silencing the BCAT1 expression are independent Mission® shRNA constructs targeting different regions of th human BCAT1 (BCAT1 shRNAI NM — 005504.3-1064s1c1 and BCAT shRNAII NM — 005504.3-751s1c1) and human IDH1 (IDH1 shRNA NM — 005896.2-1363s1c1 and IDH1 shRNAII NM — 005896.2-292s1c1 mRNA transcripts.
  • suitable antisense oligonucleotides include determination of a site or sites within the BCAT1 encoding gene for the antisense interaction to occur such that the desired effect, e.g., inhibition of the expression of the protein, will result.
  • a preferred intragenic site is (a) the region encompassing the translation initiation or termination codon of the open reading frame (ORF) of the gene or (b) region of the mRNA which is a “loop” or “bulge”, i.e., no part of a secondary structure. If one or more target site have been identified, oligonucleotides are chosen which are sufficiently complementary to the target, i.e., hybridiz sufficiently well and with sufficient specificity, to give the desired effect.
  • “hybridization” means hydrogen bonding, which may be Watson Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding between complementary nucleoside or nucleotide bases
  • oligonucleotide and the DNA or RNA are complementary to each other when a sufficient number of corresponding positions in each molecule are occupied by nucleotides which can make hydrogen bonds with each other
  • “specifically hybridizable” and “complementary” are terms which are used to indicate a sufficient degree of complementarity or precise pairing such that stable an specific binding occurs between the oligonucleotide and the DNA or RNA target. It is understood in the art that the sequence of an antisense compound does not need to be 100 complementary to that of its target nucleic acid to be specifically hybridizable.
  • An antisense compound i specifically hybridizable when binding of the compound to the target DNA or RNA molecule interferes with the normal function of the target DNA or RNA to cause a loss of utility, and there is a sufficient degree of complementarity to avoid non-specific binding of the antisense compound to non-targe sequences under conditions in which specific binding i desired, i.e., in the case of therapeutic treatment.
  • the skilled person can generate antisense compounds and siRNA or shRNAs according to the present invention on the basis of the known DNA sequence for BCAT1.
  • Oligonucleotide refers to an oligomer or polymer o ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) o mimetics thereof. This term includes oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent internucleoside (backbone) linkages as well a oligonucleotides having non-naturally-occurring portions which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms because of desirable properties such as, for example, enhance cellular uptake, enhanced affinity for nucleic acid target an increased stability in the presence of nucleases.
  • antisense oligonucleotides are a preferred form of the antisense compound
  • the present invention comprehends other oligomeric antisense compounds, including but not limited to oligonucleotide mimetics such as are described below.
  • the antisense compounds in accordance with this invention compris from about 8 to about 50 nucleobases (i.e. from about 8 t about 50 linked nucleosides).
  • antisense compounds are antisense oligonucleotides, even more preferable those comprising from about 15 to about 25 nucleobases
  • Antisense compounds include ribozymes, external guide sequences (EGS), oligonucleotides (oligozymes), and other short catalytic RNAs or catalytic oligonucleotides which hybridize to the target nucleic acid and inhibit it expression.
  • the compound of the invention is a vector allowing to transcribe an antisense oligonucleotide of the invention, e.g., in a mammalian host.
  • a vector useful for gene therapy are viral vectors, e.g. adenovirus herpes virus, vaccinia, or, more preferably, an RNA virus suc as a retrovirus.
  • the retroviral vector is a derivative of a murine or avian retrovirus.
  • retroviral vectors which can be used in the presen invention are: Moloney murine leukemia virus (MoMuLV), Harve murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV) and Rous sarcoma virus (RSV).
  • MoMuLV Moloney murine leukemia virus
  • HaMuSV Harve murine sarcoma virus
  • MoMTV murine mammary tumor virus
  • RSV Rous sarcoma virus
  • a non-human primate retroviral vector is employed, such as th gibbon ape leukemia virus (GaLV), providing a broader hos range compared to murine vectors. Since recombinan retroviruses are defective, assistance is required in order to produce infectious particles.
  • GaLV gibbon ape leukemia virus
  • helper cell lines that contain plasmid encoding all of the structural genes of the retrovirus unde the control of regulatory sequences within the LTR.
  • Suitable helper cell lines are well known to those skilled in the art
  • Said vectors can additionally contain a gene encoding selectable marker so that the transduced cells can b identified.
  • the retroviral vectors can be modified in such a way that they become target specific. This can be achieved, e.g., by inserting a polynucleotide encoding sugar, a glycolipid, or a protein, preferably an antibody
  • Those skilled in the art know additional methods for generating target specific vectors.
  • Further suitable vector and methods for in vitro- or in vivo-gene therapy ar described in the literature and are known to the person skilled in the art; see, e.g., WO 94/29469 or WO 97/00957.
  • the DNA sequences for transcription of the antisense oligonucleotides can be linked to a tissue specific promoter and used for gene therapy.
  • tissue specific promoters are well known to those skilled in the art (see e.g. Zimmermann et al. (1994) Neuron 12, 11-24; Vidal et al.; (1990) EMBO J. 9, 833 840; Mayford et al., (1995), Cell 81, 891-904; Pinkert et al. (1987) Genes & Dev. 1, 268-76).
  • the phosphate groups are commonly referred to as forming the internucleoside backbon of the oligonucleotide.
  • the normal linkage or backbone of RNA and DNA is a 3′ to 5′ phosphodiester linkage.
  • Specific examples of preferred antisense compounds useful in the present invention include oligonucleotides containing modified backbones or non-natural internucleoside linkages Oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone. Modified oligonucleotide backbones which can result in increased stability are known to the person skilled in the art preferably such modification is a phosphorothioate linkage.
  • a preferred oligonucleotide mimetic is an oligonucleotide mimetic that has been shown to have excellent hybridizatio properties, and is referred to as a peptide nucleic aci (PNA).
  • PNA peptide nucleic aci
  • the sugar-backbone of a oligonucleotide is replaced with an amide containing backbone in particular an aminoethylglycine backbone.
  • the nucleobase are retained and are bound directly or indirectly to az nitrogen atoms of the amide portion of the backbone (see e.g., Nielsen et al., Science 254 (1991), 1497-1500.)
  • Modified oligonucleotides may also contain one or more substituted or modified sugar moieties.
  • Preferred oligonucleotides comprise one of the following at the 2 position: OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O- S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C 1 t C 10 alkyl or C 2 to C 10 alkenyl and alkynyl.
  • a particularly preferred modified sugar moiety is a 2′-O-methoxyethyl suga moiety.
  • Oligonucleotides of the invention may also include nucleobas modifications or substitutions.
  • Modified nucleobases include other synthetic and natural nucleobases such as 5 methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine hypoxanthine, 2-aminoadenine, 6-methyl and other alky derivatives of adenine and guanine, 2-propyl and other alky derivatives of adenine and guanine, 2-thiouracil, 2 thiothymine and 2-thiocytosine etc., with 5-methylcytosin substitutions being preferred since these modifications have been shown to increase nucleic acid duplex stability.
  • oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide.
  • moieties include lipid moieties such as a cholesterol moiety cholic acid, a thioether, a thiocholesterol, an aliphati chain, e.g., dodecandiol or undecyl residues, a phospholipid a polyamine or a polyethylene glycol chain, or adamantan acetic acid, a palmityl moiety, or an octadecylamine o hexylamino-carbonyl-oxycholesterol moiety.
  • the present invention also includes antisense compounds which are chimeric compounds.
  • “Chimeric” antisense compounds o “chimeras,” in the context of this invention, are antisens compounds, particularly oligonucleotides, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of a oligonucleotide compound.
  • oligonucleotides typically contain at least one region wherein the oligonucleotide i modified so as to confer upon the oligonucleotide increase resistance to nuclease degradation, increased cellular uptake and/or increased binding affinity for the target nucleic acid
  • An additional region of the oligonucleotide may serve as substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids.
  • RNase H is a cellular endonucleas which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target thereby greatly enhancing the efficiency of oligonucleotide inhibition of gene expression.
  • Chimeric antisense compounds of the invention may be formed as composite structures of two or more oligonucleotides, modified oligonucleotides, oligonucleoside and/or oligonucleotide mimetics as described above. Such compounds have also been referred to in the art as hybrids o gapmers.
  • the compounds for use in a method of treating a neoplasia are compounds reducing or inhibiting the biological activity o BCAT1.
  • Such compounds are described in the art for medica indications differing from the indication of the present invention and, preferably, comprise compounds like 1 (aminomethyl)cyclohexaneacetic acid (gabapentin) or a compound described in Goto et al., The Journal of Biological Chemistry 280(44) (2005), 37246-56, Hu et al., Bioorganic & Medicinal Chemistry Letters 16 (2006), 2337-40, Caballero et al. Molecular Diversity 13 (2009), 493-500, U.S. Pat. No. 6,632,831, U.S. Pat. No. 6,809,119, and EP-B1 1 157 000.
  • gabapentin aminomethylcyclohexaneacetic acid
  • antibodies capable of reducing o inhibiting the biological activity of BCAT1 are (neutralizing antibodies directed against BCAT1 or fragments thereof having substantially the same binding specificity.
  • antibody preferably, relates to antibodies which consists essentially of pooled monoclonal antibodies with different epitopic specificities, as well as distinct monoclonal antibody preparations.
  • Monoclonal antibodies are made from a antigen containing, e.g., a fragment of BCAT1 by methods well known to those skilled in the art (see, e.g., Köhler et al. Nature 256 (1975), 495).
  • antibody or “monoclonal antibody” (Mab) is meant to include intact molecules as well as antibody fragments (such as, for example Fab and F(ab′).2 fragments) which are capable of specifically binding to protein.
  • Fab and F(ab′)2 fragments lack the F fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody. (Wahl et al., J. Nucl. Med. 24: 316 325 (1983)). Thus, these fragments are preferred, as well a the products of a FAB or other immunoglobulin expression library.
  • antibodies useful for the purposes of the present invention include chimerical, single chain, an humanized antibodies.
  • preferred compounds for the purpose of the invention are inactive versions of BCAT1 or nucleic acid sequences encoding inactive versions of BCAT1 that can be introduced according to the approaches/vectors describe above.
  • Such inactive versions can be generated according to well known methods of mutagenesis.
  • Such compounds can have therapeutic effect in the human body by displacing their functionally active counterpart, in particular when applied i excess.
  • Analyses of potentially inactive versions of BCAT1 can be carried out by assaying the (reversible) transamination o branched-chain L-amino acids to branched-chain alpha-ket acids, e.g., by determining the production of glutamate Suitable assays are described in the literature and, e.g., i Example 39 of U.S. Pat. No. 6,809,119, Hu et al. (2006), an EP-B1 1 157 000.
  • the compound described in detail above is used in a method o treating an astrocytic brain tumor or glioblastoma.
  • the present invention also relates to a method for identifying a compound reducing or inhibiting the biological activity o BCAT1 and/or its expression, comprising the steps of:
  • Step (b) can be carried out by assaying the (reversible transamination of branched-chain L-amino acids to branched chain alpha-keto acids using an assay as described above.
  • candidate molecules include antibodies oligonucleotides, proteins, or small molecules. Such molecule can be rationally designed using known techniques.
  • said test system used for screening comprise substances of similar chemical and/or physical properties most preferably said substances are almost identical.
  • the compounds which can be prepared and identified according to use of the present invention may be expression libraries e.g., cDNA expression libraries, peptides, proteins, nucleic acids, antibodies, small organic compounds, ligands, hormones peptidomimetics, PNAs or the like.
  • WO 98/25146 describes further methods for screening libraries of complexes for compounds having a desired property especially, the capacity to agonize, bind to, or antagonize polypeptide or its cellular receptor.
  • the complexes in such libraries comprise a compound under test, a tag recording a least one step in synthesis of the compound, and a tethe susceptible to modification by a reporter molecule Modification of the tether is used to signify that a comple contains a compound having a desired property.
  • the tag can be decoded to reveal at least one step in the synthesis of such compound.
  • Other methods for identifying compounds which interac with BCAT1 or nucleic acid molecules encoding such molecule are, for example, the in vitro screening with the phage display system as well as filter binding assays or “real time” measuring of interaction.
  • All these methods can be used in accordance with the present invention to identify a compound reducing or inhibiting the biological activity of BCAT1 or its expression.
  • the gene encoding BCAT1 can also serve as a target for the screening of inhibitors.
  • Inhibitors may comprise, for example proteins that bind to the mRNA of the genes encoding BCAT1 thereby destabilizing the native conformation of the mRNA an hampering transcription and/or translation.
  • nucleic acid molecules such as a RNA fragment that mimics the structure of a defined or undefined target RNA molecule t which a compound binds inside of a cell resulting in the retardation of the cell growth or cell death; see, e.g., W 98/18947 and references cited therein.
  • These nucleic acid molecules can be used for identifying unknown compounds o pharmaceutical interest, and for identifying unknown RNA targets for use in treating a disease.
  • These methods an compositions can be used for identifying compounds useful to reduce expression levels of BCAT1.
  • the compounds which can be tested and identified according to the method of the invention may be expression libraries, e.g. cDNA expression libraries, peptides, proteins, nucleic acids antibodies, small organic compounds, hormones peptidomimetics, PNAs or the like (Milner, Nature Medicine (1995), 879-880; Hupp, Cell 83 (1995), 237-245; Gibbs, Cell 7 (1994), 193-198 and references cited supra).
  • genes encoding a putative regulator of BCAT1 and/or which exert their effects up- or downstream of BCAT1 may be identified using, for example, insertion mutagenesis using for example, gene targeting vectors known in the art.
  • Sai compounds can also be functional derivatives or analogues o known inhibitors.
  • Such useful compounds can be for example transacting factors which bind to BCAT1 or regulator sequences of the gene encoding it.
  • Identification o transacting factors can be carried out using standard method in the art. To determine whether a protein binds to the protein itself or regulatory sequences, standard native gel shift analyses can be carried out. In order to identify transacting factor which binds to the protein or regulator sequence, the protein or regulatory sequence can be used as a affinity reagent in standard protein purification methods, o as a probe for screening an expression library.
  • Th identification of nucleic acid molecules which encod polypeptides which interact with BCAT1 can also be achieved, for example, as described in Scofield (Science 274 (1996), 2063 2065) by use of the so-called yeast “two-hybrid system”.
  • BCAT1 is linked to the DNA-binding domain of the GAL transcription factor.
  • a yeast strain expressing this fusio polypeptide and comprising a lacZ reporter gene driven by a appropriate promoter, which is recognized by the GAL transcription factor, is transformed with a library of cDNA which will express plant proteins or peptides thereof fused t an activation domain.
  • a peptide encoded by one of the cDNAs is able to interact with the fusion peptide comprising peptide of BCAT1, the complex is able to direct expression o the reporter gene.
  • BCAT1 and the gene encoding BCAT1 can be used to identify peptides and proteins interacting with BCAT1. It is apparent to the person skilled in the art that this and similar systems may then further be exploited for the identification of inhibitors.
  • Total RNA 50 ng was reverse-transcribed using random primers an superscript II (Invitrogen, Düsseldorf, Germany) according to the manufacturer's instructions.
  • Each complimentary DNA sample was analysed in triplicate with the Applied Biosystems Pris 7900HT Fast Real-Time PCR System (Applied Biosystems, Foste City, Calif., USA) using Absolute SYBR Green ROX Mix (ABgene Epsom, UK). The relative amount of specific mRNA was normalised to ARF, B2M and TBP. Primer sequences are given in the following Table 1A.
  • monoclonal mouse antibod against ⁇ -tubulin (1:2000, #T9026, Sigma-Aldrich, St. Louis Mo. 63178, USA)
  • monoclonal mouse antibody against BCAT (1:2000, ECA39, clone 51, #611270, BD Biosciences, San Jose Calif., USA
  • monoclonal rat antiserum against IDH1 (1:1, provided by A.
  • Tumor sections were deparaffinised using xylol and rehydrated in decreasing concentrations of ethanol.
  • Antigen retrieval was performed by heating for 40 min in a steamer in 10 mM sodium citrate buffer (pH 6.0). Endogenous peroxidase was inactivate by incubating the tumor sections in 3% hydrogen peroxide Sections were incubated overnight with primary anti-BCAT (ECA39) monoclonal mouse antibody, clone 51 (BD Biosciences San Jose, Calif.) diluted 1:500 or mouse anti-human IDH1 R132 antibody (Dianova, Hamburg, Germany) diluted 1:20 in Dak REALTM antibody diluent (Dako, Glostrup, Denmark).
  • DNA methylation was analyzed by MassARRAY technique. Briefly 500 ng genomic DNA was chemically modified with sodium bisulfite using the EZ methylation kit (Zymo Research according to the manufacturer's instructions. The bisulfite treated DNA was PCR-amplified with primers generating fou amplicons (A1-A4) from ⁇ 990 bp to +612 bp around TSS of BCAT transcript 1 (T1, ENST00000261192), and three amplicons (A5 A7) of the promoter of BCAT1 transcript 4 (T4 ENST00000539282) and 6 (T6, ENST00000538118) from ⁇ 198 bp t +63 bp.
  • the amplicons were transcribed by T7 polymerase followed by T specific-RNAaseA cleavage.
  • the digested fragments were quantified by mass-spectrometry.
  • the prime sequences are given in Table 1B.
  • DNA methylation standard (0%, 20%, 40%, 60%, 80%, and 100% methylated genomic DNA) were used to confirm the unbiased amplification of the amplicons.
  • the human glioma cell lines U-87MG (HTB-14), LN-229 (CRL 2611), Hs683 (HTB-138) and U-373MG (HTB-17) were cultured i Dulbecco's Minimal Essential Medium (DMEM) supplemented with 10% fetal calf serum and 1% penicillin/streptomycin-mix.
  • DMEM Dulbecco's Minimal Essential Medium
  • Cel lines were authenticated by short tandem repeat (STR analysis.
  • the brain cancer stem-like cells NCH421k were established from primary glioblastoma patients undergoing surgical resection according to the research proposal approved by the institutional review board at the Medica Faculty, University of Heidelberg. They were characterized genotypically and phenotypically in a previous study (Campos B., et al.
  • NCH421k cells were grown as floating aggregates (neurospheres on uncoated tissue culture dishes in DMEM/F-12 mediu containing 20% BIT serum-free supplement, basic fibroblas growth factor and epidermal growth factor at a concentration of 20 ng/ml each (all from Provitro, Berlin, Germany).
  • HEK293 and HEK293 cells were maintained as monolayer cultures in DME containing 10% fetal calf serum without antibiotics.
  • ChIP was performed on HEK293 cells overexpressing flag-tagged HEY1, since the currently available antibodies against HEY were not specific for ChIP assays.
  • Constructs for HEY overexpression were prepared using gateway compatible vector tagged with FLAG; pDest26 (C-terminal tag) and pDest11 (N terminal tag). 1 ⁇ g of either the control vectors or HEY expression vectors were transfected into 2.5 ⁇ 10 ⁇ 6 HEK293 cell per 15 cm plate using TransIT®-LT1 (Mirus Bio LLC, Madison, W 53711, USA) according to slightly modified manufacturer' instructions. The cells from two plates were harvested 4 hours after transfection.
  • Chromatin was prepared using non ionic shearing buffers with Covaris S2, according to the manufacturer's protocol (Covaris Inc.). ChIP was performed using anti-FLAG antibody (Cell signaling #2368). The DNA recovered after ChIP was used for qRT-PCR with input chromati and mock immunoprecipitation (anti-IgG antibody, Diagenode Kch-803-015) serving as controls. qRT-PCR was performed i triplicate with SYBR green detection using primers listed i Table 2. Ratios of bound to input signals are reported.
  • HEK293T cells were transfected with siRNA duplexes from Ambio (HEY1: s23868-70) or Dharmacon (control non-targeting: D 001810-10) using DhamaFect1 following slight modifications t the manufacturer's instructions (Dharmacon). Briefly, eac siRNA pool was diluted in 1 ⁇ siRNA buffer (Dharmacon), wa mixed with RPMI, and then distributed into 6 wells of 96 wel plates. 1.2 ⁇ 10 ⁇ 4 HEK293 or 9 ⁇ 10 ⁇ 3 HEK293T cells were seeded o top of the siRNA/DharmaFECT mixture (the volume was 15 ⁇ l/well and 20 nM of siRNAs in final). After 48 hour incubation at 37° C., RNAs were isolated from the wells fo further analysis.
  • Lentiviral vectors were produced by cotransfection of HEK293 cells with the psPAX2 (Addgene 12260, Didier Trono, packaging vector), pMD2.G (Addgene 12259, Didier Trono, envelop plasmid), and pLKO.1 shRNA constructs (Sigma-Aldrich) Transfections were carried out using TransIT®-LT1 (Mirus Bi LLC) and virus was harvested at 48 and 72 hours after transfection and combined.
  • Infection of glioma cells with virus at an M.O.I. of 2 was carried out in the presence of 8 ⁇ g/ml of polybrene (Chemicon Billerica, Mass., USA). Virus-containing supernatant was removed after 24 hours and cells were split on day 3, day 5 and day after transduction.
  • 5 ⁇ 10 ⁇ 4 cells/well were seeded in 24-well plates in a total volume of 500 ⁇ l cell culture medium.
  • the medium was removed 16 hours after seeding the cells, and replaced with 500 ⁇ medium containing 5 mM or 10 mM dimethyl- ⁇ -ketoglutarat (Dimethyl 2-oxoglutarate; Sigma-Aldrich) or 5 mM, 10 mM or 2 mM gabapentin (1-(Aminomethyl)-cyclohexane; Sigma-Aldrich)
  • 5 mM, 10 mM or 2 mM gabapentin (1-(Aminomethyl)-cyclohexane; Sigma-Aldrich) Corresponding volumes of 200 mM HEPES buffer (pH 7.4) were added to the respective control wells.
  • NCH421k cells were incubated with annexin V-P (Phycoerythrin) and 7-AAD (7-aminoactinomycin D, B Biosciences) for 15 minutes in the dark, immediately followed by flow cytometry.
  • Glutamate concentration in the supernatant of cells treated with gabapentin was determined using the glutamine/glutamat determination Kit (GLN-1; Sigma-Aldrich) according to the manufacturer's instructions.
  • the reaction volumes were scaled down to 100 ⁇ l total volume and absorbance was measured in triplicate in a microplate (Corning® 96 Well Clear Flat Bottom UV-Transparent Microplate) using a Tecan Infinite M200 plat reader (Tecan, Austria). Data were normalised to the number o cells per well.
  • Cells were plated on glass coverslips five days after lentiviral transduction. Cells were fixed in 4% formaldehyde rinsed twice in 1 ⁇ PBS, and permeabilized in PBS containing 0.2% Triton. Following rinsing with 1 ⁇ PBS cells were incubated in 10% goat serum for 30 minutes at room temperature. Samples were incubated 1 hour with the primary antibody (mouse anti ⁇ -tubulin antibody, 1:200, #T9026, Sigma Aldrich) and 1 hour at room temperature with the secondary antibody (FITC-conjugated goat anti mouse antibody, 1:100 ab6785, Abcam) following Mounting with DAPI-containing Vectashield mounting medium (Vector Laboratories, Burlingame Calif., USA). For fluorescence imaging, images were taken using 40 ⁇ objective lense on a Zeiss Axioplan microscope.
  • PDMS Poly(dimethylsiloxane) based microchannel chips were kindly provided by Dr. Ralf Kemkemer (Max Planck Institute for Intelligent Systems, Germany). Microfabricated channe structures with the dimensions of 5 ⁇ 11 ⁇ 300 ⁇ m (W ⁇ H ⁇ L) were bio-functionalized by incubation with a 50 ⁇ g/ml fibronecti solution prior to use. The chip was fixed on a Teflon holde and 2 ⁇ 10 ⁇ 5 cells were seeded on the chip in close proximity t the channels. After cells were attached on the chip, live-cell imaging was performed for 25 hours.
  • a total of 2 ⁇ 10 ⁇ 5 U-87MG cells with BCAT1 shRNAI knockdown o nontargeting shRNA were stereotactically implanted into the brain of six 7-9-week-old athymic mice (CD1 nu/nu; Charle River Laboratories, Wilmington, Mass., USA), respectively.
  • CD1 nu/nu Charle River Laboratories, Wilmington, Mass., USA
  • Fou weeks after implantation animals were sacrificed, brain removed and cryosectioned. Brain sections were stained with hematoxylin and eosin and the tumor volume was calculate using ImageJ.
  • the lyophilised extract residues were dissolved in 0.5 ml of D 2 O buffer (99.9% D) containing 30 mM sodium phosphate and 0. mM sodium azide (pH 7.02).
  • D 2 O buffer 99.9% D
  • capillary 1.5 mm OD, 0.99 mm ID
  • TSP trimethylsilyl-2,2,3,3-tetradeuteropropanoi acid
  • a calibration solution was prepared by placin weighed amounts of glucose and citric acid in the D 2 O buffer t give the calculated concentrations of 10.49 mM glucose an 4.95 mm glucose.
  • the signal integrals for citrate and glucose were set to value corresponding to the concentrations given above, and th integral of the TSP methyl signal was found to be equivalen to 4.60 ⁇ 0.10 mM protons.
  • Cell pellets were diluted in 100 ⁇ l water and homogenized b. sonification (Ultrasonic device, 3 ⁇ 5 ⁇ 20 cycle, output 80% Branson Sonifier 450, Dietzenbach, Germany). Protein wa determined according to Lowry (Lowry, O. H., Rosebrough, N. J. Farr, A. L. & Randall, R. J. Protein measurement with the Foli phenol reagent. J Biol Chem 193, 265-275 (1951) with th modifications of Helenius and Simons (Helenius, A. & Simons K. The binding of detergents to lipophilic and hydrophili proteins. J Biol Chem 247, 3656-3661 (1972)) using bovine seru albumin as a standard. The final protein concentrations of th homogenates should be in a range of 2-4 mg/ml. These dilution were used for all analyses.
  • the human glioblastoma cell lines U87-MG (HTB-14), HS683 (HTB 138) and U373-MG (HTB-17; LGC Standards, Teddington TW11 0LY U.K.) were cultured in Dulbecco's Minimal Essential Mediu supplemented with 10% fetal calf serum and 1 penicillin/streptomycin-mix.
  • 5 ⁇ 10 4 cells/well were seeded in 2 well plates in a total volume of 500 ⁇ l cell culture medium
  • the medium was removed 4 hours after seeding the cells, an replaced with 500 ⁇ l medium containing 1-(Aminomethyl)cyclohexylessigklare (Gabapentin, dissolved in 200 mM HEPE buffer at a concentration of 500 mM) at final concentrations o 5 mM, 10 mM and 20 mM, respectively.
  • Corresponding volumes o 200 mM HEPES were added to the respective control wells.
  • 5-ethynyl-2′-deoxyuridin EdU; Carlsbad, Calif.
  • Lentiviral vectors were produced by the cotransfection o HEK293T cells with the psPAX2 (Addgene 12260, Didier Trono packaging vector), pMD2.G (Addgene 12259, Didier Trono envelope plasmid), and pLKO.1 shRNA constructs (Sigma-Aldrich St. Louis, Mo. 63178, USA). Transfections were carried ou using TransIT®-LT1 (Mirus Bio LLC, Madison, Wis. 53711, USA) an virus was harvested at 48 and 72 hours after transfection.
  • Tw independent shRNA constructs targeting different regions o the BCAT1 mRNA transcript were used: MISSION shRN TRCN0000005907 NM — 005504.3-1064s1c1 (shRNA1) an TRCN0000005909 NM — 005504.3-751s1c1 (shRNA2); all Sigma Aldrich, St. Louis, Mo. 63178, USA. Nontargeting shRNA was use as a control.
  • the human glioblastoma cell lines U87-MG (HTB-14), HS683 (HTB 138) and U373-MG (HTB-17; LGC Standards, Teddington TW11 0LY U.K.) were seeded in 24 well plates (5 ⁇ 104 cells/well) in total volume of 500 ⁇ l cell culture medium. After 24 hour cells were transduced with virus in the presence of 8 ⁇ g/ml o polybrene. Virus-containing supernatant was removed after 2 hours and cells were split on day 3 and day 5 afte transduction. Decreased BCAT1 mRNA and protein expression wa detected using quantitative real-time PCR and Western blo (ECA39 antibody, BD Biosciences Pharmingen, San Diego, Calif.
  • a proliferation assay was carried out on day 6 using the Click-iT® EdU cell proliferation kit after incubating cells with 10 ⁇ M EdU for 16 hours. In all of the BCAT1 shRNA transduced cells a reduction of proliferation was observed ranging from 20-80% depending on the cell line. These results are shown in FIG. 11 .
  • BCAT1 was found to be expressed strongly in the glioblastoma cell lines LN-229, U-87MG, U-373MG and to a lesser extent in A172 ( FIG. 2 a ), all of which were confirmed to carry IDH1 and IDH2 wildtype genes. BCAT1 expression also was elevated in the Hs683 cell line, which was originally derived from an oligodendroglioma but nevertheless displays an IDH wt genotype. Thus, all these cell lines can be considered suitable models for studying BCAT1 function. BCAT1 RNA and protein expression were upregulated under hypoxic conditions, which are frequently present in glioblastoma ( FIG. 2 b ).
  • BCAT1 expression correlated with the concentration of the substrate alpha-KG and was upregulated after Increasing the concentration of cell-permeable dimethyl-alpha-KG-substrate in the culture medium ( FIG. 2 c ).
  • shRNA-mediated knockdown of IDH1 a major source of alpha-KG in the cytoplasm, led to strong downregulation of BCAT1 expression ( FIG. 2 d ).
  • transcript expression in patient samples was quantified.
  • expression of three protein-coding BCAT1 transcripts listed in the Ensembl database in IDH wt astrocytic primary tumors as well as in a pool of 23 normal brain tissues were confirmed.
  • These three transcripts (T1, T4 and T6) encode proteins of 386, 398, and 385 amino acids, all three of which correspond to the single protein band of 43 kD as identified by Western blot analysis.
  • These transcripts originate from two alternative promoters ( FIG.
  • T6 the predominant transcript representing 73% of all BCAT1 mRNA in primary tumors ( FIG. 3 b ). Notably, expression of all BCAT1 transcripts was significantly higher in IDH wt compared to IDH mut tumors.
  • siRNA-mediated HEY1 knockdown in HEK293T cells increased transcript-T1 expression ( FIG. 3 h,i ).
  • ChIP analysis demonstrated that compared to an upstream control region and to a region close to the translation start site, the strongest binding of two different HEY1 constructs occurs in the DMR ( FIG. 3 j,k ). Together, these data strongly show differential expression of BCAT1 transcripts in astrocytomas is regulated by DNA methylation involving broad methylation of promoter 2 in IDH mut tumors and CpG-site specific methylation in a HEY1-repressor binding site in promoter 1 of IDH wt tumors.
  • BCAT1 knockdown led to strong downregulation of 3-hydroxyacyl CoA dehydrogenase (HADH), an enzyme participating in the catabolism of valine and isoleucine downstream of BCAT1 ( FIG. 4 f, g ). Since HADH is central to fatty acid metabolism, BCAT1 knockdown would also alter synthesis or degradation of fatty acids essential for membrane synthesis.
  • HADH 3-hydroxyacyl CoA dehydrogenase
  • BCAT1 knockdown strongly affected cell morphology, resulting in a rounded, less extended appearance ( FIG. 5 a - b ).
  • a microchannel migration chip to simulate a three-dimensional environment was used ( FIG. 5 c ).
  • the majority of U-87MG cells (55%) penetrated short distances into the microchannels, but were unable to actively deform themselves in order to completely invade the microchannels whereas most control cells (78%) completely invaded the channels ( FIG. 5 d ).
  • This reduced invasiveness of the BCAT1 knockdown cells might be due to altered membrane composition caused by the observed lower abundance of long-chain fatty acids and differences in cholesterol metabolism ( FIG. 4 g ); such changes might sufficiently affect general availability of membrane components as well as membrane elasticity to hinder cell invasion.
  • BCAT1 is Essential for Glioblastoma Growth

