WO2002098468A1 - Slc13a utiles en tant que modificateurs de la voie p53 et methodes d'utilisation - Google Patents

Slc13a utiles en tant que modificateurs de la voie p53 et methodes d'utilisation Download PDF

Info

Publication number
WO2002098468A1
WO2002098468A1 PCT/US2002/017460 US0217460W WO02098468A1 WO 2002098468 A1 WO2002098468 A1 WO 2002098468A1 US 0217460 W US0217460 W US 0217460W WO 02098468 A1 WO02098468 A1 WO 02098468A1
Authority
WO
WIPO (PCT)
Prior art keywords
slc13a
assay
agent
pathway
candidate
Prior art date
Application number
PCT/US2002/017460
Other languages
English (en)
Inventor
Lori Friedman
Gregory D. Plowman
Marcia Belvin
Helen Francis-Lang
Danxi Li
Roel P. Funke
Mario N. Lioubin
Original Assignee
Exelixis, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Exelixis, Inc. filed Critical Exelixis, Inc.
Publication of WO2002098468A1 publication Critical patent/WO2002098468A1/fr

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/5005Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells
    • G01N33/5008Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics
    • G01N33/5011Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving human or animal cells for testing or evaluating the effect of chemical or biological compounds, e.g. drugs, cosmetics for testing antineoplastic activity
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/527Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving lyase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57415Specifically defined cancers of breast
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57419Specifically defined cancers of colon
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57449Specifically defined cancers of ovaries
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4739Cyclin; Prad 1
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/82Translation products from oncogenes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/988Lyases (4.), e.g. aldolases, heparinase, enolases, fumarase
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/10Screening for compounds of potential therapeutic value involving cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2510/00Detection of programmed cell death, i.e. apoptosis

Definitions

  • the p53 gene is mutated in over 50 different types of human cancers, including familial and spontaneous cancers, and is believed to be the most commonly mutated gene in human cancer (Zambetti and Levine, FASEB (1993) 7:855-865; Hollstein, et al, Nucleic Acids Res. (1994) 22:3551-3555). Greater than 90% of mutations in the p53 gene are missense mutations that alter a single amino acid that inactivates p53 function.
  • the human p53 protein normally functions as a central integrator of signals including DNA damage, hypoxia, nucleotide deprivation, and oncogene activation (Prives, Cell (1998) 95:5-8). In response to these signals, p53 protein levels are greatly increased with the result that the accumulated p53 activates cell cycle arrest or apoptosis depending on the nature and strength of these signals. Indeed, multiple lines of experimental evidence have pointed to a key role for p53 as a tumor suppressor (Levine, Cell (1997) 88:323-331). For example, homozygous p53 "knockout" mice are developmentally normal but exhibit nearly 100% incidence of neoplasia in the first year of life (Donehower et al, Nature (1992) 356:215-221).
  • p53 function is its activity as a gene-specific transcriptional activator.
  • genes with known p53-response elements are several with well-characterized roles in either regulation of the cell cycle or apoptosis, including GADD45, p21/Wafl/Cipl, cyclin G, Bax, IGF- BP3, and MDM2 (Levine, Cell (1997) 88:323-331).
  • Inorganic sulfate is present in nearly all cell types and is abundant in plasma. This type of sulfate must enter into cells across hydrophobic membranes, which requires a transport system due to inorganic sulfate's hydrophilic nature (Lee, A. et al. (2000) Genomics 70: 354-363). Disturbances in inorganic sulfate supply and metabolism, which is regulated in the kidney, cause a variety of fatal syndromes and several congenital chondrodysplasias (Karniski, L. (2001) Hum. Molec. Genet 10: 1485-1490). Sodium-sulfate cotransporter 2 (SLC13A1) mediates sodium-dependent transport of sulfate, which is an important element for sulfate reabsorption in the kidney (Lee, A. et al. (2000) supra)
  • Mammalian sodium-dicarboxylate cotransporters which transport succinate and other Krebs cycle intermediates, fall into two categories based on their substrate affinity: low affinity (SLC13A2) and high affinity (SLC13A3). Both types of transporters play an important role in the handling of citrate by the kidneys (Pajor, A. et al (1996) Am. J. Physiol. 270: F642-F648). P element insertions in a Drosophila gene called Indy (for "I'm not dead yet") seem to double the average adult life span without a decline in fertility or physical activity. Indy shows 50% similarity to human and rat renal sodium dicarboxylate cotransporters. (Rogina, B. et al. (2000) Science 290: 2137-2140).
  • Sodium-dicarboxylate cotransporter couples three sodium ions to the transport of each divalent anion substrate.
  • SLC13A2 may possibly be involved in the development of kidney stones in humans by determining citrate concentrations in the urine. Urinary citrate inhibits calcium stone formation, and hypocitraturia is associated with the formation of kidney stones (Pajor, A. et al (1996) supra).
  • Human SLC13A2 sequence and function is evolutionarily conserved, and the properties of human SLC13A2 closely resemble those of the low-affinity SLC13A2 found on the apical membrane of the rabbit renal proximal tubule (Pajor, A. et al (1996) supra).
  • Sodium-dependent dicarboxylate transporter, member 3 is a transporter protein that mediates succinate, citrate and di-carboxylic acid sodium-dependent cotransport (Wang, H. et al. (2000) Am. J. Physiol. Cell Physiol. 278: C1019-C1030).
  • the ability to manipulate the genomes of model organisms such as Drosophila provides a powerful means to analyze biochemical processes that, due to significant evolutionary conservation, have direct relevance to more complex vertebrate organisms.
  • a genetic screen can be carried out in an invertebrate model organism having underexpression (e.g. knockout) or overexpression of a gene (referred to as a "genetic entry point") that yields a visible phenotype. Additional genes are mutated in a random or targeted manner. When a gene mutation changes the original phenotype caused by the mutation in the genetic entry point, the gene is identified as a "modifier" involved in the same or overlapping pathway as the genetic entry point.
  • SLC13A genes that modify the p53 pathway in Drosophila, and identified their human orthologs, hereinafter referred to as SLC13A.
  • the invention provides methods for utilizing these p53 modifier genes and polypep tides to identify candidate therapeutic agents that can be used in the treatment of disorders associated with defective p53 function.
  • Preferred SLC13A-modulating agents specifically bind to SLC13A polypeptides and restore p53 function.
  • Other preferred SLC13A-modulating agents are nucleic acid modulators such as antisense oligomers and RNAi that repress SLC13A gene expression or product activity by, for example, binding to and inhibiting the respective nucleic acid (i.e. DNA or mRNA).
  • SLC13A-specific modulating agents may be evaluated by any convenient in vitro or in vivo assay for molecular interaction with an SLC13A polypeptide or nucleic acid.
  • candidate p53 modulating agents are tested with an assay system comprising a SLC13A polypeptide or nucleic acid.
  • Candidate agents that produce a change in the activity of the assay system relative to controls are identified as candidate p53 modulating agents.
  • the assay system may be cell-based or cell-free.
  • SLC13A-modulating agents include SLC13A related proteins (e.g.
  • a small molecule modulator is identified using a transporter assay.
  • the screening assay system is selected from a binding assay, an apoptosis assay, a cell proliferation assay, an angiogenesis assay, and a hypoxic induction assay.
  • candidate p53 pathway modulating agents are further tested using a second assay system that detects changes in the p53 pathway, such as angiogenic, apoptotic, or cell proliferation changes produced by the originally identified candidate agent or an agent derived from the original agent.
  • the second assay system may use cultured cells or non-human animals.
  • the secondary assay system uses non-human animals, including animals predetermined to have a disease or disorder implicating the p53 pathway, such as an angiogenic, apoptotic, or cell ' proliferation disorder (e.g. cancer).
  • the invention further provides methods for modulating the p53 pathway in a mammalian cell by contacting the mammalian cell with an agent that specifically binds a SLC13A polypeptide or nucleic acid.
  • the agent may be a small molecule modulator, a nucleic acid modulator, or an antibody and may be administered to a mammalian animal predetermined to have a pathology associated the p53 pathway.
  • SLC13A modulating agents that act by inhibiting or enhancing SLC13A expression, directly or indirectly, for example, by affecting an SLC13A function such as transporter or binding activity, can be identified using methods provided herein.
  • SLC13A modulating agents are useful in diagnosis, therapy and pharmaceutical development.
  • Nucleic acids and polypeptides of the invention Sequences related to SLC13A nucleic acids and polypeptides that can be used in the invention are disclosed in Genbank (referenced by Genbank identifier (GI) number) as GI#s 14750418 (SEQ ID NO:l), 19923837 (SEQ ID NO:2), 10439271 (SEQ ID NO:4), 4506978 (SEQ ID NO:5), 17483941 (SEQ ID NO:7), 12383071 (SEQ ID NO:8), and 16552194 (SEQ ID NO: 11) for nucleic acid, and GI#s 14750419 (SEQ ID NO: 12), 2499523 (SEQ ID NO: 13), 4506979 (SEQ ID NO: 14), and 13653602 (SEQ ID NO: 15) for polypeptides. Additionally, nucleic acid sequences of SEQ ID NOs:6 and 10, and novel nucleic acid sequences of SEQ ID NOs: 3 and 9 can also be used in the invention
  • SLC13As are transmembrane proteins with sodium sulfate cotransporter domains.
  • the term "SLC13A polypeptide” refers to a full-length SLC13A protein or a functionally active fragment or derivative thereof.
  • a "functionally active" SLC13A fragment or derivative exhibits one or more functional activities associated with a full-length, wild- type SLC13A protein, such as antigenic or immunogenic activity, transport activity, ability to bind natural cellular substrates, etc.
  • SLC13A proteins, derivatives and fragments can be assayed by various methods known to one skilled in the art (Current Protocols in Protein Science (1998) Coligan et al, eds., John Wiley & Sons, Inc., Somerset, New Jersey) and as further discussed below.
  • functionally active fragments also include those fragments that comprise one or more structural domains of an SLC13A, such as a transporter domain or a binding domain. Protein domains can be identified using the PFAM program (Bateman A., et al., Nucleic Acids Res, 1999, 27:260-2; http://pfam.wustl.edu).
  • the sodium sulfate symporter transmembrane region domains (PFAM 00939) of SLC13A from GI#s 14750419 (SEQ ID NO:12), 4506979 (SEQ ID NO:14), and 13653602 (SEQ ID NO:15) are located at approximately amino acid residues 3 to 579, 26 to 569, and 7 to 573, respectively.
  • Methods for obtaining SLC13A polypeptides are also further described below.
  • preferred fragments are functionally active, domain- containing fragments comprising at least 25 contiguous amino acids, preferably at least 50, more preferably 75, and most preferably at least 100 contiguous amino acids of any one of SEQ ID NOs: 12, 13, 14, or 15 (an SLC13A).
  • the fragment comprises the entire transporter (functionally active) domain.
  • SLC13A nucleic acid refers to a DNA or RNA molecule that encodes a SLC13A polypeptide.
  • the SLC13A polypeptide or nucleic acid or fragment thereof is from a human, but can also be an ortholog, or derivative thereof with at least 70% sequence identity, preferably at least 80%, more preferably 85%, still more preferably 90%, and most preferably at least 95% sequence identity with SLC13A.
  • orthologs in different species retain the same function, due to presence of one or more protein motifs and/or 3-dimensional structures.
  • Orthologs are generally identified by sequence homology analysis, such as BLAST analysis, usually using protein bait sequences. Sequences are assigned as a potential ortholog if the best hit sequence from the forward BLAST result retrieves the original query sequence in the reverse BLAST (Huynen MA and Bork P, Proc Natl Acad Sci (1998) 95:5849-5856; Huynen MA et al, Genome Research (2000) 10:1204-1210).
  • Programs for multiple sequence alignment may be used to highlight conserved regions and/or residues of orthologous proteins and to generate phylogenetic trees.
  • CLUSTAL Thimpson JD et al, 1994, Nucleic Acids Res 22:4673-4680
  • orthologous sequences from two species generally appear closest on the tree with respect to all other sequences from these two species.
  • Structural threading or other analysis of protein folding may also identify potential orthologs.
  • a single gene in one species may correspond to multiple genes (paralogs) in another, such as human.
  • paralogs encompasses paralogs.
  • percent (%) sequence identity with respect to a subject sequence, or a specified portion of a subject sequence, is defined as the percentage of nucleotides or amino acids in the candidate derivative sequence identical with the nucleotides or amino acids in the subject sequence (or specified portion thereof), after aligning the sequences and introducing gaps, if necessary to achieve the maximum percent sequence identity, as generated by the program WU-BLAST-2.0al9 (Altschul et al, J. Mol. Biol.
  • HSP S and HSP S2 parameters are dynamic values and are established by the program itself depending upon the composition of the particular sequence and composition of the particular database against which the sequence of interest is being searched.
  • a % identity value is determined by the number of matching identical nucleotides or amino acids divided by the sequence length for which the percent identity is being reported. "Percent (%) amino acid sequence similarity" is determined by doing the same calculation as for determining % amino acid sequence identity, but including conservative amino acid substitutions in addition to identical amino acids in the computation.
  • Aromatic amino acids that can be substituted for each other are phenylalanine, tryptophan, and tyrosine; interchangeable hydrophobic amino acids are leucine, isoleucine, methionine, and valine; interchangeable polar amino acids are glutamine and asparagine; interchangeable basic amino acids are arginine, lysine and histidine; interchangeable acidic amino acids are aspartic acid and glutamic acid; and interchangeable small amino acids are alanine, serine, threonine, cysteine and glycine.
  • nucleic acid sequences are provided by the local homology algorithm of Smith and Waterman (Smith and Waterman, 1981, Advances in Applied Mathematics 2:482-489; database: European Bioinformatics Institute http://www.ebi.ac.uk/MPsrch/; Smith and Waterman, 1981, J. of Molec.Biol, 147:195- 197; Nicholas et al., 1998, "A tutorial on Searching Sequence Databases and Sequence Scoring Methods” (www.psc.edu) and references cited therein.; W.R. Pearson, 1991, Genomics 11:635-650).
  • This algorithm can be applied to amino acid sequences by using the scoring matrix developed by Dayhoff (Dayhoff: Atlas of Protein Sequences and Structure, M. O. Dayhoff ed., 5 suppl. 3:353-358, National Biomedical Research Foundation, Washington, D.C., USA), and normalized by Gribskov (Gribskov 1986 Nucl. Acids Res. 14(6):6745-6763).
  • the Smith-Waterman algorithm may be employed where default parameters are used for scoring (for example, gap open penalty of 12, gap extension penalty of two). From the data generated, the "Match" value reflects "sequence identity.”
  • Derivative nucleic acid molecules of the subject nucleic acid molecules include sequences that hybridize to the nucleic acid sequence of any of SEQ ID NOs:l, 2, 3, 4, 5, 6, 7, 8, 9, 10, or 11.
  • the stringency of hybridization can be controlled by temperature, ionic strength, pH, and the presence of denaturing agents such as formamide during hybridization and washing. Conditions routinely used are set out in readily available procedure texts (e.g., Current Protocol in Molecular Biology, Vol. 1, Chap. 2.10, John Wiley & Sons, Publishers (1994); Sambrook et al., Molecular Cloning, Cold Spring Harbor (1989)).
  • a nucleic acid molecule of the invention is capable of hybridizing to a nucleic acid molecule containing the nucleotide sequence of any one of SEQ ID NOs:l, 2, 3, 4, 5, ,6 ,7 ,8 ,9 10, or 11 under stringent hybridization conditions that comprise: prehybridization of filters containing nucleic acid for 8 hours to overnight at 65° C in a solution comprising 6X single strength citrate (SSC) (IX SSC is 0.15 M NaCl, 0.015 M Na citrate; pH 7.0), 5X Denhardt's solution, 0.05% sodium pyrophosphate and 100 ⁇ g/ml herring sperm DNA; hybridization for 18-20 hours at 65° C in a solution containing 6X SSC, IX Denhardt's solution, 100 ⁇ g/ml yeast tRNA and 0.05% sodium pyrophosphate; and washing of filters at 65° C for lh in a solution containing 0.2X SSC and 0.1% S
  • moderately stringent hybridization conditions comprise: pretreatment of filters containing nucleic acid for 6 h at 40° C in a solution containing 35% formamide, 5X SSC, 50 mM Tris-HCl (pH7.5), 5mM EDTA, 0.1% PVP, 0.1% Ficoll, 1% BSA, and 500 ⁇ g/ml denatured salmon sperm DNA; hybridization for 18-20h at 40° C in a solution containing 35% formamide, 5X SSC, 50 mM Tris-HCl (pH7.5), 5mM EDTA, 0.02% PVP, 0.02% Ficoll, 0.2% BSA, 100 ⁇ g/ml salmon sperm DNA, and 10% (wt/vol) dextran sulfate; followed by washing twice for 1 hour at 55° C in a solution containing 2X SSC and 0.1% SDS.