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Biomedical Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Zoology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Biophysics (AREA)
  • Plant Pathology (AREA)
  • Physics & Mathematics (AREA)
  • Immunology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Virology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Oncology (AREA)
  • Mycology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

Described is a compound capable of reducing or inhibiting (a) the biological activity of branched-chain-aminotrans-ferase-1 (BCATI) or (b) the expression of the gene encoding BCATI for use in a method of treating a neoplasia. A preferred compound is 1-(aminomethyl)cyclohexaneacetic acid (gabapentin).

Description

  • The present invention provides a compound capable of reducing or inhibiting (a) the biological activity of branched-chain aminotransferase-1 (BCAT1) or (b) the expression of the gen
    Figure US20150374800A1-20151231-P00999
    encoding BCAT1 for use in a method of treating a brain tumor.
  • Malignant human glioblastomas account for the largest number of human malignant brain tumors. So far, the treatment o
    Figure US20150374800A1-20151231-P00999
    gliomas includes neurosurgical techniques (resection o
    Figure US20150374800A1-20151231-P00999
    stereotactic procedures), radiation therapy and chemotherapy The current standard of care for, e.g., astrocytic tumor involves surgical tumor resection that can be followed b
    Figure US20150374800A1-20151231-P00999
    chemotherapy with the oral alkylating agent temozolamide (TMZ and radiotherapy. However, glioblastomas are considered a being incurable as they fail to respond to ionising radiation chemotherapy and surgical resection. In other words, with these therapies only a very limited prolongation of the lifespan of patients can be achieved. This means that despite these therapies, the average life span after the cancer diagnosis is merely 12 to 16 months.
  • Thus, the technical problem underlying the present invention is to provide means for the therapy of brain tumors preferably glioblastomas or astrocytic brain tumors, which overcome the disadvantages of the current therapies and improve the survival of patients.
  • The solution of said technical problem is achieved b
    Figure US20150374800A1-20151231-P00999
    providing the embodiments characterized in the claims.
  • During the experiments resulting in the present invention it was found that known BACT1 inhibitors, like gabapentin, which have been used so far for example as anticonvulsant drugs represent a novel treatment option for cancer therapy, i.e. an effective therapy for neoplasias in general and in particular for the treatment of brain tumors like astrocytic brain tumors. They potentially act by targeting a molecular pathway that is not targeted by any established chemotherapy.
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1: IDHwt astrocytic gliomas are characterized by hig
    Figure US20150374800A1-20151231-P00999
    BCAT1 expression.
  • (a) Schematic representation of BCAA catabolism. BCAAs branched-chain amino acids; BCKAs, branched-chain ketoacids (b,c) RNA expression of BCAT1 (b) and BCAT2 (c) in 7 astrocytic gliomas (41 IDHwt and 29 IDHmut) normalized t
    Figure US20150374800A1-20151231-P00999
    expression in normal brain (dashed line). Data are expressed as mean±s.d. (two-tailed Student's t-test). *, P<0.05; ** P, <0.001. (d) Western blot showing expression of BCAT protein in astrocytic gliomas with IDH1 and IDH2 wildtype genes (lanes 1-5), different mutations in the IDH2 (lanes 6-7 or IDH1 (lanes 8-12) genes, and normal brain (lane 13). AII diffuse astrocytoma WHO grade II; AAIII, anaplasti astrocytoma WHO grade III; sGBIV, secondary glioblastoma WH grade IV; pGBIV, primary glioblastoma WHO grade IV; AOIII anaplastic oligodendroglioma WHO grade III. (e-h Immunohistochemical stainings of BCAT1 in an IDHwt primary glioblastoma (e), a primary glioblastoma with IDH1-R132 mutation (f), a diffuse astrocytoma with IDH1-R132C mutation (g), and an anaplastic oligodendroglioma with IDH2-R172 mutation (h). (i,j) Immunohistochemical staining of IDH1-R132 in the same tumors as in panels b and c, respectively demonstrating the complementarity of BCAT1 and IDH1-R132 staining. Scale bars: 50 μm. (k) Two-by-two table showing the significant correlation of BCAT1 protein expression an
    Figure US20150374800A1-20151231-P00999
    mutation status of the IDH1 and IDH2 genes in 81 gliomas (p
    Figure US20150374800A1-20151231-P00999
    0.0001; Fisher's Exact Test).
  • FIG. 2: BCAT1 shows substrate-dependent expression i
    Figure US20150374800A1-20151231-P00999
    glioblastoma cell lines.
  • (a) Western blot showing BCAT1 protein expression in IDH glioma cell lines. (b-d) Effects of hypoxia and alpha-KG o
    Figure US20150374800A1-20151231-P00999
    BCAT1 expression. RNA and protein expression are shown at the bottom and the top of each panel. The numbers above the Western blots indicate the fold ratios of expression relative to control cells. The mRNA expression values represent mean s.d. of triplicate samples. (b) BCAT1 is upregulated under hypoxic (1% O2) conditions. (c) BCAT1 expression is induced by supplementation of the culture media with cell-permeable dimethyl-alpha KG. (d) Knockdown by two different shRNAs o
    Figure US20150374800A1-20151231-P00999
    the alpha-KG-producing cytoplasmatic IDH1 leads the downregulation of BCAT1 expression.
  • FIG. 3: Expression levels of the three BCAT1 transcripts ar associated with differential methylation of two alternative promoters.
  • (a) Schematic drawing of exons 1 to 4 of BCATI showing the exon structure of the three transcripts T1, T4 and T6. The two alternative promoters 1 and 2 are shown in the enlarge sections on the lower left and lower right, respectively. (b
    Figure US20150374800A1-20151231-P00999
    qRT-PCR quantification of RNA expression of transcripts T1, T
    Figure US20150374800A1-20151231-P00999
    and T6 in astrocytic gliomas and a pool of RNAs from normal brain tissues (n=23). Values represent mean±s.d. o
    Figure US20150374800A1-20151231-P00999
    triplicate samples. (c) DNA-methylation patterns detected i
    Figure US20150374800A1-20151231-P00999
    promoters 1 (left) and 2 (right) by massarray analysis o
    Figure US20150374800A1-20151231-P00999
    bisulfite-PCR amplicons A1 to A8 in normal brain an astrocytic gliomas WHO grade II-IV. *, IDHwt anaplasti
    Figure US20150374800A1-20151231-P00999
    astrocytoma. (d-f), Extent of DNA methylation in normal brain (Nbr), IDHwt and IDHmut tumors in (d) average of all CpGs i
    Figure US20150374800A1-20151231-P00999
    promoter 2 (two-tailed Student's t-test, P<0.0001) (e) CpG4, (P=0.0003) and (f) CpG6 in promoter 1 (P<0.0001). (g
    Figure US20150374800A1-20151231-P00999
    Correlation of CpG6 methylation grade and BCAT1 T1 expression (h,i) Knockdown of HEY1 in HEK293T cells (h) results in upregulation of BCAT1 expression (i). Values represent mean s.d. (n=3). (j,k) ChIP assays showing preferred binding o
    Figure US20150374800A1-20151231-P00999
    HEY1 to the DMR (amplicon c1) as compared to control amplicon in promoter 1 (c2), about 5kb upstream of BCAT1, and unrelate positive (BHLH15) and negative (PTPRD) controls. (j) Flag-HEY construct. (k) HEY1-Flag construct. qRT-PCR was performed i
    Figure US20150374800A1-20151231-P00999
    triplicate and ratios of bound to input are shown a
    Figure US20150374800A1-20151231-P00999
    mean±s.d.
  • FIG. 4: BCAT1 suppression reduces glutamate release b
    Figure US20150374800A1-20151231-P00999
    glioma cells and affects concentrations of membrane fatt
    Figure US20150374800A1-20151231-P00999
    acids.
  • (a,b) NMR spectra of U-87MG (a) and U-373MG (b) cells after treatment with control (−) or 20 mM gabapentin (+) for 2 hours. Difference spectra are shown near the top of the panels. Upon inhibitor treatment levels of valine (Val) leucine (Leu) and isoleucine (Ile) increased by factors 1.09 1.38, 1.19, respectively, in U-87MG cells and by factors 1.83 2.18% and 2.32, respectively, in U-373MG cells. (c) Glutamat release by glioma cells at 6 and 12 hours after start o treatment with control (−) or with BCAT1 inhibition (+) by 2 mM gabapentin (n=3). (d) Tandem-MS analysis of amino acid concentrations in culture media of BCAT1 knockdown and control U-87MG cells 8 days after lentiviral transduction. Values ar shown as difference to the media starting concentrations Positive and negative values indicate amino acid release an
    Figure US20150374800A1-20151231-P00999
    uptake, respectively (n=6). (e) Intracellular amino acid concentrations of the same cells as in (d) (n=6). (f) Western blot showing downregulation of HADH upon BCAT1 knockdown. (g
    Figure US20150374800A1-20151231-P00999
    Relative depletion of cholesterol and very long chain fatt
    Figure US20150374800A1-20151231-P00999
    acids in BCAT1 knockdown vs. control U-87MG cells (n=3). Dat
    Figure US20150374800A1-20151231-P00999
    are expressed as mean±s.d. *, P<0.05; **, P<0.01; **
    Figure US20150374800A1-20151231-P00999
    P<0.001.
  • FIG. 5: BCAT1 knockdown limits glioblastoma cell invasion potential.
  • (a,b) Immuno-fluorescence labeling of alpha-tubulin in control (a) and BCAT1 knockdown (b) U-87MG cells. blue: DAPI; scal
    Figure US20150374800A1-20151231-P00999
    bar: 50 μm. (c) Sequential images showing the permeation of U-87MG cell through a 5×11×300 μm microchannel over a period of 9 hours; scale bar: 50 μm. (d) BCAT1 knockdown significantly inhibits the invasion potential of U-87MG cell compared to nontarget shRNA transduced cells. Results indicate the mean±s.d. of three independent experiments. P=0.0146.
  • FIG. 6: BCAT1 is essential for glioblastoma progression.
  • (a-d) Cell proliferation and cell cycle analyses of glioma cells upon BCAT1 suppression. Proliferation of cells was examined using the Click-iT® EdU cell proliferation assay Cell cycle analysis was performed after DNA staining with propidium iodide. The DNA distribution is shown for living cells. Values in graphs represent mean±s.d. for n=3. nt nontarget shRNA. *, P<0.05; **, P<0.01; *** P<0.001 compared to respective controls. (a) Treatment with the BCAT inhibitor gabapentin for 20 hours suppressed proliferation of glioma cell lines in a concentration-dependent manner relative to control cells treated with solvent only. (b) Cell cycle analysis of gabapentin-treated glioma cells showed a accumulation of cells in 01-phase. (c) Knockdown of BCAT caused a significant reduction of cell proliferation relative to samples treated with nontarget shRNA (nt) in all three glioma cell lines and (d) resulted in the accumulation of the G1-arrest marker CDKN1B/p27KIP1. Cell cycle analysis showed significant increases of the proportions of cells in G1 phase (e) BCAT1 knockdown results in decreased phosphorylation o
    Figure US20150374800A1-20151231-P00999
    AKT. (f-i) Cross-sections of tumors induced by intracrania injection of U-87MG glioblastoma cells into CD-1 nude mice Hematoxylin-eosin staining is shown for mice injected with (f
    Figure US20150374800A1-20151231-P00999
    control nontarget-shRNA or (g) BCAT1-shRNA. (h) Quantification of tumor volumes (n=5 mice for each group, P=0.0091).
  • FIG. 7: BCAT1 transcripts.
  • (a) Gene structure of BCAT1 showing 11 exons and the three transcripts T1 (ENST00000261192), T4 (ENST00000539282) and T
    Figure US20150374800A1-20151231-P00999
    (ENST00000538118) which originate in two different promoters The regions around the transcription start sitzes (TSS) an exon 5 are enlarged to show primer locations. (b) Agarose ge
    Figure US20150374800A1-20151231-P00999
    image of PCR products amplified from an IDHwt glioblastom (left) and a pool of normal brain RNAs from 23 individual (right) using the reverse primer and the transcript-specific exon 1 primers. Band sizes match the expected sizes of the respective spliced mRNAs.
  • FIG. 8: Western blot analysis
  • Western Blot analysis of total and phosphorylated protein of the mTOR target RPS6K upon (a) gabapentin treatment and (b
    Figure US20150374800A1-20151231-P00999
    BCAT1 knockdown in the cell lines U-87MG, U-373MG and Hs683.
  • FIG. 9: BCAT1 knockdown
  • BCAT1 knockdown causes apoptosis in a glioblastoma spheroi
    Figure US20150374800A1-20151231-P00999
    primary culture. (a) Reduction of proliferation induced b
    Figure US20150374800A1-20151231-P00999
    lentiviral transduction of two shRNA constructs (n=3, *** P 0.0001). (b) Cell cycle analysis showing strong increase of the subG1 fraction following BCAT1 knockdown (n=3). The Western blot at the top shows the increased presence of the G1-arrest marker CDKN1B in the knockdown cells. (c
    Figure US20150374800A1-20151231-P00999
    AnnexinV/7AAD assay confirming apoptotic death of spheroi
    Figure US20150374800A1-20151231-P00999
    cells with BCAT1 knockdown.
  • FIG. 10: Click-iT EdU assay after 5 mM Gabapentin treatment for 23 h
  • See Example 2 for experimental details.
  • FIG. 11: Click-iT EdU assay after shRNA-mediated knock down
  • See Example 3 for experimental details
  • Thus, the present invention relates to a compound capable of reducing or inhibiting (a) the biological activity o
    Figure US20150374800A1-20151231-P00999
    branched-chain-aminotransferase-1 (BCAT1) or (b) the expression of the gene encoding BCAT1 for use in a method of treating a neoplasia.
  • “Neoplasm” is an abnormal mass of tissue as a result of neoplasia. “Neoplasia” is the abnormal proliferation of cells The growth of the cells exceeds and is uncoordinated with respect to the normal tissues around it. The growth persistant in the same excessive manner, even after cessation of the stimuli. It usually causes a lump or tumor. Neoplasms may be benign, pre-malignant (carcinoma-in-situ) or malignant (cancer). The neoplasms to be treated according to the present invention are those which (over)express BCAT1. Thus, the determination of BCAT1 in a neoplasm is an indication to start with a BCAT inhibiting therapy. Neoplasms to be treated ar
    Figure US20150374800A1-20151231-P00999
    brain tumors, particularly an astrocytic brain tumor, glioma
    Figure US20150374800A1-20151231-P00999
    or glioblastoma, in particular those expressing IDH1 wildtype.
  • The branched-chain amino acids (BCAAs) valine, leucine an isoleucine are essential amino acids that escape live catabolism and are available in the general circulation. BCA metabolism provides an important transport system to move nitrogen throughout the body for the synthesis of non-essential amino acids, including the neurotransmitte glutamate in the central nervous system. Deregulation of the BCAA catabolic pathways frequently results in neura
    Figure US20150374800A1-20151231-P00999
    dysfunction. The first step of BCAA catabolism involves the transfer of the alpha-amino group to alpha-ketoglutarat
    Figure US20150374800A1-20151231-P00999
    (alpha-KG) by the cytosolic branched-chain amino acid transaminase 1 (BCAT1) or the mitochondrial BCAT2 isoenzyme with glutamate and the respective branched chain ketoaci
    Figure US20150374800A1-20151231-P00999
    (BCKA) as products (Ichihara et al., J. Biochem. 59, pp. 160 169, (1966); Taylor et al., J. Biol. Chem. 241, pp. 4396-4405 (1966)). Expression of BCAT2 is ubiquitous, whereas expression of BCAT1 is restricted to a small number of tissues including brain, where BCAAs are a major source of nitrogen for the synthesis of the neurotransmitter glutamate. Following transamination, BCKAs are catabolized further to acetyl-Co
    Figure US20150374800A1-20151231-P00999
    and succinyl-CoA, which enter the tricarboxylic acid (TCA cycle (FIG. 1 a). NADH and FADH2, which are generated as a by product of BCKA catabolism, are used to transfer reducing equivalents to complex III of the respiratory chain for AT
    Figure US20150374800A1-20151231-P00999
    production.
  • Mutations in IDH1 (isocitrat dehydrogenase 1), originally detected in a fraction of glioblastomas are present in the great majority of World Health Organization (WHO) grade II an
    Figure US20150374800A1-20151231-P00999
    III gliomas and secondary glioblastomas, but are rare in primary glioblastomas (Bales et al., Acta Neuropath. 116, pp 597-602 (2008)). Mutations in IDH2 also have been detected albeit at a lower frequency of 5-10% (Bales, Bales et al. Acta Neuropath. 116, pp. 597-602 (2008); Yan et al., N. Eng
    Figure US20150374800A1-20151231-P00999
    J. Med. 360, pp. 765-773 (2009)). IDH mutations play a central role in glioma pathogenesis (Parsons et al, Science 321, pp 1807-1812 (2008)) and have been shown to constitute a ke
    Figure US20150374800A1-20151231-P00999
    classifier distinguishing two major glioma subgroups that were identified initially based on RNA expression and DN
    Figure US20150374800A1-20151231-P00999
    methylation patterns. It has been revealed that brain tumor showing an IDH1 mutation (IDHmut) have a better prognosis that those where IDH1 is not mutated (IDHwt) (Hartmann et al., Act
    Figure US20150374800A1-20151231-P00999
    Neuropathol. 120, pp. 707-718 (2010)). The inventors found out that one difference between IDHm
    Figure US20150374800A1-20151231-P00999
    (having IDH1-R132H mutation) and IDHwt brain tumors is that BCAT1 overexpression is a highly specific feature of IDH
    Figure US20150374800A1-20151231-P00999
    glioblastomas, the most common and most aggressive adult brain tumor. The observed specific methylation of the BCAT1 promoted in IDHmut tumors, but not in IDHwt tumors and normal brain strongly show that low BCAT1 expression in IDHmut tumors is consequence of IDH1 mutation-associated DNA methylation, which is thought to be mediated through inhibition of histon
    Figure US20150374800A1-20151231-P00999
    demethylases and the TET family of 5-methylcytosin hydroxylases by the product of mutant IDH1 and IDH2 enzymes the oncometabolite 2-hydroxyglutarate. Interestingly regulation of BCAT1 mRNA expression originating from promote appears to be achieved by an IDH-independent epigeneti
    Figure US20150374800A1-20151231-P00999