  • low stringency conditions can be used that comprise: incubation for 8 hours to overnight at 37° C in a solution comprising 20% formamide, 5 x SSC, 50 mM sodium phosphate (pH 7.6), 5X Denhardt's solution, 10% dextran sulfate, and 20 ⁇ g/ml denatured sheared salmon sperm DNA; hybridization in the same buffer for 18 to 20 hours; and washing of filters in 1 x SSC at about 37° C for 1 hour.
  • SLC13A nucleic acids and polypeptides useful for identifying and testing agents that modulate SLC13A function and for other applications related to the involvement of SLC13A in the p53 pathway.
  • SLC13A nucleic acids and derivatives and orthologs thereof may be obtained using any available method. For instance, techniques for isolating cDNA or genomic DNA sequences of interest by screening DNA libraries or by using polymerase chain reaction (PCR) are well known in the art.
  • PCR polymerase chain reaction
  • the particular use for the protein will dictate the particulars of expression, production, and purification methods. For instance, production of proteins for use in screening for modulating agents may require methods that preserve specific biological activities of these proteins, whereas production of proteins for antibody generation may require structural integrity of particular epitopes.
  • Proteins to be purified for screening or antibody production may require the addition of specific tags (e.g., generation of fusion proteins).
  • Overexpression of an SLC13A protein for assays used to assess SLC13A function, such as involvement in cell cycle regulation or hypoxic response, may require expression in eukaryotic cell lines capable of these cellular activities.
  • recombinant SLC13A is expressed in a cell line known to have defective p53 function (e.g. SAOS-2 osteoblasts, H1299 lung cancer cells, C33A and HT3 cervical cancer cells, HT-29 and DLD-1 colon cancer cells, among others, available from
  • the recombinant cells are used in cell-based screening assay systems of the invention, as described further below.
  • the nucleotide sequence encoding an SLC13A polypeptide can be inserted into any appropriate expression vector.
  • the necessary transcriptional and translational signals, including promoter/enhancer element, can derive from the native SLC13A gene and/or its flanking regions or can be heterologous.
  • a variety of host-vector expression systems may be utilized, such as mammalian cell systems infected with virus (e.g. vaccinia virus, adenovirus, etc.); insect cell systems infected with virus (e.g.
  • microorganisms such as yeast containing yeast vectors, or bacteria transformed with bacteriophage, plasmid, or cosmid DNA.
  • a host cell strain that modulates the expression of, modifies, and/or specifically processes the gene product may be used.
  • the expression vector can comprise a promoter operably linked to an SLC13A gene nucleic acid, one or more origins of replication, and, one or more selectable markers (e.g. thymidine kinase activity, resistance to antibiotics, etc.).
  • selectable markers e.g. thymidine kinase activity, resistance to antibiotics, etc.
  • recombinant expression vectors can be identified by assaying for the expression of the SLC13A gene product based on the physical or functional properties of the SLC13A protein in in vitro assay systems (e.g. immunoassays).
  • the SLC13A protein, fragment, or derivative may be optionally expressed as a fusion, or chimeric protein product (i.e.
  • a chimeric product can be made by ligating the appropriate nucleic acid sequences encoding the desired amino acid sequences to each other using standard methods and expressing the chimeric product.
  • a chimeric product may also be made by protein synthetic techniques, e.g. by use of a peptide synthesizer (Hunkapiller et al., Nature (1984) 310:105-111).
  • the gene product can be isolated and purified using standard methods (e.g. ion exchange, affinity, and gel exclusion chromatography; centrifugation; differential solubility; electrophoresis, cite purification reference).
  • native SLC13A proteins can be purified from natural sources, by standard methods (e.g. immunoaffinity purification). Once a protein is obtained, it may be quantified and its activity measured by appropriate methods, such as immunoassay, bioassay, or other measurements of physical properties, such as crystallography.
  • the methods of this invention may also use cells that have been engineered for altered expression (mis-expression) of SLC13A or other genes associated with the p53 pathway.
  • mis-expression encompasses ectopic expression, over-expression, under- expression, and non-expression (e.g. by gene knock-out or blocking expression that would otherwise normally occur).
  • Animal models that have been genetically modified to alter SLC13A expression may be used in in vivo assays to test for activity of a candidate p53 modulating agent, or to further assess the role of SLC13A in a p53 pathway process such as apoptosis or cell proliferation.
  • the altered SLC13A expression results in a detectable phenotype, such as decreased or increased levels of cell proliferation, angiogenesis, or apoptosis compared to control animals having normal SLC13A expression.
  • the genetically modified animal may additionally have altered p53 expression (e.g. p53 knockout).
  • Preferred genetically modified animals are mammals such as primates, rodents (preferably mice), cows, horses, goats, sheep, pigs, dogs and cats.
  • Preferred non- mammalian species include zebrafish, C. elegans, and Drosophila.
  • Preferred genetically modified animals are transgenic animals having a heterologous nucleic acid sequence present as an extrachromosomal element in a portion of its cells, i.e. mosaic animals (see, for example, techniques described by Jakobovits, 1994, Curr. Biol. 4:761-763.) or stably integrated into its germ line DNA (i.e., in the genomic sequence of most or all of its cells).
  • Heterologous nucleic acid is introduced into the germ line of such transgenic animals by genetic manipulation of, for example, embryos or embryonic stem cells of the host animal.
  • transgenic mice see Brinster et al., Proc. Nat. Acad. Sci. USA 82: 4438-4442 (1985), U.S. Pat. Nos. 4,736,866 and 4,870,009, both by Leder et al., U.S. Pat. No. 4,873,191 by Wagner et al., and Hogan, B., Manipulating the Mouse Embryo, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., (1986); for particle bombardment see U.S. Pat.
  • Clones of the nonhuman transgenic animals can be produced according to available methods (see Wilmut, I. et al. (1997) Nature 385:810-813; and PCT International Publication Nos. WO 97/07668 and WO 97/07669).
  • the transgenic animal is a "knock-out" animal having a heterozygous or homozygous alteration in the sequence of an endogenous SLC13A gene that results in a decrease of SLC13A function, preferably such that SLC13A expression is undetectable or insignificant.
  • Knock-out animals are typically generated by homologous recombination with a vector comprising a transgene having at least a portion of the gene to be knocked out. Typically a deletion, addition or substitution has been introduced into the transgene to functionally disrupt it.
  • the transgene can be a human gene (e.g., from a human genomic clone) but more preferably is an ortholog of the human gene derived from the transgenic host species.
  • a mouse SLC13A gene is used to construct a homologous recombination vector suitable for altering an endogenous SLC13A gene in the mouse genome.
  • homologous recombination vector suitable for altering an endogenous SLC13A gene in the mouse genome.
  • Detailed methodologies for homologous recombination in mice are available (see Capecchi, Science (1989) 244:1288-1292; Joyner et al, Nature (1989) 338:153-156). Procedures for the production of non-rodent transgenic mammals and other animals are also available (Houdebine and Chourrout, supra; Pursel et al., Science (1989) 244:1281-1288; Simms et al, Bio/Technology (1988) 6:179-183).
  • knock-out animals such as mice harboring a knockout of a specific gene, may be used to produce antibodies against the human counterpart of the gene that has been knocked out (Claesson MH et al., (1994) Scan J Immunol 40:257-264; Declerck PJ et al, (1995) J Biol Chem. 270:8397-400).
  • the transgenic animal is a "knock-in" animal having an alteration in its genome that results in altered expression (e.g., increased (including ectopic) or decreased expression) of the SLC13A gene, e.g., by introduction of additional copies of SLC13A, or by operatively inserting a regulatory sequence that provides for altered expression of an endogenous copy of the SLC13A gene.
  • a regulatory sequence include inducible, tissue-specific, and constitutive promoters and enhancer elements.
  • the knock-in can be homozygous or heterozygous.
  • Transgenic nonhuman animals can also be produced that contain selected systems allowing for regulated expression of the transgene.
  • cre/loxP recombinase system of bacteriophage PI (Lakso et al, PNAS (1992) 89:6232-6236; U.S. Pat. No. 4,959,317). If a cre/loxP recombinase system is used to regulate expression of the transgene, animals containing transgenes encoding both the Cre recombinase and a selected protein are required.
  • Such animals can be provided through the construction of "double" transgenic animals, e.g., by mating two transgenic animals, one containing a transgene encoding a selected protein and the other containing a transgene encoding a recombinase.
  • a recombinase system is the FLP recombinase system of Saccharomyces cerevisiae (O'Gorman et al. (1991) Science 251:1351-1355; U.S. Pat. No. 5,654,182).
  • both Cre-LoxP and Flp-Frt are used in the same system to regulate expression of the transgene, and for sequential deletion of vector sequences in the same cell (Sun X et al (2000) Nat Genet 25:83-6).
  • the genetically modified animals can be used in genetic studies to further elucidate the p53 pathway, as animal models of disease and disorders implicating defective p53 function, and for in vivo testing of candidate therapeutic agents, such as those identified in screens described below.
  • the candidate therapeutic agents are administered to a genetically modified animal having altered SLC13A function and phenotypic changes are compared with appropriate control animals such as genetically modified animals that receive placebo treatment, and/or animals with unaltered SLC13A expression that receive candidate therapeutic agent.
  • animal models having defective p53 function can be used in the methods of the present invention.
  • a p53 knockout mouse can be used to assess, in vivo, the activity of a candidate p53 modulating agent identified in one of the in vitro assays described below.
  • p53 knockout mice are described in the literature (Jacks et al., Nature 2001;410:1111-1116, 1043-1044; Donehower et al, supra).
  • the candidate p53 modulating agent when administered to a model system with cells defective in p53 function, produces a detectable phenotypic change in the model system indicating that the p53 function is restored, i.e., the cells exhibit normal cell cycle progression.
  • the invention provides methods to identify agents that interact with and/or modulate the function of SLC13A and/or the p53 pathway. Such agents are useful in a variety of diagnostic and therapeutic applications associated with the p53 pathway, as well as in further analysis of the SLC13A protein and its contribution to the p53 pathway. Accordingly, the invention also provides methods for modulating the p53 pathway comprising the step of specifically modulating SLC13A activity by administering a SLC13A-interacting or -modulating agent.
  • SLC13A-modulating agents inhibit or enhance SLC13A activity or otherwise affect normal SLC13A function, including transcription, protein expression, protein localization, and cellular or extra-cellular activity.
  • the candidate p53 pathway- modulating agent specifically modulates the function of the SLC13A.
  • the phrases "specific modulating agent”, “specifically modulates”, etc., are used herein to refer to modulating agents that directly bind to the SLC13A polypeptide or nucleic acid, and preferably inhibit, enhance, or otherwise alter, the function of the SLC13A.
  • the term also encompasses modulating agents that alter the interaction of the SLC13A with a binding partner or substrate (e.g. by binding to a binding partner of an SLC13A, or to a protein/binding partner complex, and inhibiting function).
  • Preferred SLC13A-modulating agents include small molecule compounds; SLC ISA- interacting proteins, including antibodies and other biotherapeutics; and nucleic acid modulators such as antisense and RNA inhibitors.
  • the modulating agents may be formulated in pharmaceutical compositions, for example, as compositions that may comprise other active ingredients, as in combination therapy, and/or suitable carriers or excipients. Techniques for formulation and administration of the compounds may be found in "Remington's Pharmaceutical Sciences” Mack Publishing Co., Easton, PA, 19 th edition.
  • Small molecules are often preferred to modulate function of proteins with enzymatic function, and/or containing protein interaction domains.
  • Chemical agents referred to in the art as "small molecule” compounds are typically organic, non-peptide molecules, having a molecular weight less than 10,000, preferably less than 5,000, more preferably less than 1,000, and most preferably less than 500.
  • This class of modulators includes chemically synthesized molecules, for instance, compounds from combinatorial chemical libraries. Synthetic compounds may be rationally designed or identified based on known or inferred properties of the SLC13A protein or may be identified by screening compound libraries.
  • modulators of this class are natural products, particularly secondary metabolites from organisms such as plants or fungi, which can also be identified by screening compound libraries for SLC13A-modulating activity. Methods for generating and obtaining compounds are well known in the art (Schreiber SL, Science (2000) 151: 1964-1969; Radmann J and Gunther J, Science (2000) 151:1947-1948).
  • Small molecule modulators identified from screening assays, as described below, can be used as lead compounds from which candidate clinical compounds may be designed, optimized, and synthesized. Such clinical compounds may have utility in treating pathologies associated with the p53 pathway.
  • the activity of candidate small molecule modulating agents may be improved several-fold through iterative secondary functional validation, as further described below, structure determination, and candidate modulator modification and testing.
  • candidate clinical compounds are generated with specific regard to clinical and pharmacological properties.
  • the reagents may be derivatized and re-screened using in vitro and in vivo assays to optimize activity and minimize toxicity for pharmaceutical development. Protein Modulators
  • SLC13A-interacting proteins are useful in a variety of diagnostic and therapeutic applications related to the p53 pathway and related disorders, as well as in validation assays for other SLC13A-modulating agents.
  • SLC13A-interacting proteins affect normal SLC13A function, including transcription, protein expression, protein localization, and cellular or extra-cellular activity.
  • SLC13A-interacting proteins are useful in detecting and providing information about the function of SLC13A proteins, as is relevant to p53 related disorders, such as cancer (e.g., for diagnostic means).
  • An SLC13A-interacting protein may be endogenous, i.e.
  • SLC13A-modulators include dominant negative forms of SLC13A-interacting proteins and of SLC13A proteins themselves.
  • Yeast two-hybrid and variant screens offer preferred methods for identifying endogenous SLC13A-interacting proteins (Finley, R. L. et al. (1996) in DNA Cloning- Expression Systems: A Practical Approach, eds. Glover D. & Hames B. D (Oxford University Press, Oxford, England), pp.
  • Mass spectrometry is an alternative preferred method for the elucidation of protein complexes (reviewed in, e.g., Pandley A and Mann M, Nature (2000) 405:837-846; Yates JR 3 rd , Trends Genet (2000) 16:5-8).
  • An SLC13A-interacting protein may be an exogenous protein, such as an SLC ISA-specific antibody or a T-cell antigen receptor (see, e.g., Harlow and Lane (1988) Antibodies, A Laboratory Manual, Cold Spring Harbor Laboratory; Harlow and Lane (1999) Using antibodies: a laboratory manual. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press). SLC13A antibodies are further discussed below.
  • an SLC13A-interacting protein specifically binds an SLC13A protein.
  • an SLC13A-modulating agent binds an SLC13A substrate, binding partner, or cofactor.
  • the protein modulator is an SLC 13 A specific antibody agonist or antagonist.
  • the antibodies have therapeutic and diagnostic utilities, and can be used in screening assays to identify SLC13A modulators.
  • the antibodies can also be used in dissecting the portions of the SLC13A pathway responsible for various cellular responses and in the general processing and maturation of the SLC13A.
  • Antibodies that specifically bind SLC13A polypeptides can be generated using known methods.
  • the antibody is specific to a mammalian ortholog of SLC13A polypeptide, and more preferably, to human SLC 13 A.
  • Antibodies may be polyclonal, monoclonal (mAbs), humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab').sub.2 fragments, fragments produced by a FAb expression library, anti- idiotypic (anti-Id) antibodies, and epitope-binding fragments of any of the above.