    mechanism, through methylation of a bindung site for the HEY transcriptional repressor in IDHwt but not IDHmut tumors. These diametrically opposed patterns of DNA methylation in the two promoters make clear that BCAT1 suppression does not occur a
    Figure US20150374800A1-20151231-P00999
    a mere byproduct of IDH1 mutation through “passenger methylation, but rather that the differential regulation of cell metabolism in IDHwt and IDHmut tumors requires tight control of BCAT1 expression in each group.
  • Similar to the commonly used diagnostic IDH1-R132H staining BCAT1 staining may help to distinguish IDHwt from IDHm gliomas; however, BCAT1 staining offers the added advantage c
    Figure US20150374800A1-20151231-P00999
    also distinguishing IDHwt primary glioblastoma from the clos
    Figure US20150374800A1-20151231-P00999
    to 10% of IDHmut astrocytomas with non-IDH1-R132H mutations Most importantly, the present invention shows that BCAT1 an
    Figure US20150374800A1-20151231-P00999
    BCAA metabolism provide the basis for the development of nove
    Figure US20150374800A1-20151231-P00999
    metabolism-based approaches in glioma therapy.
  • This means that suppression of BCAT1 in IDHwt glioblastomas has the potential to significantly impede tumor growth as well a
    Figure US20150374800A1-20151231-P00999
    the excretion of glutamate by the tumor cells, which frequently causes neurotoxicity to surrounding brain tissue and leads to tumor-associated epilepsy in brain tumor patients.
  • In addition, the data of the present invention show that availability of large amounts of glucose and glutamine, the two nutrients considered to be most important for supporting malignant cell growth, is not sufficient to support sustained fast growth of IDHwt glioblastoma.
  • In other words, in the present invention it has been shown that BCAT1 overexpression is a highly characteristic feature of glioblastoma, in particular IDHwt glioblastoma, and essential for their aggressive clinical behavior. Thus, BCAT1 expression and BCAA catabolism are promising markers for the diagnostic and prognostic assessment of gliomas and serve a
    Figure US20150374800A1-20151231-P00999
    novel therapeutic targets. Furthermore, the present invention represents the first example of silencing of a metabolic gen
    Figure US20150374800A1-20151231-P00999
    that is central to glioma pathomechanism by IDH1 mutation associated aberrant DNA methylation. Silencing of BCAT1 earl in tumor development will prevent IDHmut gliomas from utilizing BCAAs as a metabolic resource and offers an explanation for the less malignant growth behaviour of IDHmut gliomas relative to the BCAT1-dependent IDHwt glioblastomas.
  • The reduction, silencing or inhibition of the biological activity can be effected by direct interaction or binding of compound to BCAT1 or by indirect interaction, e.g., b
    Figure US20150374800A1-20151231-P00999
    interacting with a compound that is associated with the biological activity of BCAT1. The reduction or inhibition of the biological activity can also be achieved by the application of altered, e.g., inactive forms of BCAT1 preferably in excess.
  • Examples of suitable compounds reducing, silencing o
    Figure US20150374800A1-20151231-P00999
    inhibiting the biological activity of BCAT1 or the expression of the gene encoding BCAT1 with the aim to get a therapeutic effect are:
  • (a) Plasmids, vectors or natural/synthetic/mutated viruses oligonucleotides of various types of modification (e.g. PTO LNA, 2′F-ANA, protein-nucleotide complexes, RNAi, siRNA o
    Figure US20150374800A1-20151231-P00999
    mikromiRNA, shRNA, Methylmethoxy-, Phosphoroamidates, PNA Morpholino, Phosphoramidate, Cyclohexen (CeNA), gap-meres ribozymes, aptamers, CpG-oligos, DNA-zymes, riboswitches, o
    Figure US20150374800A1-20151231-P00999
    lipids or lipid containing molecules;
    (b) peptides, peptide complexes, including all types of linkers,
    (c) small molecules;
    (d) antibodies and their derivatives, especially chimeras Fab-fragments, Fc-fragments, or
    (e) carriers, liposomes, nanoparticles, complexes, or an other delivery systems containing the above named constructs,
    (f) oxidizing agents or sulfhydryl (SH groups) modifying agents.
  • Further compounds suitable for the purposes of the present invention and methods how to identify/select such compound are in more detail described below.
  • Preferably, in a pharmaceutical composition, such compounds as described above are combined with a pharmaceuticall acceptable carrier. “Pharmaceutically acceptable” is meant to encompass any carrier, which does not interfere with the effectiveness of the biological activity of the activ
    Figure US20150374800A1-20151231-P00999
    ingredient and that is not toxic to the host to which it is administered. Examples of suitable pharmaceutical carriers are well known in the art and include phosphate buffered salin
    Figure US20150374800A1-20151231-P00999
    solutions, water, emulsions, such as oil/water emulsions various types of wetting agents, sterile solutions etc. Suc
    Figure US20150374800A1-20151231-P00999
    carriers can be formulated by conventional methods and the active compound can be administered to the subject at an effective dose.
  • An “effective dose” refers to an amount of the activ
    Figure US20150374800A1-20151231-P00999
    ingredient that is sufficient to affect the course and the severity of the neoplasia, leading to the reduction of remission of such a pathology. An “effective dose” useful for treating and/or preventing neoplasias may be determined using methods known to one skilled in the art (see for example Fingl et al., The Pharmocological Basis of Therapeutics Goodman and Gilman, eds. Macmillan Publishing Co., New York pp. 1-46 ((1975)).
  • Administration of the suitable compositions may be effected b
    Figure US20150374800A1-20151231-P00999
    different ways, e.g. by intravenous, intraperitoneal subcutaneous, intramuscular, topical or intraderma
    Figure US20150374800A1-20151231-P00999
    administration. The route of administration, of course depends on the kind of therapy and the kind of compound contained in the pharmaceutical composition. The dosage regimen will be determined by the attending physician and other clinical factors. As is well known in the medical arts dosages for any one patient depends on many factors, including the patient's size, body surface area, age, sex, th
    Figure US20150374800A1-20151231-P00999
    particular compound to be administered, time and route o
    Figure US20150374800A1-20151231-P00999
    administration, the kind of therapy, general health and of the drugs being administered concurrently.
  • The person skilled in the art can easily identify or generate compounds useful for the treatments of the present invention based on the knowledge of the amino acid sequence of BCAT1 and the nucleotide sequence of the gene encoding this protein Respective sequences are found in the UniProtKB/Swiss-Pro
    Figure US20150374800A1-20151231-P00999
    database (P54687; BCAT1 HUMAN), in Genbank (NCBI Reference Sequence: NM005504) and the Human Genome Organization Gen
    Figure US20150374800A1-20151231-P00999
    Nomenclature Committee (HGNC) database (HGNC ID: 976).
  • In a further preferred embodiment of the present invention the compound useful for reducing or inhibiting the expression of the gene encoding BCAT1 is an antisense oligonucleotide shRNA or siRNA specific for BCAT1. Preferably, the compound useful for silencing the BCAT1 expression are independent Mission® shRNA constructs targeting different regions of th human BCAT1 (BCAT1 shRNAI NM005504.3-1064s1c1 and BCAT shRNAII NM005504.3-751s1c1) and human IDH1 (IDH1 shRNA NM005896.2-1363s1c1 and IDH1 shRNAII NM005896.2-292s1c1 mRNA transcripts.
  • The generation of suitable antisense oligonucleotides include determination of a site or sites within the BCAT1 encoding gene for the antisense interaction to occur such that the desired effect, e.g., inhibition of the expression of the protein, will result. A preferred intragenic site is (a) the region encompassing the translation initiation or termination codon of the open reading frame (ORF) of the gene or (b)
    Figure US20150374800A1-20151231-P00999
    region of the mRNA which is a “loop” or “bulge”, i.e., no
    Figure US20150374800A1-20151231-P00999
    part of a secondary structure. If one or more target site have been identified, oligonucleotides are chosen which are sufficiently complementary to the target, i.e., hybridiz
    Figure US20150374800A1-20151231-P00999
    sufficiently well and with sufficient specificity, to give the desired effect. In the context of this invention “hybridization” means hydrogen bonding, which may be Watson Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding between complementary nucleoside or nucleotide bases
  • “Complementary” as used herein, refers to the capacity for precise pairing between two nucleotides. For example, if
    Figure US20150374800A1-20151231-P00999
    nucleotide at a certain position of an oligonucleotide i
    Figure US20150374800A1-20151231-P00999
    capable of hydrogen bonding with a nucleotide at the same position of a DNA or RNA molecule, then the oligonucleotide and the DNA or RNA are considered to be complementary to each other at that position. The oligonucleotide and the DNA or RNA are complementary to each other when a sufficient number of corresponding positions in each molecule are occupied by nucleotides which can make hydrogen bonds with each other Thus, “specifically hybridizable” and “complementary” are terms which are used to indicate a sufficient degree of complementarity or precise pairing such that stable an
    Figure US20150374800A1-20151231-P00999
    specific binding occurs between the oligonucleotide and the DNA or RNA target. It is understood in the art that the sequence of an antisense compound does not need to be 100 complementary to that of its target nucleic acid to be specifically hybridizable. An antisense compound i
    Figure US20150374800A1-20151231-P00999
    specifically hybridizable when binding of the compound to the target DNA or RNA molecule interferes with the normal function of the target DNA or RNA to cause a loss of utility, and there is a sufficient degree of complementarity to avoid non-specific binding of the antisense compound to non-targe sequences under conditions in which specific binding i
    Figure US20150374800A1-20151231-P00999
    desired, i.e., in the case of therapeutic treatment.
  • The skilled person can generate antisense compounds and siRNA or shRNAs according to the present invention on the basis of the known DNA sequence for BCAT1.
  • “Oligonucleotide” refers to an oligomer or polymer o
    Figure US20150374800A1-20151231-P00999
    ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) o
    Figure US20150374800A1-20151231-P00999
    mimetics thereof. This term includes oligonucleotides composed of naturally-occurring nucleobases, sugars and covalent internucleoside (backbone) linkages as well a
    Figure US20150374800A1-20151231-P00999
    oligonucleotides having non-naturally-occurring portions which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms because of desirable properties such as, for example, enhance
    Figure US20150374800A1-20151231-P00999
    cellular uptake, enhanced affinity for nucleic acid target an increased stability in the presence of nucleases. While antisense oligonucleotides are a preferred form of the antisense compound, the present invention comprehends other oligomeric antisense compounds, including but not limited to oligonucleotide mimetics such as are described below. The antisense compounds in accordance with this invention compris
    Figure US20150374800A1-20151231-P00999
    from about 8 to about 50 nucleobases (i.e. from about 8 t about 50 linked nucleosides). Particularly preferred antisense compounds are antisense oligonucleotides, even more preferable those comprising from about 15 to about 25 nucleobases Antisense compounds include ribozymes, external guide sequences (EGS), oligonucleotides (oligozymes), and other short catalytic RNAs or catalytic oligonucleotides which hybridize to the target nucleic acid and inhibit it
    Figure US20150374800A1-20151231-P00999
    expression.
  • Alternatively, the compound of the invention is a vector allowing to transcribe an antisense oligonucleotide of the invention, e.g., in a mammalian host. Preferably, such vector is a vector useful for gene therapy. Preferred vector useful for gene therapy are viral vectors, e.g. adenovirus herpes virus, vaccinia, or, more preferably, an RNA virus suc
    Figure US20150374800A1-20151231-P00999
    as a retrovirus. Even more preferably, the retroviral vector is a derivative of a murine or avian retrovirus. Examples o
    Figure US20150374800A1-20151231-P00999
    such retroviral vectors which can be used in the presen invention are: Moloney murine leukemia virus (MoMuLV), Harve
    Figure US20150374800A1-20151231-P00999
    murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV) and Rous sarcoma virus (RSV). Most preferably, a non-human primate retroviral vector is employed, such as th gibbon ape leukemia virus (GaLV), providing a broader hos
    Figure US20150374800A1-20151231-P00999
    range compared to murine vectors. Since recombinan retroviruses are defective, assistance is required in order to produce infectious particles. Such assistance can be provided e.g., by using helper cell lines that contain plasmid encoding all of the structural genes of the retrovirus unde
    Figure US20150374800A1-20151231-P00999
    the control of regulatory sequences within the LTR. Suitable helper cell lines are well known to those skilled in the art Said vectors can additionally contain a gene encoding selectable marker so that the transduced cells can b
    Figure US20150374800A1-20151231-P00999
    identified. Moreover, the retroviral vectors can be modified in such a way that they become target specific. This can be achieved, e.g., by inserting a polynucleotide encoding sugar, a glycolipid, or a protein, preferably an antibody Those skilled in the art know additional methods for generating target specific vectors. Further suitable vector and methods for in vitro- or in vivo-gene therapy ar
    Figure US20150374800A1-20151231-P00999
    described in the literature and are known to the person skilled in the art; see, e.g., WO 94/29469 or WO 97/00957.
  • In order to achieve expression only in the target organ, e.g. a brain tumor, the DNA sequences for transcription of the antisense oligonucleotides can be linked to a tissue specific promoter and used for gene therapy. Such promoters are well known to those skilled in the art (see e.g. Zimmermann et al. (1994) Neuron 12, 11-24; Vidal et al.; (1990) EMBO J. 9, 833 840; Mayford et al., (1995), Cell 81, 891-904; Pinkert et al. (1987) Genes & Dev. 1, 268-76).
  • Within an oligonucleotide structure, the phosphate groups are commonly referred to as forming the internucleoside backbon of the oligonucleotide. The normal linkage or backbone of RNA and DNA is a 3′ to 5′ phosphodiester linkage. Specific examples of preferred antisense compounds useful in the present invention include oligonucleotides containing modified backbones or non-natural internucleoside linkages Oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone. Modified oligonucleotide backbones which can result in increased stability are known to the person skilled in the art preferably such modification is a phosphorothioate linkage.
  • A preferred oligonucleotide mimetic is an oligonucleotide mimetic that has been shown to have excellent hybridizatio properties, and is referred to as a peptide nucleic aci
    Figure US20150374800A1-20151231-P00999
    (PNA). In PNA compounds, the sugar-backbone of a oligonucleotide is replaced with an amide containing backbone in particular an aminoethylglycine backbone. The nucleobase are retained and are bound directly or indirectly to az
    Figure US20150374800A1-20151231-P00999
    nitrogen atoms of the amide portion of the backbone (see e.g., Nielsen et al., Science 254 (1991), 1497-1500.)
  • Modified oligonucleotides may also contain one or more substituted or modified sugar moieties. Preferred oligonucleotides comprise one of the following at the 2 position: OH; F; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; O- S- or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C1 t C10 alkyl or C2 to C10 alkenyl and alkynyl. A particularly preferred modified sugar moiety is a 2′-O-methoxyethyl suga
    Figure US20150374800A1-20151231-P00999
    moiety.
  • Oligonucleotides of the invention may also include nucleobas modifications or substitutions. Modified nucleobases include other synthetic and natural nucleobases such as 5 methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine hypoxanthine, 2-aminoadenine, 6-methyl and other alky
    Figure US20150374800A1-20151231-P00999
    derivatives of adenine and guanine, 2-propyl and other alky
    Figure US20150374800A1-20151231-P00999
    derivatives of adenine and guanine, 2-thiouracil, 2 thiothymine and 2-thiocytosine etc., with 5-methylcytosin
    Figure US20150374800A1-20151231-P00999
    substitutions being preferred since these modifications have been shown to increase nucleic acid duplex stability.
  • Another modification of the oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide. Such moieties include lipid moieties such as a cholesterol moiety cholic acid, a thioether, a thiocholesterol, an aliphati
    Figure US20150374800A1-20151231-P00999
    chain, e.g., dodecandiol or undecyl residues, a phospholipid
    Figure US20150374800A1-20151231-P00999
    a polyamine or a polyethylene glycol chain, or adamantan
    Figure US20150374800A1-20151231-P00999
    acetic acid, a palmityl moiety, or an octadecylamine o
    Figure US20150374800A1-20151231-P00999
    hexylamino-carbonyl-oxycholesterol moiety.
  • The present invention also includes antisense compounds which are chimeric compounds. “Chimeric” antisense compounds o
    Figure US20150374800A1-20151231-P00999
    “chimeras,” in the context of this invention, are antisens compounds, particularly oligonucleotides, which contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of a oligonucleotide compound. These oligonucleotides typically contain at least one region wherein the oligonucleotide i