  • Epitopes of SLC13A which are particularly antigenic can be selected, for example, by routine screening of SLC13A polypeptides for antigenicity or by applying a theoretical method for selecting antigenic regions of a protein (Hopp and Wood (1981), Proc. Nati. Acad. Sci. U.S.A. 78:3824-28; Hopp and Wood, (1983) Mol. Immunol. 20:483-89; Sutcliffe et al., (1983) Science 219:660-66) to the amino acid sequence shown in any of SEQ ID NOs: 12, 13, 14, or 15.
  • Monoclonal antibodies with affinities of 10 8 M “1 preferably 10 9 M 1 to 10 10 M "1 , or stronger can be made by standard procedures as described (Harlow and Lane, supra; Goding (1986) Monoclonal Antibodies: Principles and Practice (2d ed) Academic Press, New York; and U.S. Pat. Nos. 4,381,292; 4,451,570; and 4,618,577).
  • Antibodies may be generated against crude cell extracts of SLC13A or substantially purified fragments thereof. If SLC13A fragments are used, they preferably comprise at least 10, and more preferably, at least 20 contiguous amino acids of an SLC13A protein.
  • SLC13A-specific antigens and/or immunogens are coupled to carrier proteins that stimulate the immune response.
  • the subject polypeptides are covalently coupled to the keyhole limpet hemocyanin (KLH) carrier, and the conjugate is emulsified in Freund's complete adjuvant, which enhances the immune response.
  • KLH keyhole limpet hemocyanin
  • An appropriate immune system such as a laboratory rabbit or mouse is immunized according to conventional protocols.
  • SLC13A-specific antibodies is assayed by an appropriate assay such as a solid phase enzyme-linked immunosorbant assay (ELISA) using immobilized corresponding SLC13A polypeptides.
  • an appropriate assay such as a solid phase enzyme-linked immunosorbant assay (ELISA) using immobilized corresponding SLC13A polypeptides.
  • ELISA enzyme-linked immunosorbant assay
  • Other assays such as radioimmunoassays or fluorescent assays might also be used.
  • Chimeric antibodies specific to SLC13A polypeptides can be made that contain different portions from different animal species.
  • a human immunoglobulin constant region may be linked to a variable region of a murine mAb, such that the antibody derives its biological activity from the human antibody, and its binding specificity from the murine fragment.
  • Chimeric antibodies are produced by splicing together genes that encode the appropriate regions from each species (Morrison et al., Proc. Natl. Acad. Sci. (1984) 81:6851-6855; Neuberger et al., Nature (1984) 312:604-608; Takeda et al., Nature (1985) 31:452-454).
  • Humanized antibodies which are a form of chimeric antibodies, can be generated by grafting complementary-determining regions (CDRs) (Carlos, T. M., J. M. Harlan. 1994. Blood 84:2068-2101) of mouse antibodies into a background of human framework regions and constant regions by recombinant DNA technology (Riechmann LM, et al., 1988 Nature 323: 323-327). Humanized antibodies contain ⁇ 10% murine sequences and ⁇ 90% human sequences, and thus further reduce or eliminate immunogenicity, while retaining the antibody specificities (Co MS, and Queen C. 1991 Nature 351: 501-501; Morrison SL. 1992 Ann. Rev. Immun. 10:239-265). Humanized antibodies and methods of their production are well-known in the art (U.S. Pat. Nos. 5,530,101, 5,585,089, 5,693,762, and 6,180,370).
  • SLC13A-specific single chain antibodies which are recombinant, single chain polypeptides formed by linking the heavy and light chain fragments of the Fv regions via an amino acid bridge, can be produced by methods known in the art (U.S. Pat. No. 4,946,778; Bird, Science (1988) 242:423-426; Huston et al., Proc. Natl. Acad. Sci. USA (1988) 85:5879-5883; and Ward et al., Nature (1989) 334:544-546).
  • T-cell antigen receptors are included within the scope of antibody modulators (Harlow and Lane, 1988, supra).
  • polypeptides and antibodies of the present invention may be used with or without modification. Frequently, antibodies will be labeled by joining, either covalently or non- covalently, a substance that provides for a detectable signal, or that is toxic to cells that express the targeted protein (Menard S, et al., Int J. Biol Markers (1989) 4:131-134).
  • labels and conjugation techniques are known and are reported extensively in both the scientific and patent literature. Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, fluorescent emitting lanthanide metals, chemiluminescent moieties, bioluminescent moieties, magnetic particles, and the like (U.S. Pat. Nos.
  • the antibodies of the subject invention are typically administered parenterally, when possible at the target site, or intravenously.
  • the therapeutically effective dose and dosage regimen is determined by clinical studies.
  • the amount of antibody administered is in the range of about 0.1 mg/kg -to about 10 mg/kg of patient weight.
  • the antibodies are formulated in a unit dosage injectable form (e.g., solution, suspension, emulsion) in association with a pharmaceutically acceptable vehicle.
  • a pharmaceutically acceptable vehicle are inherently nontoxic and non-therapeutic. Examples are water, saline, Ringer's solution, dextrose solution, and 5% human serum albumin.
  • Nonaqueous vehicles such as fixed oils, ethyl oleate, or liposome carriers may also be used.
  • the vehicle may contain minor amounts of additives, such as buffers and preservatives, which enhance isotonicity and chemical stability or otherwise enhance therapeutic potential.
  • the antibodies' concentrations in such vehicles are typically in the range of about 1 mg/ml to aboutlO mg/ml.
  • an SLC13A-interacting protein may have biotherapeutic applications.
  • Biotherapeutic agents formulated in pharmaceutically acceptable carriers and dosages may be used to activate or inhibit signal transduction pathways. This modulation may be accomplished by binding a ligand, thus inhibiting the activity of the pathway; or by binding a receptor, either to inhibit activation of, or to activate, the receptor.
  • the biotherapeutic may itself be a ligand capable of activating or inhibiting a receptor. Biotherapeutic agents and methods of producing them are described in detail in U.S. Pat. No. 6,146,628.
  • SLC13A its ligand(s), antibodies to the ligand(s) or the SLC13A itself may be used as biotherapeutics to modulate the activity of SLC13A in the p53 pathway.
  • SLC13A-modulating agents comprise nucleic acid molecules, such as antisense oligomers or double stranded RNA (dsRNA), which generally inhibit SLC13A activity.
  • Preferred nucleic acid modulators interfere with the function of the SLC13A nucleic acid such as DNA replication, transcription, translocation of the SLC13A RNA to the site of protein translation, translation of protein from the SLC13A RNA, splicing of the SLC13A RNA to yield one or more mRNA species, or catalytic activity which may be engaged in or facilitated by the SLC13A RNA.
  • the antisense oligomer is an oligonucleotide that is sufficiently complementary to an SLC13A mRNA to bind to and prevent translation, preferably by binding to the 5' untranslated region.
  • SLC13A-specific antisense oligonucleotides preferably range from at least 6 to about 200 nucleotides. In some embodiments the oligonucleotide is preferably at least 10, 15, or 20 nucleotides in length. In other embodiments, the oligonucleotide is preferably less than 50, 40, or 30 nucleotides in length.
  • the oligonucleotide can be DNA or RNA or a chimeric mixture or derivatives or modified versions thereof, single-stranded or double-stranded.
  • the oligonucleotide can be modified at the base moiety, sugar moiety, or phosphate backbone.
  • the oligonucleotide may include other appending groups such as peptides, agents that facilitate transport across the cell membrane, hybridization-triggered cleavage agents, and intercalating agents.
  • the antisense oligomer is a phosphothioate morpholino oligomer (PMO).
  • PMOs are assembled from four different morpholino subunits, each of which contain one of four genetic bases (A, C, G, or T) linked to a six-membered morpholine ring. Polymers of these subunits are joined by non-ionic phosphodiamidate intersubunit linkages. Details of how to make and use PMOs and other antisense oligomers are well known in the art (e.g. see W099/18193; Probst JC, Antisense Oligodeoxynucleotide and Ribozyme Design, Methods. (2000) 22(3): 271-281; Summerton J, and Weller D. 1997 Antisense Nucleic Acid Drag Dev. :7:187-95; US Pat. No. 5,235,033; and US Pat No. 5,378,841).
  • RNAi is the process of sequence-specific, post-transcriptional gene silencing in animals and plants, initiated by double-stranded RNA (dsRNA) that is homologous in sequence to the silenced gene.
  • dsRNA double-stranded RNA
  • Methods relating to the use of RNAi to silence genes in C. elegans, Drosophila, plants, and humans are known in the art (Fire A, et al., 1998 Nature 391:806-811; Fire, A. Trends Genet. 15, 358-363 (1999); Sharp, P. A. RNA interference 2001. Genes Dev. 15, 485-490 (2001); Hammond, S.
  • Nucleic acid modulators are commonly used as research reagents, diagnostics, and therapeutics. For example, antisense oligonucleotides, which are able to inhibit gene expression with seventeen specificity, are often used to elucidate the function of particular genes (see, for example, U.S. Pat. No. 6,165,790). Nucleic acid modulators are also used, for example, to distinguish between functions of various members of a biological pathway.
  • an SLC13A-specific nucleic acid modulator is used in an assay to further elucidate the role of the SLC13A in the p53 pathway, and/or its relationship to other members of the pathway.
  • an SLC13A-specific antisense oligomer is used as a therapeutic agent for treatment of p53- related disease states.
  • an "assay system” encompasses all the components required for performing and analyzing results of an assay that detects and/or measures a particular event.
  • primary assays are used to identify or confirm a modulator's specific biochemical or molecular effect with respect to the SLC13A nucleic acid or protein.
  • secondary assays further assess the activity of a SLC13A modulating agent identified by a primary assay and may confirm that the modulating agent affects SLC13A in a manner relevant to the p53 pathway. In some cases, SLC13A modulators will be directly tested in a secondary assay.
  • the screening method comprises contacting a suitable assay system comprising an SLC13A polypeptide with a candidate agent under conditions whereby, but for the presence of the agent, the system provides a reference activity (e.g. transporter activity), which is based on the particular molecular event the screening method detects.
  • a reference activity e.g. transporter activity
  • the type of modulator tested generally determines the type of primary assay.
  • screening assays are used to identify candidate modulators. Screening assays may be cell-based or may use a cell-free system that recreates or retains the relevant biochemical reaction of the target protein (reviewed in Sittampalam GS et al, Curr Opin Chem Biol (1997) 1:384-91 and accompanying references).
  • the term "cell-based” refers to assays using live cells, dead cells, or a particular cellular fraction, such as a membrane, endoplasmic reticulum, or mitochondrial fraction.
  • cell free encompasses assays using substantially purified protein (either endogenous or recombinantly produced), partially purified or crude cellular extracts.
  • Screening assays may detect a variety of molecular events, including protein-DNA interactions, protein-protein interactions (e.g., receptor-ligand binding), transcriptional activity (e.g., using a reporter gene), enzymatic activity (e.g., via a property of the substrate), activity of second messengers, immunogenicty and changes in cellular morphology or other cellular characteristics.
  • Appropriate screening assays may use a wide range of detection methods including fluorescent, radioactive, colorimetric, spectrophotometric, and amperometric methods, to provide a read-out for the particular molecular event detected.
  • Cell-based screening assays usually require systems for recombinant expression of SLC13A and any auxiliary proteins demanded by the particular assay. Appropriate methods for generating recombinant proteins produce sufficient quantities of proteins that retain their relevant biological activities and are of sufficient purity to optimize activity and assure assay reproducibility. Yeast two-hybrid and variant screens, and mass spectrometry provide preferred methods for determining protein-protein interactions and elucidation of protein complexes. In certain applications, when SLC13A-interacting proteins are used in screens to identify small molecule modulators, the binding specificity of the interacting protein to the SLC13A protein may be assayed by various known methods such as substrate processing (e.g.
  • binding equilibrium constants usually at least about 10 7 M _1 , preferably at least about 10 8 M "1 , more preferably at least about 10 9 M "1
  • immunogenicity e.g. ability to elicit SLC13A specific antibody in a heterologous host such as a mouse, rat, goat or rabbit.
  • binding may be assayed by, respectively, substrate and ligand processing.
  • the screening assay may measure a candidate agent's ability to specifically bind to or modulate activity of a SLC13A polypeptide, a fusion protein thereof, or to cells or membranes bearing the polypeptide or fusion protein.
  • the SLC13A polypeptide can be full length or a fragment thereof that retains functional SLC13A activity.
  • the SLC13A polypeptide may be fused to another polypeptide, such as a peptide tag for detection or anchoring, or to another tag.
  • the SLC13A polypeptide is preferably human SLC 13 A, or is an ortholog or derivative thereof as described above.
  • the screening assay detects candidate agent-based modulation of SLC13A interaction with a binding target, such as an endogenous or exogenous protein or other substrate that has SLC13A -specific binding activity, and can be used to assess normal SLC13A gene function.
  • screening assays are high throughput or ultra high throughput and thus provide automated, cost-effective means of screening compound libraries for lead compounds (Fernandes PB, Curr Opin Chem Biol (1998) 2:597-603; Sundberg SA, Curr Opin Biotechnol 2000, 11:47-53).
  • screening assays uses fluorescence technologies, including fluorescence polarization, time-resolved fluorescence, and fluorescence resonance energy transfer.
  • a variety of suitable assay systems may be used to identify candidate SLC 13 A and p53 pathway modulators (e.g. U.S. Pat. Nos. 5,550,019 and 6,133,437 (apoptosis assays); U.S. Pat. No. 6,020,135 (p53 modulation), among others). Specific preferred assays are described in more detail below.
  • Transporter assays carry a range of substrates, including nutrients, ions, amino acids, and drugs, across cell membranes.
  • Assays for modulators of transporters may use labeled substrates.
  • exemplary high throughput screens to identify compounds that interact with different peptide and anion transporters both use fluorescently labeled substrates; the assay for peptide transport additionally uses multiscreen filtration plates (Blevitt JM et al., J Biomol Screen 1999, 4:87-91; Cihlar T and Ho ES, Anal Biochem 2000, 283:49-55).
  • Apoptosis assays may be performed by terminal deoxynucleotidyl transferase-mediated digoxigenin-11-dUTP nick end labeling (TUNEL) assay.
  • TUNEL terminal deoxynucleotidyl transferase-mediated digoxigenin-11-dUTP nick end labeling
  • the TUNEL assay is used to measure nuclear DNA fragmentation characteristic of apoptosis ( Lazeb ik et al, 1994, Nature 371, 346), by following the incorporation of fluorescein-dUTP (Yonehara et al, 1989, J. Exp. Med. 169, 1747).
  • Apoptosis may further be assayed by acridine orange staining of tissue culture cells (Lucas, R., et al., 1998, Blood 15:4730-41).
  • An apoptosis assay system may comprise a cell that expresses an SLC13A, and that optionally has defective p53 function (e.g. p53 is over-expressed or under- expressed relative to wild-type cells).
  • a test agent can be added to the apoptosis assay system and changes in induction of apoptosis relative to controls where no test agent is added, identify candidate p53 modulating agents.
  • an apoptosis assay may be used as a secondary assay to test a candidate p53 modulating agents that is initially identified using a cell-free assay system.
  • An apoptosis assay may also be used to test whether SLC13A function plays a direct role in apoptosis. For example, an apoptosis assay may be performed on cells that over- or under-express
  • Cell proliferation and cell cycle assays may be assayed via bromodeoxyuridine (BRDU) incorporation.
  • BRDU bromodeoxyuridine
  • This assay identifies a cell population undergoing DNA synthesis by incorporation of BRDU into newly-synthesized DNA. Newly-synthesized DNA may then be detected using an anti-BRDU antibody (Hoshino et al, 1986, Int. J. Cancer 38, 369; Campana et al, 1988, J. Immunol. Meth. 107, 79), or by other means.
  • Cell Proliferation may also be examined using [ 3 H]-thymidine incorporation (Chen, J., 1996, Oncogene 13:1395-403; Jeoung, J., 1995, J. Biol. Chem. 270:18367-73).
  • This assay allows for quantitative characterization of S-phase DNA syntheses.
  • cells synthesizing DNA will incorporate [ 3 H]-thymidine into newly synthesized DNA.
  • Incorporation can then be measured by standard techniques such as by counting of radioisotope in a scintillation counter (e.g., Beckman LS 3800 Liquid Scintillation Counter).
  • Cell proliferation may also be assayed by colony formation in soft agar (Sambrook et al., Molecular Cloning, Cold Spring Harbor (1989)). For example, cells transformed with SLC13A are seeded in soft agar plates, and colonies are measured and counted after two weeks incubation.