    Figure US20150374800A1-20151231-P00999
    modified so as to confer upon the oligonucleotide increase resistance to nuclease degradation, increased cellular uptake and/or increased binding affinity for the target nucleic acid An additional region of the oligonucleotide may serve as substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids. By way of example, RNase H is a cellular endonucleas
    Figure US20150374800A1-20151231-P00999
    which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target thereby greatly enhancing the efficiency of oligonucleotide inhibition of gene expression. Consequently, comparable results can often be obtained with shorter oligonucleotide when chimeric oligonucleotides are used, compared to phosphorothioate deoxyoligonucleotides hybridizing to the same target region. Chimeric antisense compounds of the invention may be formed as composite structures of two or more oligonucleotides, modified oligonucleotides, oligonucleoside and/or oligonucleotide mimetics as described above. Such compounds have also been referred to in the art as hybrids o
    Figure US20150374800A1-20151231-P00999
    gapmers.
  • In a further preferred embodiment of the present invention the compounds for use in a method of treating a neoplasia are compounds reducing or inhibiting the biological activity o
    Figure US20150374800A1-20151231-P00999
    BCAT1.
  • Such compounds are described in the art for medica
    Figure US20150374800A1-20151231-P00999
    indications differing from the indication of the present invention and, preferably, comprise compounds like 1 (aminomethyl)cyclohexaneacetic acid (gabapentin) or a compound described in Goto et al., The Journal of Biological Chemistry 280(44) (2005), 37246-56, Hu et al., Bioorganic & Medicinal Chemistry Letters 16 (2006), 2337-40, Caballero et al. Molecular Diversity 13 (2009), 493-500, U.S. Pat. No. 6,632,831, U.S. Pat. No. 6,809,119, and EP-B1 1 157 000.
  • Further examples of compounds capable of reducing o
    Figure US20150374800A1-20151231-P00999
    inhibiting the biological activity of BCAT1 are (neutralizing antibodies directed against BCAT1 or fragments thereof having substantially the same binding specificity. The term “antibody”, preferably, relates to antibodies which consists essentially of pooled monoclonal antibodies with different epitopic specificities, as well as distinct monoclonal antibody preparations. Monoclonal antibodies are made from a antigen containing, e.g., a fragment of BCAT1 by methods well known to those skilled in the art (see, e.g., Köhler et al. Nature 256 (1975), 495). As used herein, the term “antibody
    Figure US20150374800A1-20151231-P00999
    (Ab) or “monoclonal antibody” (Mab) is meant to include intact molecules as well as antibody fragments (such as, for example Fab and F(ab′).2 fragments) which are capable of specifically binding to protein. Fab and F(ab′)2 fragments lack the F
    Figure US20150374800A1-20151231-P00999
    fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding than an intact antibody. (Wahl et al., J. Nucl. Med. 24: 316 325 (1983)). Thus, these fragments are preferred, as well a
    Figure US20150374800A1-20151231-P00999
    the products of a FAB or other immunoglobulin expression library. Moreover, antibodies useful for the purposes of the present invention include chimerical, single chain, an
    Figure US20150374800A1-20151231-P00999
    humanized antibodies.
  • Alternatively, preferred compounds for the purpose of the invention are inactive versions of BCAT1 or nucleic acid sequences encoding inactive versions of BCAT1 that can be introduced according to the approaches/vectors describe above. Such inactive versions can be generated according to well known methods of mutagenesis. Such compounds can have therapeutic effect in the human body by displacing their functionally active counterpart, in particular when applied i
    Figure US20150374800A1-20151231-P00999
    excess. Analyses of potentially inactive versions of BCAT1 can be carried out by assaying the (reversible) transamination o
    Figure US20150374800A1-20151231-P00999
    branched-chain L-amino acids to branched-chain alpha-ket
    Figure US20150374800A1-20151231-P00999
    acids, e.g., by determining the production of glutamate
    Figure US20150374800A1-20151231-P00999
    Suitable assays are described in the literature and, e.g., i
    Figure US20150374800A1-20151231-P00999
    Example 39 of U.S. Pat. No. 6,809,119, Hu et al. (2006), an EP-B1 1 157 000.
  • In a preferred embodiment of the present invention the compound described in detail above is used in a method o
    Figure US20150374800A1-20151231-P00999
    treating an astrocytic brain tumor or glioblastoma.
  • The present invention also relates to a method for identifying a compound reducing or inhibiting the biological activity o
    Figure US20150374800A1-20151231-P00999
    BCAT1 and/or its expression, comprising the steps of:
      • (a) incubating a candidate compound with a test system comprising BCAT1 or its gene; and
      • (b) assaying a biological activity of BCAT1;
      • wherein an inhibition or loss of a biological activity o
        Figure US20150374800A1-20151231-P00999
        BCAT1, preferably compared to a test system in the absence of said test compound, is indicative of the presence of
        Figure US20150374800A1-20151231-P00999
        candidate compound having the desired property.
  • Step (b) can be carried out by assaying the (reversible transamination of branched-chain L-amino acids to branched chain alpha-keto acids using an assay as described above.
  • Examples of such candidate molecules include antibodies oligonucleotides, proteins, or small molecules. Such molecule can be rationally designed using known techniques.
  • Preferably, said test system used for screening comprise substances of similar chemical and/or physical properties most preferably said substances are almost identical. The compounds which can be prepared and identified according to use of the present invention may be expression libraries e.g., cDNA expression libraries, peptides, proteins, nucleic acids, antibodies, small organic compounds, ligands, hormones
    Figure US20150374800A1-20151231-P00999
    peptidomimetics, PNAs or the like.
  • WO 98/25146 describes further methods for screening libraries of complexes for compounds having a desired property especially, the capacity to agonize, bind to, or antagonize polypeptide or its cellular receptor. The complexes in such libraries comprise a compound under test, a tag recording a least one step in synthesis of the compound, and a tethe
    Figure US20150374800A1-20151231-P00999
    susceptible to modification by a reporter molecule Modification of the tether is used to signify that a comple
    Figure US20150374800A1-20151231-P00999
    contains a compound having a desired property. The tag can be decoded to reveal at least one step in the synthesis of such compound. Other methods for identifying compounds which interac
    Figure US20150374800A1-20151231-P00999
    with BCAT1 or nucleic acid molecules encoding such molecule are, for example, the in vitro screening with the phage display system as well as filter binding assays or “real time” measuring of interaction.
  • It is also well known to the person skilled in the art, that it is possible to design, synthesize and evaluate mimetics o
    Figure US20150374800A1-20151231-P00999
    small organic compounds that, for example, can act as substrate or ligand to BCAT1. For example, it has been described that D-glucose mimetics of hapalosin exhibited similar efficiency as hapalosin in antagonizing multidru
    Figure US20150374800A1-20151231-P00999
    resistance assistance-associated protein in cytotoxicity; se
    Figure US20150374800A1-20151231-P00999
    Dinh, J. Med. Chem. 41 (1998), 981-987.
  • All these methods can be used in accordance with the present invention to identify a compound reducing or inhibiting the biological activity of BCAT1 or its expression.
  • The gene encoding BCAT1 can also serve as a target for the screening of inhibitors. Inhibitors may comprise, for example proteins that bind to the mRNA of the genes encoding BCAT1 thereby destabilizing the native conformation of the mRNA an hampering transcription and/or translation. Furthermore methods are described in the literature for identifying nucleic acid molecules such as a RNA fragment that mimics the structure of a defined or undefined target RNA molecule t
    Figure US20150374800A1-20151231-P00999
    which a compound binds inside of a cell resulting in the retardation of the cell growth or cell death; see, e.g., W
    Figure US20150374800A1-20151231-P00999
    98/18947 and references cited therein. These nucleic acid molecules can be used for identifying unknown compounds o
    Figure US20150374800A1-20151231-P00999
    pharmaceutical interest, and for identifying unknown RNA targets for use in treating a disease. These methods an
    Figure US20150374800A1-20151231-P00999
    compositions can be used for identifying compounds useful to reduce expression levels of BCAT1.
  • The compounds which can be tested and identified according to the method of the invention may be expression libraries, e.g. cDNA expression libraries, peptides, proteins, nucleic acids antibodies, small organic compounds, hormones peptidomimetics, PNAs or the like (Milner, Nature Medicine (1995), 879-880; Hupp, Cell 83 (1995), 237-245; Gibbs, Cell 7
    Figure US20150374800A1-20151231-P00999
    (1994), 193-198 and references cited supra). Furthermore genes encoding a putative regulator of BCAT1 and/or which exert their effects up- or downstream of BCAT1 may be identified using, for example, insertion mutagenesis using for example, gene targeting vectors known in the art. Sai
    Figure US20150374800A1-20151231-P00999
    compounds can also be functional derivatives or analogues o
    Figure US20150374800A1-20151231-P00999
    known inhibitors. Such useful compounds can be for example transacting factors which bind to BCAT1 or regulator sequences of the gene encoding it. Identification o
    Figure US20150374800A1-20151231-P00999
    transacting factors can be carried out using standard method in the art. To determine whether a protein binds to the protein itself or regulatory sequences, standard native gel
    Figure US20150374800A1-20151231-P00999
    shift analyses can be carried out. In order to identify transacting factor which binds to the protein or regulator sequence, the protein or regulatory sequence can be used as a affinity reagent in standard protein purification methods, o
    Figure US20150374800A1-20151231-P00999
    as a probe for screening an expression library. Th
    Figure US20150374800A1-20151231-P00999
    identification of nucleic acid molecules which encod polypeptides which interact with BCAT1 can also be achieved, for example, as described in Scofield (Science 274 (1996), 2063 2065) by use of the so-called yeast “two-hybrid system”. In this system BCAT1 is linked to the DNA-binding domain of the GAL transcription factor. A yeast strain expressing this fusio
    Figure US20150374800A1-20151231-P00999
    polypeptide and comprising a lacZ reporter gene driven by a appropriate promoter, which is recognized by the GAL transcription factor, is transformed with a library of cDNA which will express plant proteins or peptides thereof fused t
    Figure US20150374800A1-20151231-P00999
    an activation domain. Thus, if a peptide encoded by one of the cDNAs is able to interact with the fusion peptide comprising peptide of BCAT1, the complex is able to direct expression o
    Figure US20150374800A1-20151231-P00999
    the reporter gene. In this way, BCAT1 and the gene encoding BCAT1 can be used to identify peptides and proteins interacting with BCAT1. It is apparent to the person skilled in the art that this and similar systems may then further be exploited for the identification of inhibitors.
  • The below example explains the invention in more detail bu
    Figure US20150374800A1-20151231-P00999
    are not construed as a limitation of the invention.
  • Example 1 General Methods
  • (a) RNA Isolation and Quantitative Real-Time PCR (qRT-PCR)
  • Total RNA was extracted using the AllPrep DNA/RNA/Protein Min
    Figure US20150374800A1-20151231-P00999
    Kit (Qiagen) according to the manufacturer's instructions FirstChoice® Human Brain Reference Total RNA from Ambio
    Figure US20150374800A1-20151231-P00999
    served as normal brain RNA pool (n=23 donors). Total RNA (50 ng) was reverse-transcribed using random primers an superscript II (Invitrogen, Karlsruhe, Germany) according to the manufacturer's instructions. Each complimentary DNA sample was analysed in triplicate with the Applied Biosystems Pris
    Figure US20150374800A1-20151231-P00999
    7900HT Fast Real-Time PCR System (Applied Biosystems, Foste
    Figure US20150374800A1-20151231-P00999
    City, Calif., USA) using Absolute SYBR Green ROX Mix (ABgene
    Figure US20150374800A1-20151231-P00999
    Epsom, UK). The relative amount of specific mRNA was normalised to ARF, B2M and TBP. Primer sequences are given in the following Table 1A.
  • TABLE 1A
    Expression primer
    BCAT1 Forward CAACTATGGAGAATGGTC
    (all isoforms) CTAAGCT
    Reverse TGTCCAGTCGCTCTCTTC
    TCTTC
    BCAT1 T1 Forward GCTACGACCCTTGGGATC
    (ENST00000261192) T
    BCAT1 T4 Forward GTGCCACTGCCGCTCTCT
    (ENST00000539282)
    BCAT1 T6 Forward TGGTTGTCTGAGCCTCCT
    (ENST00000538118) TT
    BCAT1 Exon
     2 Reverse AAGTCCCCACCACCTCTT
    TT
    BCAT1 Exon
     5 Reverse CCCATTCTTGATCCAATT
    TCA
    HEY1 Forward CGAGCTGGACGAGACCAT
    Reverse GAGCCGAACTCAAGTTTC
    CA
    ARF Forward GACCACGATCCTCTACAA
    GC
    Reverse TCCCACACAGTGAAGCTG
    ATG
    B2M Forward ACTGAATTCACCCCCACT
    GA
    Reverse CCTCCATGATGCTGCTTA
    CA
    TBP Forward GAACCACGGCACTGATTT
    TC
    Reverse CCCCACCATGTTCTGAAT
    CT
    Reverse AAATAACTTAACACCCCA
    ATCTAAAC
  • (b) Western Blot Analysis
  • All experiments involving the use of human tissues are carried out in line with the guidelines of the institutional revie
    Figure US20150374800A1-20151231-P00999
    board of the Medical Faculty at Heinrich Heine University Düsseldorf. Fresh frozen human tumor tissue samples and a non-neoplastic brain tissue sample were lysed in guanidin isothiocyanate solution (4M) using an ULTRA-TURRAX (IKA Staufen, Germany) and subjected to CsCl-ultracentrifugation Diafiltration of the obtained protein fractions was performed using the Amicon Ultra-0.5 centrifugal filter device (Millipore, Schwalbach, Germany; 3 kDa cut-off) essentially a described45. Total protein of cell lines was extracted using the AllPrep DNA/RNA/Protein Mini kit (Qiagen). 10 μg o
    Figure US20150374800A1-20151231-P00999
    protein were separated by 10% SDS-PAGE and transferred to PVD membranes (Millipore). The membrane was blocked in blocking solution (5% BSA in TBS, 0.1% Tween 20) and incubated with primary antibodies overnight at 4° C. Horseradish peroxidas
    Figure US20150374800A1-20151231-P00999
    (HRP)-conjugated secondary antibodies were incubated for hour at room temperature prior to the chemiluminescent detection of protein (ECL kit; GE Healthcare, Little Chalfont
    Figure US20150374800A1-20151231-P00999
    UK).
  • The following antibodies were used: monoclonal mouse antibod
    Figure US20150374800A1-20151231-P00999
    against α-tubulin (1:2000, #T9026, Sigma-Aldrich, St. Louis Mo. 63178, USA), monoclonal mouse antibody against BCAT (1:2000, ECA39, clone 51, #611270, BD Biosciences, San Jose Calif., USA), monoclonal rat antiserum against IDH1 (1:1, provided by A. von Deimling, DKFZ, Heidelberg, Germany; commercially available from Dianova, Hamburg, Germany), monoclonal mous
    Figure US20150374800A1-20151231-P00999
    antibody against HADHSC (1:500, #H00003033-M01, Abnova) monoclonal rabbit antibody against CDKN1B (1:1000, p27 Kip1 #3686, Cell Signaling Technology), monoclonal rabbit antibod against Akt (1:1000, #9272, Cell Signaling Technology) monoclonal rabbit antibody against phospho-Akt (Ser473 (1:1000, #193H12, Cell Signaling Technology), polyclona rabbit antibody against p70 S6 kinase (1:1000, #9202, Cell Signaling Technology), polyclonal rabbit antibody against phospho-p70 S6 kinase (Thr389) (1:1000, #9205, Cell Signaling Technology), HRP-conjugated anti-rat IgG (1:10000, provided by A. von Deimling, DKFZ, Heidelberg, Germany), HRP-conjugated horse anti-mouse IgG (1:5000, #7076, Cell Signaling Technology), HRP-conjugated goat anti-rabbit IgG (1:2000 #7074, Cell Signaling Technology).
  • (c) Immunohistochemical Staining
  • Tumor sections were deparaffinised using xylol and rehydrated in decreasing concentrations of ethanol. Antigen retrieval was performed by heating for 40 min in a steamer in 10 mM sodium citrate buffer (pH 6.0). Endogenous peroxidase was inactivate by incubating the tumor sections in 3% hydrogen peroxide Sections were incubated overnight with primary anti-BCAT (ECA39) monoclonal mouse antibody, clone 51 (BD Biosciences San Jose, Calif.) diluted 1:500 or mouse anti-human IDH1 R132 antibody (Dianova, Hamburg, Germany) diluted 1:20 in Dak
    Figure US20150374800A1-20151231-P00999
    REAL™ antibody diluent (Dako, Glostrup, Denmark). Staining for detection of bound antibody was performed according to standard protocols using the EnVision™ detection system (peroxidase/DAB+, rabbit/mouse) (Dako, Glostrup, Denmark) subsequent counterstaining was done using hematoxylin.
  • (d) DNA Methylation Analysis
  • DNA methylation was analyzed by MassARRAY technique. Briefly 500 ng genomic DNA was chemically modified with sodium bisulfite using the EZ methylation kit (Zymo Research according to the manufacturer's instructions. The bisulfite
    Figure US20150374800A1-20151231-P00999
    treated DNA was PCR-amplified with primers generating fou
    Figure US20150374800A1-20151231-P00999
    amplicons (A1-A4) from −990 bp to +612 bp around TSS of BCAT transcript 1 (T1, ENST00000261192), and three amplicons (A5 A7) of the promoter of BCAT1 transcript 4 (T4 ENST00000539282) and 6 (T6, ENST00000538118) from −198 bp t
    Figure US20150374800A1-20151231-P00999
    +63 bp. The amplicons were transcribed by T7 polymerase followed by T specific-RNAaseA cleavage. The digested fragments were quantified by mass-spectrometry. The prime sequences are given in Table 1B. DNA methylation standard (0%, 20%, 40%, 60%, 80%, and 100% methylated genomic DNA) were used to confirm the unbiased amplification of the amplicons.
  • TABLE 1B
    Methylation analysis primer
    A1 * Forward AAGTTTTTGTTGTGGAAGTTGTTTT
    Reverse CACCTAACCAACAATCATTAAACACTA
    A2 * Forward TTTGTTTGAGGGTATTGGAAGTAAT
    Reverse TAACTCCTACCCACCTCCCTACTAT
    A3 * Forward ATAGTAGGGAGGTGGGTAGGAGTTA
    Reverse AAACACTAAAACTACTCCCCCAAAC
    A4 * Forward GTTTGGGGGAGTAGTTTTAGTG
    Reverse CTCCCTACCAACTATATTTCTTA
    A5 * Forward ATTTATGAGGGTGTTAGATTTGGGT
    Reverse TTAAAAACTCCTCCCCAAAAAATAC
    A6 * Forward TTGTTTAGGTTTAGTATTTTTATGGG
    Reverse ACCATTTATAAAAAAATCTCCAAAA
    A7 * Forward AAATTATTATTAAGTAAATGTAGGTGGG
  • (e) Cell Culture
  • The human glioma cell lines U-87MG (HTB-14), LN-229 (CRL 2611), Hs683 (HTB-138) and U-373MG (HTB-17) were cultured i
    Figure US20150374800A1-20151231-P00999
    Dulbecco's Minimal Essential Medium (DMEM) supplemented with 10% fetal calf serum and 1% penicillin/streptomycin-mix. Cel lines were authenticated by short tandem repeat (STR analysis. The brain cancer stem-like cells NCH421k were established from primary glioblastoma patients undergoing surgical resection according to the research proposal approved by the institutional review board at the Medica
    Figure US20150374800A1-20151231-P00999
    Faculty, University of Heidelberg. They were characterized genotypically and phenotypically in a previous study (Campos
    Figure US20150374800A1-20151231-P00999
    B., et al. Differentiation therapy exerts antitumor effects o
    Figure US20150374800A1-20151231-P00999
    stem-like glioma cells. Clin Cancer Res 16, 2715-2728 (2010)) NCH421k cells were grown as floating aggregates (neurospheres
    Figure US20150374800A1-20151231-P00999
    on uncoated tissue culture dishes in DMEM/F-12 mediu
    Figure US20150374800A1-20151231-P00999
    containing 20% BIT serum-free supplement, basic fibroblas
    Figure US20150374800A1-20151231-P00999
    growth factor and epidermal growth factor at a concentration of 20 ng/ml each (all from Provitro, Berlin, Germany). HEK293 and HEK293 cells were maintained as monolayer cultures in DME
    Figure US20150374800A1-20151231-P00999
    containing 10% fetal calf serum without antibiotics.
  • All cell lines were cultivated at 37° C. in a humidified incubator with 5% CO2. For hypoxia experiments, cells were cultured at 1% O2 in a nitrogen-supplied C16 hypoxia incubato
    Figure US20150374800A1-20151231-P00999
    (Labotect, Goettingen, Germany).
  • (f) Chromatin Immunoprecipitation (ChIP)
  • ChIP was performed on HEK293 cells overexpressing flag-tagged HEY1, since the currently available antibodies against HEY were not specific for ChIP assays. Constructs for HEY overexpression were prepared using gateway compatible vector tagged with FLAG; pDest26 (C-terminal tag) and pDest11 (N
    Figure US20150374800A1-20151231-P00999
    terminal tag). 1 μg of either the control vectors or HEY expression vectors were transfected into 2.5×10̂6 HEK293 cell per 15 cm plate using TransIT®-LT1 (Mirus Bio LLC, Madison, W
    Figure US20150374800A1-20151231-P00999
    53711, USA) according to slightly modified manufacturer' instructions. The cells from two plates were harvested 4 hours after transfection. Chromatin was prepared using non
    Figure US20150374800A1-20151231-P00999
    ionic shearing buffers with Covaris S2, according to the manufacturer's protocol (Covaris Inc.). ChIP was performed using anti-FLAG antibody (Cell signaling #2368). The DNA recovered after ChIP was used for qRT-PCR with input chromati
    Figure US20150374800A1-20151231-P00999
    and mock immunoprecipitation (anti-IgG antibody, Diagenode Kch-803-015) serving as controls. qRT-PCR was performed i
    Figure US20150374800A1-20151231-P00999
    triplicate with SYBR green detection using primers listed i
    Figure US20150374800A1-20151231-P00999
    Table 2. Ratios of bound to input signals are reported.
  • TABLE 2
    Promoter primer
    PTPRD Forward GAGGAGGAGGAGAAAGAGCA
    Reverse GACAGAGCCTCGCCTCCT
    BCAT1_neg Forward TCCCTAGTCCTCCGTTCTGA
    Reverse ATTCCAAGGAGCATTTGTGC
    BCAT1_DMR(c1) Forward GAGGGTGACTAAGGAACTCTGG
    Reverse ATTGCCATTCCGTCATCACT
    BCAT1_c2 Forward GCTACGACCCTTGGGATCT
    Reverse TCGATTCACGCACACATTTT
    BHLHA15 Forward CCGAGGGCTCATTTGCAT
    Reverse CACCCGAGTTCCCAGCTC
  • (g) Transient Transfection of siRNA
  • HEK293T cells were transfected with siRNA duplexes from Ambio
    Figure US20150374800A1-20151231-P00999
    (HEY1: s23868-70) or Dharmacon (control non-targeting: D
    Figure US20150374800A1-20151231-P00999
    001810-10) using DhamaFect1 following slight modifications t
    Figure US20150374800A1-20151231-P00999
    the manufacturer's instructions (Dharmacon). Briefly, eac
    Figure US20150374800A1-20151231-P00999
    siRNA pool was diluted in 1× siRNA buffer (Dharmacon), wa
    Figure US20150374800A1-20151231-P00999
    mixed with RPMI, and then distributed into 6 wells of 96 wel
    Figure US20150374800A1-20151231-P00999
    plates. 1.2×10̂4 HEK293 or 9×10̂3 HEK293T cells were seeded o top of the siRNA/DharmaFECT mixture (the volume was 15 μl/well and 20 nM of siRNAs in final). After 48 hour incubation at 37° C., RNAs were isolated from the wells fo
    Figure US20150374800A1-20151231-P00999
    further analysis.
  • (h) Virus Production and Transduction
  • Lentiviral vectors were produced by cotransfection of HEK293 cells with the psPAX2 (Addgene 12260, Didier Trono, packaging vector), pMD2.G (Addgene 12259, Didier Trono, envelop
    Figure US20150374800A1-20151231-P00999
    plasmid), and pLKO.1 shRNA constructs (Sigma-Aldrich) Transfections were carried out using TransIT®-LT1 (Mirus Bi
    Figure US20150374800A1-20151231-P00999
    LLC) and virus was harvested at 48 and 72 hours after transfection and combined.
  • Infection of glioma cells with virus at an M.O.I. of 2 was carried out in the presence of 8 μg/ml of polybrene (Chemicon Billerica, Mass., USA). Virus-containing supernatant was removed after 24 hours and cells were split on day 3, day 5 and day
    Figure US20150374800A1-20151231-P00999
    after transduction. Two independent Mission® shRNA construct
    Figure US20150374800A1-20151231-P00999
    targeting different regions of the human BCATI (BCAT1 shRNA NM005504.3-1064s1c1 and BCAT1 shRNAII NM005504.3-751s1c1 and human IDH1 (IDH1 shRNAI NM005896.2-1363s1c1 and IDH
    Figure US20150374800A1-20151231-P00999
    shRNAII NM005896.2-292s1c1) mRNA transcripts were used Nontargeting shRNA (Mission SHC002) was used as a control. Quantification of BCAT1 and IDH1 knockdown was assessed b
    Figure US20150374800A1-20151231-P00999
    quantitative real-time PCR and Western blot.
  • (i) Treatment with Dimethyl-α-Ketoglutarate and Gabapentin
    Figure US20150374800A1-20151231-P00999
  • 5×10̂4 cells/well were seeded in 24-well plates in a total volume of 500 μl cell culture medium. The medium was removed 16 hours after seeding the cells, and replaced with 500μ medium containing 5 mM or 10 mM dimethyl-α-ketoglutarat
    Figure US20150374800A1-20151231-P00999
    (Dimethyl 2-oxoglutarate; Sigma-Aldrich) or 5 mM, 10 mM or 2 mM gabapentin (1-(Aminomethyl)-cyclohexane; Sigma-Aldrich) Corresponding volumes of 200 mM HEPES buffer (pH 7.4) were added to the respective control wells.
  • (j) Cell-Cycle Analysis and Detection of Apoptosis
  • Cell-cycle analysis was performed 20 hours after treatment with gabapentin and 6 days after lentiviral transduction Nicoletti buffer (0.1% sodium citrate, pH7.4, 0.05 % Triton X 100, 50 μg ml−1 propidium iodide) was added to the well
    Figure US20150374800A1-20151231-P00999
    containing both dead and living cells. After 4 hours in the dark at 4° C., DNA content was analysed by flow cytometry using FACS Canto II (BD Biosciences, San Jose, Calif., USA). FACS Div
    Figure US20150374800A1-20151231-P00999
    software was used to quantify the distribution of cells i
    Figure US20150374800A1-20151231-P00999
    each cell-cycle phase: sub-G1 (dead cells), G1, S, and G2/M. For investigation of apoptotic activity after lentivira
    Figure US20150374800A1-20151231-P00999
    knockdown NCH421k cells were incubated with annexin V-P (Phycoerythrin) and 7-AAD (7-aminoactinomycin D, B
    Figure US20150374800A1-20151231-P00999
    Biosciences) for 15 minutes in the dark, immediately followed by flow cytometry.
  • (k) Proliferation Analysis
  • To assess the proliferation of glioma cells after treatment with gabapentin or after lentiviral knockdown, the Click-iT
    Figure US20150374800A1-20151231-P00999
    EdU cell proliferation assay (Invitrogen, Karlsruhe, Germany was used following the manufacturer's instructions. The cell were incubated with 10 μM of the nucleoside analog EdU (5 ethynyl-2′deoxyuridine) for 16 hours. Quantification of cell that incorporated EdU was performed using FACS Canto II (B
    Figure US20150374800A1-20151231-P00999
    Biosciences).
  • (l) Glutamate Quantification
  • Glutamate concentration in the supernatant of cells treated with gabapentin was determined using the glutamine/glutamat
    Figure US20150374800A1-20151231-P00999
    determination Kit (GLN-1; Sigma-Aldrich) according to the manufacturer's instructions. The reaction volumes were scaled down to 100 μl total volume and absorbance was measured in triplicate in a microplate (Corning® 96 Well Clear Flat Bottom UV-Transparent Microplate) using a Tecan Infinite M200 plat
    Figure US20150374800A1-20151231-P00999
    reader (Tecan, Austria). Data were normalised to the number o
    Figure US20150374800A1-20151231-P00999
    cells per well.
  • (m) Immunofluorescence Staining
  • Cells were plated on glass coverslips five days after lentiviral transduction. Cells were fixed in 4% formaldehyde rinsed twice in 1×PBS, and permeabilized in PBS containing 0.2% Triton. Following rinsing with 1×PBS cells were incubated in 10% goat serum for 30 minutes at room temperature. Samples were incubated 1 hour with the primary antibody (mouse anti α-tubulin antibody, 1:200, #T9026, Sigma Aldrich) and 1 hour at room temperature with the secondary antibody (FITC-conjugated goat anti mouse antibody, 1:100 ab6785, Abcam) following Mounting with DAPI-containing Vectashield mounting medium (Vector Laboratories, Burlingame Calif., USA). For fluorescence imaging, images were taken using 40× objective lense on a Zeiss Axioplan microscope.
  • (n) 3D Microchannel Migration Assay
  • Poly(dimethylsiloxane) (PDMS) based microchannel chips were kindly provided by Dr. Ralf Kemkemer (Max Planck Institute for Intelligent Systems, Germany). Microfabricated channe structures with the dimensions of 5×11×300 μm (W×H×L) were bio-functionalized by incubation with a 50 μg/ml fibronecti
    Figure US20150374800A1-20151231-P00999
    solution prior to use. The chip was fixed on a Teflon holde
    Figure US20150374800A1-20151231-P00999
    and 2×10̂5 cells were seeded on the chip in close proximity t
    Figure US20150374800A1-20151231-P00999
    the channels. After cells were attached on the chip, live-cell imaging was performed for 25 hours. During the experiments n
    Figure US20150374800A1-20151231-P00999
    chemical gradient or flow inside the channels was applied Phase-contrast time-lapse pictures of multiple positions were captured every 10 minutes with an automated inverted microscope (Zeiss Cell Observer; Carl Zeiss) equipped with a
    Figure US20150374800A1-20151231-P00999
    air-humidified and heated chamber. Images were recorded an
    Figure US20150374800A1-20151231-P00999
    processed with Zeiss AxioVision and ImageJ software. Cel
    Figure US20150374800A1-20151231-P00999
    behaviors were analyzed and categorized as reported previously (Bai, A. H., et al. MicroRNA-182 promotes leptomeningeal sprea
    Figure US20150374800A1-20151231-P00999
    of non-sonic hedgehog-medulloblastoma. Acta Neuropatho (2011); Rolli, C. G., Seufferlein, T., Kemkemer, R. & Spatz
    Figure US20150374800A1-20151231-P00999
    J. P. Impact of tumor cell cytoskeleton organization o
    Figure US20150374800A1-20151231-P00999
    invasiveness and migration: a microchannel-based approach PLoS One 5, e8726 (2010)).
  • (o) Animal Experiments
  • Animal work was approved by the governmental authorities (Regierungspraesidium Karlsruhe, Germany) and supervised b
    Figure US20150374800A1-20151231-P00999
    institutional animal protection officials in accordance with the National Institutes of Health guidelines Guide for the Care and Use of Laboratory Animals.
  • (p) Orthotopic Brain Tumor Model
  • A total of 2×10̂5 U-87MG cells with BCAT1 shRNAI knockdown o
    Figure US20150374800A1-20151231-P00999
    nontargeting shRNA were stereotactically implanted into the brain of six 7-9-week-old athymic mice (CD1 nu/nu; Charle
    Figure US20150374800A1-20151231-P00999
    River Laboratories, Wilmington, Mass., USA), respectively. Fou
    Figure US20150374800A1-20151231-P00999
    weeks after implantation, animals were sacrificed, brain removed and cryosectioned. Brain sections were stained with hematoxylin and eosin and the tumor volume was calculate using ImageJ.
  • (q) Perchloric Acid Cell Extraction and NMR Spectroscopy
  • Cells were harvested by trypsinization, washed once with ice cold PBS, and the cell pellet was frozen at −80° C. Typically 1×108 harvested cells were subjected to perchloric acid extraction followed by KOH neutralization according t
    Figure US20150374800A1-20151231-P00999
    published protocols. Briefly, 2 mL of ice-cold 1 N HClO4 were added to a frozen pellet of harvested cells. The pellet was disrupted using a Teflon pestle and a glass homogeniser. 1 m
    Figure US20150374800A1-20151231-P00999
    of ice-cold water was added to the lysate, vortexed for 90
    Figure US20150374800A1-20151231-P00999
    and centrifuged at 10000×g for 10 min at 4° C. The pellet was re-extracted and the supernatants were combined. The extrac
    Figure US20150374800A1-20151231-P00999
    was neutralised with KOH and the pH was adjusted to 6.5-7.0
    Figure US20150374800A1-20151231-P00999
    Precipitated KClO4 was pelleted at 25000×g for 20 min and the supernatant was lyophilised.
  • The lyophilised extract residues were dissolved in 0.5 ml of D2O buffer (99.9% D) containing 30 mM sodium phosphate and 0. mM sodium azide (pH 7.02). For quantitative analysis reference capillary (1.5 mm OD, 0.99 mm ID) was filled with solution of 11 mM trimethylsilyl-2,2,3,3-tetradeuteropropanoi
    Figure US20150374800A1-20151231-P00999
    acid (TSP, sodium salt) in the D2O buffer described above Subsequently, the capillary was sealed in the flame of Bunsen burner. A calibration solution was prepared by placin weighed amounts of glucose and citric acid in the D2O buffer t
    Figure US20150374800A1-20151231-P00999
    give the calculated concentrations of 10.49 mM glucose an 4.95 mm glucose. 1H-NMR spectra of the calibration solution capillary were acquired at 600 MHz (Avance AV-600, Bruke
    Figure US20150374800A1-20151231-P00999
    BioSpin GmbH) using a standard 5-mm NMR tube and a triple resonance inverse probe under the same conditions to be use for the extracts (20° C., presaturation of residual HDO signal repetition time TR=7 s, 30° flip angle). The signal integrals for citrate and glucose were set to value corresponding to the concentrations given above, and th
    Figure US20150374800A1-20151231-P00999
    integral of the TSP methyl signal was found to be equivalen
    Figure US20150374800A1-20151231-P00999
    to 4.60±0.10 mM protons. Cell extracts were measured wit
    Figure US20150374800A1-20151231-P00999
    the calibrated reference capillary inserted (512 transients i
    Figure US20150374800A1-20151231-P00999
    1 hour), and the 1H-NMR signals for the branched-chain amin
    Figure US20150374800A1-20151231-P00999
    acids, various metabolites, and the TSP reference wer
    Figure US20150374800A1-20151231-P00999
    integrated. The TSP integral was defined as 4.60 mM so tha
    Figure US20150374800A1-20151231-P00999
    the metabolite signal integrals gave directly the metabolit
    Figure US20150374800A1-20151231-P00999
    concentrations in mM (μmol/ml) for the 0.5 ml volumes used These data were then converted to femtomol/cell using the cel
    Figure US20150374800A1-20151231-P00999
    counts determined prior to extraction.
  • (r) Preparation of Cell Homogenates
  • Cell pellets were diluted in 100 μl water and homogenized b. sonification (Ultrasonic device, 3−5×20 cycle, output 80% Branson Sonifier 450, Dietzenbach, Germany). Protein wa
    Figure US20150374800A1-20151231-P00999
    determined according to Lowry (Lowry, O. H., Rosebrough, N. J. Farr, A. L. & Randall, R. J. Protein measurement with the Foli
    Figure US20150374800A1-20151231-P00999
    phenol reagent. J Biol Chem 193, 265-275 (1951) with th
    Figure US20150374800A1-20151231-P00999
    modifications of Helenius and Simons (Helenius, A. & Simons
    Figure US20150374800A1-20151231-P00999
    K. The binding of detergents to lipophilic and hydrophili
    Figure US20150374800A1-20151231-P00999
    proteins. J Biol Chem 247, 3656-3661 (1972)) using bovine seru
    Figure US20150374800A1-20151231-P00999
    albumin as a standard. The final protein concentrations of th
    Figure US20150374800A1-20151231-P00999
    homogenates should be in a range of 2-4 mg/ml. These dilution were used for all analyses.
  • (s) Statistical Analysis
  • The relationship between IDH1/IDH2 mutation status and BCAT protein expression (FIG. 1 k) was tested with the two-side Fisher's Exact Test. The Student' t test (two-tailed unpaired) was used for all other statistical comparisons. *,
    Figure US20150374800A1-20151231-P00999
    <0.05; **, P<0.01; ***, P<0.001.
  • Example 2 Inhibition of the Cell Proliferation by Gabapentin
  • The human glioblastoma cell lines U87-MG (HTB-14), HS683 (HTB 138) and U373-MG (HTB-17; LGC Standards, Teddington TW11 0LY
    Figure US20150374800A1-20151231-P00999
    U.K.) were cultured in Dulbecco's Minimal Essential Mediu