  • Involvement of a gene in the cell cycle may be assayed by flow cytometry (Gray JW et al. (1986) Int J Radiat Biol Relat Stud Phys Chem Med 49:237-55). Cells transfected with an SLC13A may be stained with propidium iodide and evaluated in a flow cytometer (available from Becton Dickinson).
  • a cell proliferation or cell cycle assay system may comprise a cell that expresses an SLC13A, and that optionally has defective p53 function (e.g. p53 is over- expressed or under-expressed relative to wild-type cells).
  • a test agent can be added to the assay system and changes in cell proliferation or cell cycle relative to controls where no test agent is added, identify candidate p53 modulating agents.
  • the cell proliferation or cell cycle assay may be used as a secondary assay to test a candidate p53 modulating agents that is initially identified using another assay system such as a cell-free assay system.
  • a cell proliferation assay may also be used to test whether SLC13A function plays a direct role in cell proliferation or cell cycle.
  • a cell proliferation or cell cycle assay may be performed on cells that over- or under-express SLC13A relative to wild type cells. Differences in proliferation or cell cycle compared to wild type cells suggest that the SLC 13 A plays a direct role in cell proliferation or cell cycle.
  • Angiogenesis may be assayed using various human endothelial cell systems, such as umbilical vein, coronary artery, or dermal cells. Suitable assays include Alamar Blue based assays (available from Biosource International) to measure proliferation; migration assays using fluorescent molecules, such as the use of Becton Dickinson Falcon HTS FluoroBlock cell culture inserts to measure migration of cells through membranes in presence or absence of angiogenesis enhancer or suppressors; and tubule formation assays based on the formation of tubular structures by endothelial cells on Matrigel® (Becton Dickinson).
  • Alamar Blue based assays available from Biosource International
  • migration assays using fluorescent molecules such as the use of Becton Dickinson Falcon HTS FluoroBlock cell culture inserts to measure migration of cells through membranes in presence or absence of angiogenesis enhancer or suppressors
  • tubule formation assays based on the formation of tubular structures by endothelial cells on Ma
  • an angiogenesis assay system may comprise a cell that expresses an SLC13A, and that optionally has defective p53 function (e.g. p53 is over-expressed or under-expressed relative to wild-type cells).
  • a test agent can be added to the angiogenesis assay system and changes in angiogenesis relative to controls where no test agent is added, identify candidate p53 modulating agents.
  • the angiogenesis assay may be used as a secondary assay to test a candidate p53 modulating agents that is initially identified using another assay system.
  • An angiogenesis assay may also be used to test whether SLC13A function plays a direct role in cell proliferation. For example, an angiogenesis assay may be performed on cells that over- or under-express SLC13A relative to wild type cells. Differences in angiogenesis compared to wild type cells suggests that the SLC13A plays a direct role in angiogenesis.
  • hypoxia inducible factor-1 The alpha subunit of the transcription factor, hypoxia inducible factor-1 (HIF-1), is upregulated in tumor cells following exposure to hypoxia in vitro.
  • HIF-1 hypoxia inducible factor-1
  • hypoxia inducible factor-1 stimulates the expression of genes known to be important in tumour cell survival, such as those encoding glyolytic enzymes and VEGF.
  • Induction of such genes by hypoxic conditions may be assayed by growing cells transfected with SLC13A in hypoxic conditions (such as with 0.1% 02, 5% CO2, and balance N2, generated in a Napco 7001 incubator (Precision Scientific)) and normoxic conditions, followed by assessment of gene activity or expression by Taqman®.
  • a hypoxic induction assay system may comprise a cell that expresses an SLC13A, and that optionally has a mutated p53 (e.g. p53 is over-expressed or under- expressed relative to wild-type cells).
  • a test agent can be added to the hypoxic induction assay system and changes in hypoxic response relative to controls where no test agent is added, identify candidate p53 modulating agents.
  • the hypoxic induction assay may be used as a secondary assay to test a candidate p53 modulating agents that is initially identified using another assay system.
  • a hypoxic induction assay may also be used to test whether SLC13A function plays a direct role in the hypoxic response.
  • hypoxic induction assay may be performed on cells that over- or under-express SLC13A relative to wild type cells. Differences in hypoxic response compared to wild type cells suggests that the SLC13A plays a direct role in hypoxic induction.
  • Cell adhesion assays measure adhesion of cells to purified adhesion proteins, or adhesion of cells to each other, in presence or absence of candidate modulating agents.
  • Cell-protein adhesion assays measure the ability of agents to modulate the adhesion of cells to purified proteins. For example, recombinant proteins are produced, diluted to 2.5g/mL in PBS, and used to coat the wells of a microtiter plate. The wells used for negative control are not coated. Coated wells are then washed, blocked with 1% BSA, and washed again. Compounds are diluted to 2x final test concentration and added to the blocked, coated wells. Cells are then added to the wells, and the unbound cells are washed off.
  • Retained cells are labeled directly on the plate by adding a membrane-permeable fluorescent dye, such as calcein-AM, and the signal is quantified in a fluorescent microplate reader.
  • a membrane-permeable fluorescent dye such as calcein-AM
  • Cell-cell adhesion assays measure the ability of agents to modulate binding of cell adhesion proteins with their native ligands. These assays use cells that naturally or recombinantly express the adhesion protein of choice.
  • cells expressing the cell adhesion protein are plated in wells of a multiwell plate.
  • Cells expressing the ligand are labeled with a membrane-permeable fluorescent dye, such as BCECF , and allowed to adhere to the monolayers in the presence of candidate agents. Unbound cells are washed off, and bound cells are detected using a fluorescence plate reader.
  • High-throughput cell adhesion assays have also been described.
  • small molecule ligands and peptides are bound to the surface of microscope slides using a microarray spotter, intact cells are then contacted with the slides, and unbound cells are washed off.
  • this assay not only the binding specificity of the peptides and modulators against cell lines are determined, but also the functional cell signaling of attached cells using immunofluorescence techniques in situ on the microchip is measured (Falsey JR et al., Bioconjug Chem. 2001 May-Jun;12(3):346-53).
  • appropriate primary assays test is a binding assay that tests the antibody's affinity to and specificity for the SLC13A protein. Methods for testing antibody affinity and specificity are well known in the art (Harlow and Lane, 1988, 1999, supra).
  • the enzyme-linked immunosorbant assay (ELISA) is a preferred method for detecting SLC13A-specific antibodies; others include FACS assays, radioimmunoassays, and fluorescent assays.
  • primary assays may test the ability of the nucleic acid modulator to inhibit or enhance SLC13A gene expression, preferably mRNA expression.
  • expression analysis comprises comparing SLC13A expression in like populations of cells (e.g., two pools of cells that endogenously or recombinantly express SLC 13 A) in the presence and absence of the nucleic acid modulator. Methods for analyzing mRNA and protein expression are well known in the art.
  • Northern blotting For instance, Northern blotting, slot blotting, ribonuclease protection, quantitative RT-PCR (e.g., using the TaqMan®, PE Applied Biosystems), or microarray analysis may be used to confirm that SLC13A mRNA expression is reduced in cells treated with the nucleic acid modulator
  • Protein expression may also be monitored. Proteins are most commonly detected with specific antibodies or antisera directed against either the SLC13A protein or specific peptides. A variety of means including Western blotting, ELISA, or in situ detection, are available (Harlow E and Lane D, 1988 and 1999, supra).
  • Secondary assays may be used to further assess the activity of SLC13A-modulating agent identified by any of the above methods to confirm that the modulating agent affects SLC13A in a manner relevant to the p53 pathway.
  • SLC13A-modulating agents encompass candidate clinical compounds or other agents derived from previously identified modulating agent.
  • Secondary assays can also be used to test the activity of a modulating agent on a particular genetic or biochemical pathway or to test the specificity of the modulating agent's interaction with SLC13A.
  • Secondary assays generally compare like populations of cells or animals (e.g., two pools of cells or animals that endogenously or recombinantly express SLC13A) in the presence and absence of the candidate modulator. In general, such assays test whether treatment of cells or animals with a candidate SLC13A-modulating agent results in changes in the p53 pathway in comparison to untreated (or mock- or placebo-treated) cells or animals. Certain assays use "sensitized genetic backgrounds", which, as used herein, describe cells or animals engineered for altered expression of genes in the p53 or interacting pathways. Cell-based assays
  • Cell based assays may use a variety of mammalian cell lines known to have defective p53 function (e.g. SAOS-2 osteoblasts, H1299 lung cancer cells, C33A and HT3 cervical cancer cells, HT-29 and DLD-1 colon cancer cells, among others, available from American Type Culture Collection (ATCC), Manassas, VA). Cell based assays may detect endogenous p53 pathway activity or may rely on recombinant expression of p53 pathway components. Any of the aforementioned assays may be used in this cell-based format.
  • Candidate modulators are typically added to the cell media but may also be injected into cells or delivered by any other efficacious means.
  • Models for defective p53 pathway typically use genetically modified animals that have been engineered to mis-express (e.g., over- express or lack expression in) genes involved in the p53 pathway.
  • Assays generally require systemic delivery of the candidate modulators, such as by oral administration, injection, etc.
  • p53 pathway activity is assessed by monitoring neovascularization and angiogenesis.
  • Animal models with defective and normal p53 are used to test the candidate modulator's affect on SLC13A in Matrigel® assays.
  • Matrigel® is an extract of basement membrane proteins, and is composed primarily of laminin, collagen IV, and heparin sulfate proteoglycan. It is provided as a sterile liquid at 4° C, but rapidly forms a solid gel at 37° C. Liquid Matrigel® is mixed with various angiogenic agents, such as bFGF and VEGF, or with human tumor cells which over-express the SLC13A.
  • mice Female athymic nude mice (Taconic, Germantown, NY) to support an intense vascular response.
  • Mice with Matrigel® pellets may be dosed via oral (PO), intraperitoneal (IP), or intravenous (IV) routes with the candidate modulator. Mice are euthanized 5 - 12 days post-injection, and the Matrigel® pellet is harvested for hemoglobin analysis (Sigma plasma hemoglobin kit). Hemoglobin content of the gel is found to correlate the degree of neovascularization in the gel.
  • the effect of the candidate modulator on SLC13A is assessed via tumorigenicity assays.
  • xenograft human tumors are implanted SC into female athymic mice, 6-7 week old, as single cell suspensions either from a pre-existing tumor or from in vitro culture.
  • the tumors which express the SLC13A endogenously are injected in the flank, 1 x 10 5 to 1 x 10 7 cells per mouse in a volume of 100 ⁇ L using a 27gauge needle. Mice are then ear tagged and tumors are measured twice weekly.
  • Candidate modulator treatment is initiated on the day the mean tumor weight reaches 100 mg.
  • Candidate modulator is delivered IV, SC, IP, or PO by bolus administration.
  • dosing can be performed multiple times per day.
  • the tumor weight is assessed by measuring perpendicular diameters with a caliper and calculated by multiplying the measurements of diameters in two dimensions.
  • the excised tumors maybe utilized for biomarker identification or further analyses.
  • immunohistochemistry staining xenograft tumors are fixed in 4% paraformaldehyde, 0.1M phosphate, pH 7.2, for 6 hours at 4°C, immersed in 30% sucrose in PBS, and rapidly frozen in isopentane cooled with liquid nitrogen.
  • Specific SLC13A-modulating agents are useful in a variety of diagnostic and therapeutic applications where disease or disease prognosis is related to defects in the p53 pathway, such as angiogenic, apoptotic, or cell proliferation disorders.
  • the invention also provides methods for modulating the p53 pathway in a cell, preferably a cell pre-determined to have defective p53 function, comprising the step of administering an agent to the cell that specifically modulates SLC13A activity.
  • the modulating agent produces a detectable phenotypic change in the cell indicating that the p53 function is restored, i.e., for example, the cell undergoes normal proliferation or progression through the cell cycle.
  • SLC13A is implicated in p53 pathway provides for a variety of methods that can be employed for the diagnostic and prognostic evaluation of diseases and disorders involving defects in the p53 pathway and for the identification of subjects having a predisposition to such diseases and disorders.
  • Various expression analysis methods can be used to diagnose whether SLC13A expression occurs in a particular sample, including Northern blotting, slot blotting, ribonuclease protection, quantitative RT-PCR, and microarray analysis, (e.g., Current Protocols in Molecular Biology (1994) Ausubel FM et al, eds., John Wiley & Sons, Inc., chapter 4; Freeman WM et al., Biotechniques (1999) 26:112-125; Kallioniemi OP, Ann Med 2001, 33:142-147; Blohm and Guiseppi-Elie, Curr Opin Biotechnol 2001, 12:41-47).
  • Tissues having a disease or disorder implicating defective p53 signaling that express an SLC 13 A are identified as amenable to treatment with an SLC13A modulating agent, hi a preferred application, the p53 defective tissue overexpresses an SLC13A relative to normal tissue.
  • an SLC13A modulating agent for example, a Northern blot analysis of mRNA from tumor and normal cell lines, or from tumor and matching normal tissue samples from the same patient, using full or partial SLC13A cDNA sequences as probes, can determine whether particular tumors express or overexpress SLC 13 A.
  • the TaqMan® is used for quantitative RT-PCR analysis of SLC13A expression in cell lines, normal tissues and tumor samples (PE Applied Biosystems).
  • reagents such as the SLC13A oligonucleotides, and antibodies directed against an SLC13A, as described above for: (1) the detection of the presence of SLC13A gene mutations, or the detection of either over- or under-expression of SLC13A mRNA relative to the non- disorder state; (2) the detection of either an over- or an under-abundance of SLC13A gene product relative to the non-disorder state; and (3) the detection of perturbations or abnormalities in the signal transduction pathway mediated by SLC 13 A.
  • the invention is drawn to a method for diagnosing a disease in a patient, the method comprising: a) obtaining a biological sample from the patient; b) contacting the sample with a probe for SLC13A expression; c) comparing results from step (b) with a control; and d) determining whether step (c) indicates a likelihood of disease.
  • the disease is cancer, most preferably a cancer as shown in TABLE 1.
  • the probe may be either DNA or protein, including an antibody.
  • the Drosophila p53 gene was overexpressed specifically in the wing using the vestigial margin quadrant enhancer.
  • Increasing quantities of Drosophila p53 (titrated using different strength transgenic inserts in 1 or 2 copies) caused deterioration of normal wing morphology from mild to strong, with phenotypes including disruption of pattern and polarity of wing hairs, shortening and thickening of wing veins, progressive crumpling of the wing and appearance of dark "death" inclusions in wing blade.
  • the sodium sulfate symporter transmembrane region domains (PFAM 00939) of SLC13A from GI#s 14750419 (SEQ ID NO:12), 4506979 (SEQ ID NO: 14), and 13653602 (SEQ ID NO: 15) are located at approximately amino acid residues 3 to 579, 26 to 569, and 7 to 573, respectively.
  • TM-HMM was used to determine the 11 transmembrane regions of each SLC13A, and their start and end amino acid positions, which are located approximately at amino acids (13,32) (42,64) (77,99) (114,136) (239,261) (281,303) (340,362) (382,399) (465,487) (507,529) (554,576) for GI#14750419 (SEQ ID NO:12), amino acids (13,30) (45,67) (87,106) (116,138) (266,288) (332,354) (367,384) (448,470) (482,501) (506,528) (540,562) for GI#4506979 (SEQ ID NO: 14), and amino acids (12,34) (54,76) (81,103) (137,159) (236,258) (278,300) (335,357) (370,392) (468,490) (505,527) (548,570) for GI#13653602 (SEQ ID NO
  • Fluorescently-labeled SLC13A peptide/substrate are added to each well of a 96-well microtiter plate, along with a test agent in a test buffer (10 mM HEPES, 10 mM NaCl, 6 mM magnesium chloride, pH 7.6). Changes in fluorescence polarization, determined by using a Fluorolite FPM-2 Fluorescence Polarization Microtiter System (Dynatech Laboratories, Inc), relative to control values indicates the test compound is a candidate modifier of SLC 13 A activity.
  • the cell lysate is incubated with 25 ⁇ l of M2 beads (Sigma) for 2 h