    Figure US20150374800A1-20151231-P00999
    supplemented with 10% fetal calf serum and 1 penicillin/streptomycin-mix. 5×104 cells/well were seeded in 2 well plates in a total volume of 500 μl cell culture medium The medium was removed 4 hours after seeding the cells, an
    Figure US20150374800A1-20151231-P00999
    replaced with 500 μl medium containing 1-(Aminomethyl)cyclohexylessigsäure (Gabapentin, dissolved in 200 mM HEPE buffer at a concentration of 500 mM) at final concentrations o
    Figure US20150374800A1-20151231-P00999
    5 mM, 10 mM and 20 mM, respectively. Corresponding volumes o
    Figure US20150374800A1-20151231-P00999
    200 mM HEPES were added to the respective control wells. hours after exchanging the medium, 5-ethynyl-2′-deoxyuridin
    Figure US20150374800A1-20151231-P00999
    (EdU; Carlsbad, Calif. 92008, USA) was added in order to measur
    Figure US20150374800A1-20151231-P00999
    cell proliferation using Invitrogens (Carlsbad, Calif. 92008, USA Click iT® proliferation kit. The cells were harvested 16 hour after EdU application and proliferation was measured according to the manufacturer's instructions using a BD Biosciences FAC
    Figure US20150374800A1-20151231-P00999
    Canto II flow cytometer (BD Biosciences, Franklin Lakes, N
    Figure US20150374800A1-20151231-P00999
    USA 07417). Three replicate measurements were obtained fo
    Figure US20150374800A1-20151231-P00999
    each treatment. In all cell lines a concentration-dependen
    Figure US20150374800A1-20151231-P00999
    significant reduction of cell proliferation of about 20-55 was observed in the gabapentin treated cells as compared t
    Figure US20150374800A1-20151231-P00999
    mock treated cells. The results are shown in FIG. 10.
  • Example 3 Inhibition of the Cell Proliferation by BCAT1 Antisense Oligonucleotides
  • Lentiviral vectors were produced by the cotransfection o
    Figure US20150374800A1-20151231-P00999
    HEK293T cells with the psPAX2 (Addgene 12260, Didier Trono
    Figure US20150374800A1-20151231-P00999
    packaging vector), pMD2.G (Addgene 12259, Didier Trono
    Figure US20150374800A1-20151231-P00999
    envelope plasmid), and pLKO.1 shRNA constructs (Sigma-Aldrich St. Louis, Mo. 63178, USA). Transfections were carried ou
    Figure US20150374800A1-20151231-P00999
    using TransIT®-LT1 (Mirus Bio LLC, Madison, Wis. 53711, USA) an virus was harvested at 48 and 72 hours after transfection. Tw
    Figure US20150374800A1-20151231-P00999
    independent shRNA constructs targeting different regions o
    Figure US20150374800A1-20151231-P00999
    the BCAT1 mRNA transcript were used: MISSION shRN
    Figure US20150374800A1-20151231-P00999
    TRCN0000005907 NM005504.3-1064s1c1 (shRNA1) an TRCN0000005909 NM005504.3-751s1c1 (shRNA2); all Sigma Aldrich, St. Louis, Mo. 63178, USA. Nontargeting shRNA was use as a control.
  • The human glioblastoma cell lines U87-MG (HTB-14), HS683 (HTB 138) and U373-MG (HTB-17; LGC Standards, Teddington TW11 0LY
    Figure US20150374800A1-20151231-P00999
    U.K.) were seeded in 24 well plates (5×104 cells/well) in total volume of 500 μl cell culture medium. After 24 hour cells were transduced with virus in the presence of 8 μg/ml o
    Figure US20150374800A1-20151231-P00999
    polybrene. Virus-containing supernatant was removed after 2 hours and cells were split on day 3 and day 5 afte
    Figure US20150374800A1-20151231-P00999
    transduction. Decreased BCAT1 mRNA and protein expression wa
    Figure US20150374800A1-20151231-P00999
    detected using quantitative real-time PCR and Western blo
    Figure US20150374800A1-20151231-P00999
    (ECA39 antibody, BD Biosciences Pharmingen, San Diego, Calif. 92121, USA). A proliferation assay was carried out on day 6 using the Click-iT® EdU cell proliferation kit after incubating cells with 10 μM EdU for 16 hours. In all of the BCAT1 shRNA transduced cells a reduction of proliferation was observed ranging from 20-80% depending on the cell line. These results are shown in FIG. 11.
  • Example 4 Determination of BCAT1 Overexpression in IDHwt Glioblastomas
  • Prediction analysis of microarrays on gene expression data from astrocytic gliomas of WHO grades II, III and IV identified BCAT1 as the best classifier distinguishing primary glioblastoma from other astrocytoma as can be seen in the following Table 3:
  • Rank Gene symbol AII_AAIII_sGBIV-score pGBIV-score
    1 BCAT1 −0.530 0.435
    2 CHI3L1 −0.503 0.413
    3 TIMP1 −0.492 0.404
    4 IGFBP2 −0.453 0.372
    5 PDPN −0.448 0.368
    6 SERPINE1 −0.442 0.363
    7 EMP3 −0.436 0.358
    8 ADM −0.413 0.338
    9 PTX3 −0.403 0.331
    10 COL6A2 −0.399 0.327
    11 NNMT −0.396 0.325
    12 LIF −0.393 0.323
    13 STEAP3 −0.371 0.304
    14 COL6A2 −0.371 0.304
    15 POSTN −0.361 0.296
    16 KCNE4 −0.359 0.295
    17 ABCC3 −0.348 0.286
    18 FABP5 −0.343 0.282
    19 LOX −0.342 0.281
    20 RANBP17 0.339 −0.278
    21 MOXD1 −0.337 0.276
    22 ADAM12 −0.336 0.276
    23 RBP1 −0.331 0.272
    24 OCIAD2 −0.330 0.270
    25 SAA2 −0.320 0.263
    26 FMOD −0.319 0.262
    27 PBEF1 −0.317 0.260
    28 ATP7B −0.316 0.259
    29 SOCS3 −0.315 0.259
    30 PLAT −0.314 0.258
    31 RARRES2 −0.312 0.256
    32 RAB34 −0.305 0.250
    33 VEGF −0.304 0.249
    34 RBP1 −0.303 0.249
    35 SAA1 −0.302 0.248
    36 NA 0.300 −0.246
    37 PCDH15 0.296 −0.243
    38 AL354720.14 −0.293 0.240
    39 PBEF1 −0.291 0.239
    40 EFEMP2 −0.291 0.238
    41 IL8 −0.288 0.236
    42 UPP1 −0.284 0.233
    43 ANXA2 −0.282 0.231
    44 TNFRSF12A −0.279 0.229
    45 ANGPT2 −0.273 0.224
    46 ANXA2 −0.272 0.224
    47 EMILIN2 −0.272 0.223
    48 TMEM158 −0.268 0.220
    49 VEGF −0.262 0.215
    50 PHYHIPL 0.262 −0.215
  • When classifying tumors based on IDH-mutation status, BCAT1 mRNA expression levels were significantly higher for IDHwt gliomas relative to IDHmut gliomas (FIG. 1 b; P<0.0001; two-tailed Student's t-test), whereas comparably enhanced levels were not observed for BCAT2 expression (FIG. 1 c; P=0.0301). Pathway analysis showed that, in addition to BCAT1, the RNA expression of several other genes of the BCAA catabolic pathway was upregulated in IDHwt compared to IDHmut tumors (FIG. 7). Consistent with the RNA expression results, Western blot analysis showed BCAT1 protein expression was high in IDHwt tumors, but essentially absent in IDHmut tumors, regardless of the specific mutation in either IDH1 or IDH2 (FIG. 1 d). Sequencing of both the promoter and coding regions of BCAT1 for 20 gliomas revealed neither nonsense nor putative activating mutations. The observed tight correlation between IDH1 or IDH2 mutation and BCAT1 expression was further confirmed through immunohistochemical staining of sections from 81 primary human gliomas (77 astrocytoma, 4 oligodendroglioma), among which 45 of 46 IDHwt tumors showed strong BCAT1 staining (FIG. 1 e), while 35 of 35 tumors with mutations in IDH1 (30 R132H, 1 R132C, 1 R132S) or IDH2 (3 R172K) were BCAT1 negative (FIG. 1 f-h); (P<0.0001; Fisher's Exact Test; FIG. 1 k). The BCAT1 staining pattern therefore is largely complementary to the pattern obtained with the widely used antibody against the IDH1-R132H mutant protein (FIG. 1 i,j). However, unlike IDH1-R132H staining, the BCAT1 antibody also distinguishes IDHwt tumors (FIG. 1 e) from tumors with less common IDH1 and IDH2 mutations that are not recognized by anti-IDH1-R132H (FIG. 1 g-h). These data show that high BCAT1 expression is a characteristic feature of IDHwt gliomas that can be used to positively identify these tumors in a diagnostic setting.
  • Example 5 Determination of Substrate-Dependent Expression of BCAT1
  • BCAT1 was found to be expressed strongly in the glioblastoma cell lines LN-229, U-87MG, U-373MG and to a lesser extent in A172 (FIG. 2 a), all of which were confirmed to carry IDH1 and IDH2 wildtype genes. BCAT1 expression also was elevated in the Hs683 cell line, which was originally derived from an oligodendroglioma but nevertheless displays an IDHwt genotype. Thus, all these cell lines can be considered suitable models for studying BCAT1 function. BCAT1 RNA and protein expression were upregulated under hypoxic conditions, which are frequently present in glioblastoma (FIG. 2 b). BCAT1 expression correlated with the concentration of the substrate alpha-KG and was upregulated after Increasing the concentration of cell-permeable dimethyl-alpha-KG-substrate in the culture medium (FIG. 2 c). Conversely, shRNA-mediated knockdown of IDH1, a major source of alpha-KG in the cytoplasm, led to strong downregulation of BCAT1 expression (FIG. 2 d).
  • Example 6 Differentially Regulated BCAT1 Expression Through DNA-Methylation in Two Alternative Promoters
  • To further elucidate the differential regulation of BCAT1 expression in IDHwt and IDHmut gliomas, transcript expression in patient samples was quantified. Using RT-PCR and sequencing, expression of three protein-coding BCAT1 transcripts listed in the Ensembl database in IDHwt astrocytic primary tumors as well as in a pool of 23 normal brain tissues (FIG. 9) were confirmed. These three transcripts (T1, T4 and T6) encode proteins of 386, 398, and 385 amino acids, all three of which correspond to the single protein band of 43 kD as identified by Western blot analysis. These transcripts originate from two alternative promoters (FIG. 3 a) and differ only in their first exons, which encode 2, 14 and 1 amino acid(s) in T1, T4 and T6, respectively. Transcript-specific qRT-PCR identified T6 as the predominant transcript representing 73% of all BCAT1 mRNA in primary tumors (FIG. 3 b). Notably, expression of all BCAT1 transcripts was significantly higher in IDHwt compared to IDHmut tumors.
  • To investigate the possible mechanisms of BCAT1 transcriptional regulation, quantitative DNA methylation analysis on astrocytomas of all grades using MassARRAY analysis of PCR products amplified from bisulfite-treated DNA covering the two alternative promoters (FIG. 3 c) were performed. Distinct methylation patterns were observed for IDHwt and IDHmut tumors. The major promoter (promoter 2) was hypermethylated in IDHmut tumors but mostly unmethylated in IDHwt tumors and normal brain (FIG. 3 c, right panel). The average degree of methylation of the A6 and A7 amplicons was strongly associated with IDH1 mutation status (FIG. 3 d). These data show the suppression of transcripts T4 and T6 by promoter-2 methylation in IDHmut tumors. Promoter 1 (FIG. 3 c, left panel) was mostly unmethylated in all tumor samples as well as in normal brain, with the exception of a stretch of hypermethylated sequences immediately upstream of the CpG island. Within this upstream region, three differentially methylated CpG-dinucleotides between IDHwt and IDHmut tumors at positions −699/−697 (CpG4;5) and −660 (CpG6) in amplicon A2 were identified. In contrast to the methylation pattern in promoter 2, CpG4;5 (FIG. 3 e) and CpG6 (FIG. 3 f) showed significantly less methylation in IDHmut than in IDHwt tumors. The degree of methylation in this differentially methylated region (DMR) correlated with the expression of the BCAT1 transcript T1 in IDHwt tumors supporting its functional relevance (FIG. 3 g). The observed CpG-specific methylation pattern is consistent with repressor binding to the DMR leading to the downregulation of T1. Binding of the repressor HEY1 to the BCAT1 promoter 1 but not promoter 2 (FIG. 3 a) previously had been suggested by ChIPseq data. Analysis of RNA expression data confirmed overexpression of the HEY1 repressor in astrocytic tumors compared to normal brain. Consistent with HEY1-repressor activity, siRNA-mediated HEY1 knockdown in HEK293T cells increased transcript-T1 expression (FIG. 3 h,i). ChIP analysis demonstrated that compared to an upstream control region and to a region close to the translation start site, the strongest binding of two different HEY1 constructs occurs in the DMR (FIG. 3 j,k). Together, these data strongly show differential expression of BCAT1 transcripts in astrocytomas is regulated by DNA methylation involving broad methylation of promoter 2 in IDHmut tumors and CpG-site specific methylation in a HEY1-repressor binding site in promoter 1 of IDHwt tumors.
  • Example 7 Reduction of the Release of Glutamate by Glioblastoma Cells Through the Suppression of BCAT1
  • To gain insight into the BCAT1 functional role in glioblastomas, cell lines U-87MG and U-373MG cell lines were treated with gabapentin, a leucine analog that specifically inhibits BCAT1, but not BCAT2. 1H-NMR spectroscopy of extracts of cells treated with 20 mM gabapentin for 20 hours demonstrated the intracellular accumulation of BCAAs, consistent with BCAT1 inhibition (FIG. 4 ab). Glioblastoma release high concentrations of glutamate which leads to neuronal death by an excitotoxic mechanism. Glutamate release was significantly reduced with inhibition of BCAT1 with gabapentin (FIG. 4 c) indicating that BCAT1 is a major contributor to glutamate production through BCAA catabolism in IDHwt glioma.
  • To independently confirm these findings, shRNA-mediated BCAT1 knockdown in U-87MG, U-373MG and Hs683 cells using two shRNAs that both target all three BCAT1 transcripts was performed. Tandem-MS analysis of the U-87MG cell culture media after 24 hour incubation revealed that glutamate release, as well as BCAA uptake were reduced in BCAT1 knockdown cells compared to control cells (FIG. 4 d). A significant increase in release of alanine, glycine and threonine was also observed. Quantification of intracellular amino acid concentrations revealed significant accumulations of aspartate, glycine and threonine (FIG. 4 e). A small but significant accumulation of glutamate also was observed following BCAT1 knockdown; however, considering the high ratio of media volume to intracellular volume, total concentration of glutamate is not significantly affected by this small intracellular accumulation. In the BCAT2 knockout mice the inhibition of BCAA catabolism results in high concentrations of BCAAs and higher rates of protein synthesis in peripheral tissues via U) the mechanistic target of rapamycin (mTOR) signaling pathway and compensatory increased protein degradation (increased protein turnover).
  • BCAT1 knockdown led to strong downregulation of 3-hydroxyacyl CoA dehydrogenase (HADH), an enzyme participating in the catabolism of valine and isoleucine downstream of BCAT1 (FIG. 4 f, g). Since HADH is central to fatty acid metabolism, BCAT1 knockdown would also alter synthesis or degradation of fatty acids essential for membrane synthesis.
  • Example 8 Limitation of Glioblastoma Migration in Microchannels by BCAT1 Knockdown
  • BCAT1 knockdown strongly affected cell morphology, resulting in a rounded, less extended appearance (FIG. 5 a-b). To test whether these morphology changes could affect the tumor cells' ability to invade adjacent tissues, a microchannel migration chip to simulate a three-dimensional environment was used (FIG. 5 c). Following BCAT1 knockdown, the majority of U-87MG cells (55%) penetrated short distances into the microchannels, but were unable to actively deform themselves in order to completely invade the microchannels whereas most control cells (78%) completely invaded the channels (FIG. 5 d). This reduced invasiveness of the BCAT1 knockdown cells might be due to altered membrane composition caused by the observed lower abundance of long-chain fatty acids and differences in cholesterol metabolism (FIG. 4 g); such changes might sufficiently affect general availability of membrane components as well as membrane elasticity to hinder cell invasion.
  • Example 9 BCAT1 is Essential for Glioblastoma Growth
  • To assess the impact of BCAT1 on tumor cell proliferation, inhibition and knockdown experiments were conducted. A concentration-dependent reduction of proliferation up to 56%, estimated based on EdU-incorporation, was observed, upon treatment with the inhibitor gabapentin (FIG. 6 a). Cell cycle analyses suggested that gabapentin treatment induced partial G1-arrest, indicated by the increased fraction of cells in G1-phase with concurrent decreases of the G2 and S-fractions (FIG. 6 b). ShRNA-mediated BCAT1 knockdown elicited similar effects (FIG. 6 c,d). BCAT1 knockdown decreased proliferation by 20-70% in all three cell lines (FIG. 6 c) and led to G1-arrest and strong increases in cellular CDKN1B/p27KIP1. Notably, the degree of CDKN1B/p27KIP1 accumulation showed a positive correlation with the size of the G1-fraction (FIG. 6 d). Cell death, indicated by the fraction of cells in sub-G1 phase, remained below 5% except in the case of Hs683, which showed a moderate increase as shown in the following Table 4
  • Cell line Treatment Average sub-G1 [%] STDEV sub-G1
    U87-MG nt shRNA 2.1 0.241
    BCAT1 shRNAI 1.4 0.100
    BCAT1 shRNAII 1.9 0.058
    20 mM Gabapentin 2.4 0.354
    Control (HEPES) 1.4 0.707
    U373-MG nt shRNA 2.4 0.283
    BCAT1 shRNAI 1.1 0.045
    BCAT1 shRNAII 2.7 0.141
    20 mM Gabapentin 2.8 0.283
    Control (HEPES) 1.4 0.212
    HS683 nt shRNA 13.3 1.344
    BCAT1 shRNAI 21.4 0.424
    BCAT1 shRNAII 16.7 2.263
    20 mM Gabapentin 10.1 0.849
    Control (HEPES) 2.2 0.424
  • Comparable reductions of cell proliferation were observed upon BCAT1 knockdown in glioblastoma primary spheroid cultures in serum-free media, except that these cells showed a higher rate of apoptosis as determined by Annexin V/7AAD staining (FIG. 9). Consistent with the observed reduction in proliferation, BCAT1 knockdown led to decreased phosphorylation of the v-akt murine thymoma viral oncogene homolog (AKT) in U-87MG, U-373MG and Hs683 cells (FIG. 6 e).
  • The effect of BCAT1 knockdown on tumor growth in vivo was evaluated by intracerebral transplantation of U-87MG cells into CD-1 nude mice (FIG. 6 f-h). Four weeks after transplantation of equal numbers of living cells, all 6 control mice, but only 1 of 6 mice transplanted with BCAT1-shRNA-transduced cells displayed neurologic symptoms such as lethargy or uncoordinated motor activities. Hematoxylin and eosin staining of mouse brain sections revealed large tumors in mice transplanted with control cells (FIG. 6 f) while significantly smaller tumors were found in mice transplanted with BCAT1-knockdown cells (FIG. 6 g). Quantitative analysis confirmed significant differences in tumor volume between the groups (FIG. 6 h, P=0.0091).

Claims (10)

1.-12. (canceled)
13. A pharmaceutical composition for treating a brain tumor, wherein said pharmaceutical composition comprises
an effective dose of a compound capable of reducing or inhibiting
(a) the biological activity of a branched-chain-aminotransferase-1 (BCAT1) or
(b) the expression of the gene encoding BCAT1,
wherein the effective dose leads to a reduction or inhibition of the proliferation of brain tumor cells in a subject,
and a pharmaceutically acceptable carrier.
14. The pharmaceutical composition of claim 13, wherein said compound is an antisense oligonucleotide or siRNA reducing or inhibiting the expression of the gene encoding BCAT1.
15. The pharmaceutical composition of claim 13, wherein said compound is a compound reducing or inhibiting the biological activity of BCAT1.
16. The pharmaceutical composition of claim 15, wherein said compound is 1-(aminomethyl)cyclohexaneacetic acid.
17. The pharmaceutical composition of claim 15, wherein said compound is an antibody directed against BCAT1 or a fragment thereof.
18. The pharmaceutical composition of claim 13, wherein said compound is an inactive version of BCAT1.
19. The pharmaceutical composition of claim 13, wherein said compound is, wherein the neoplasm to be treated shows BCAT1 (over) expression.
20. The pharmaceutical composition of claim 13, wherein said compound is, wherein said brain tumor to be treated is an astrocytic brain tumor, glioma or glioblastoma.
21. The pharmaceutical composition of claim 20, wherein said compound is, wherein the brain tumor is an IDHwt glioblastoma.
US14/752,280 2011-01-28 2015-06-26 Inhibitors of Branched-Chain-Aminotransferase-1 (BCAT1) for the Treatment of Brain Tumors Abandoned US20150374800A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/752,280 US20150374800A1 (en) 2011-01-28 2015-06-26 Inhibitors of Branched-Chain-Aminotransferase-1 (BCAT1) for the Treatment of Brain Tumors

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
EP11000720A EP2481801A1 (en) 2011-01-28 2011-01-28 Inhibitors of branched-chain-aminotransferase-1 (BCAT1) for the treatment of neoplasia
EP11000720.0 2011-01-28
PCT/EP2012/000378 WO2012100957A1 (en) 2011-01-28 2012-01-27 Inhibitors of branched-chain-aminotransferase-1 (bcat1) for the treatment of brain tumors
US201313982098A 2013-07-26 2013-07-26
US14/752,280 US20150374800A1 (en) 2011-01-28 2015-06-26 Inhibitors of Branched-Chain-Aminotransferase-1 (BCAT1) for the Treatment of Brain Tumors

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/EP2012/000378 Division WO2012100957A1 (en) 2011-01-28 2012-01-27 Inhibitors of branched-chain-aminotransferase-1 (bcat1) for the treatment of brain tumors
US13/982,098 Division US9241993B2 (en) 2011-01-28 2012-01-27 Inhibitors of branched-chain-aminotransferase-1 (BCAT1) for the treatment of brain tumors

Publications (1)

Publication Number Publication Date
US20150374800A1 true US20150374800A1 (en) 2015-12-31

Family

ID=43984084

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/982,098 Expired - Fee Related US9241993B2 (en) 2011-01-28 2012-01-27 Inhibitors of branched-chain-aminotransferase-1 (BCAT1) for the treatment of brain tumors
US14/752,280 Abandoned US20150374800A1 (en) 2011-01-28 2015-06-26 Inhibitors of Branched-Chain-Aminotransferase-1 (BCAT1) for the Treatment of Brain Tumors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US13/982,098 Expired - Fee Related US9241993B2 (en) 2011-01-28 2012-01-27 Inhibitors of branched-chain-aminotransferase-1 (BCAT1) for the treatment of brain tumors

Country Status (7)

Country Link
US (2) US9241993B2 (en)
EP (2) EP2481801A1 (en)
JP (2) JP6002152B2 (en)
CN (1) CN103380214B (en)
CA (1) CA2823774C (en)
ES (1) ES2565498T3 (en)
WO (1) WO2012100957A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10941109B2 (en) 2011-06-13 2021-03-09 Ergon Pharmaceuticals Llc Compositions and methods of treatment using a BCAT1 inhibitor
US9422224B2 (en) 2011-06-13 2016-08-23 Ergon Pharmaceuticals Llc Methods of treatment using a BCAT1 inhibitor
WO2017051347A2 (en) * 2015-09-23 2017-03-30 Pfizer Inc. Cells and method of cell culture
CA3052219A1 (en) 2017-02-01 2018-08-09 Hi-Stem Ggmbh Novel methods for sub-typing and treating cancer
WO2021063821A1 (en) * 2019-10-01 2021-04-08 Bayer Aktiengesellschaft Pyrimidinedione derivatives
CN116987680A (en) * 2022-04-25 2023-11-03 复旦大学 Functional acquisition mutant of branched-chain amino acid aminotransferase 1 and application thereof

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994029469A2 (en) 1993-06-07 1994-12-22 Vical Incorporated Plasmids suitable for gene therapy
JPH11509088A (en) 1995-06-23 1999-08-17 プレジデント アンド フェローズ オブ ハーバード カレッジ Transcriptional regulation of the gene encoding vascular endothelial growth factor receptor
WO1998018947A1 (en) 1996-10-31 1998-05-07 Smithkline Beecham Corporation Methods for the characterization and selection of rna target motifs that bind compounds of pharmaceutical use
US5958703A (en) 1996-12-03 1999-09-28 Glaxo Group Limited Use of modified tethers in screening compound libraries
MXPA01008085A (en) 1999-12-08 2002-04-24 Warner Lambert Co Branched chain amino aciddependent aminotransferase inhibitors and their use in the treatment of diabetic retinopathy.
MXPA02010430A (en) * 2001-11-27 2003-06-02 Warner Lambert Co Branched chain amino acid-dependent aminotransferase inhibitors and their use in the treatment of neurodegenerative diseases.
MXPA02010231A (en) 2001-11-27 2004-12-13 Warner Lambert Co Branched chain amino acid-dependent aminotransferase inhibitors and their use in the treatment of neurodegenerative diseases.
US7842467B1 (en) * 2005-05-12 2010-11-30 Celera Corporation Breast disease targets and uses thereof
JP2008118915A (en) * 2006-11-10 2008-05-29 Kazuto Nishio Utilization to diagnosis of gastric cancer, and drug discovery by identification of high gastric cancer-expressing gene

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Perry et al. (The Canadian Journal of Neurological Sciences. Le Journal Canadien des Sciences Neurologiques [1996, 23(2):128-131] *

Also Published As

Publication number Publication date
US9241993B2 (en) 2016-01-26
JP2014506566A (en) 2014-03-17
CA2823774A1 (en) 2012-08-02
WO2012100957A1 (en) 2012-08-02
EP2668274B1 (en) 2016-01-13
JP2016196486A (en) 2016-11-24
CA2823774C (en) 2017-11-21
WO2012100957A8 (en) 2013-04-11
EP2481801A1 (en) 2012-08-01
EP2668274A1 (en) 2013-12-04
CN103380214B (en) 2015-07-15
CN103380214A (en) 2013-10-30
JP6002152B2 (en) 2016-10-05
US20130310545A1 (en) 2013-11-21
ES2565498T3 (en) 2016-04-05

Similar Documents

Publication Publication Date Title
US20150374800A1 (en) Inhibitors of Branched-Chain-Aminotransferase-1 (BCAT1) for the Treatment of Brain Tumors
US20160186183A1 (en) Methods and compositions for treating malignant tumors associated with kras mutation
US10792299B2 (en) Methods and compositions for treating malignant tumors associated with kras mutation
US11352628B2 (en) Methods and compositions for treating malignant tumors associated with KRAS mutation
Rosenzweig et al. Related to testes-specific, vespid, and pathogenesis protein-1 (RTVP-1) is overexpressed in gliomas and regulates the growth, survival, and invasion of glioma cells
JP6078633B2 (en) Inhibitors of therapeutic thioredoxin interacting protein (TXNIP)
CN101348512B (en) Antineoplastic ad virus preparation
US11319541B2 (en) Anticancer therapeutic intervention
JP2021183618A (en) Cytostatic agent and pharmaceutical composition for treating or preventing cancer containing the same
US20210380988A1 (en) Reducing Prominin2-Mediated Resistance to Ferroptotic Cell Death
WO2009084668A1 (en) Method of inhibiting cancer cell proliferation, proliferation inhibitor and screening method
US20220275373A1 (en) Methods and compositions for treating malignant tumors associated with kras mutation
US20230287427A1 (en) Inhibition of lncExACT1 to Treat Heart Disease
KR101466661B1 (en) novel use of TMC5 gene
KR20220011698A (en) P2X7 receptor expression modulators
KR100992239B1 (en) Novel use of MIG12 gene
WO2018151840A2 (en) Methods and compositions for treating malignant tumors
KR20190113412A (en) Biomarker composition for diagnosis of glioma comprising TRIO and F-actin-binding protein
US20100292302A1 (en) Induction of apoptosis and inhibition of cell proliferation through modulation of carnitine palmitoyltransferase 1c activity

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION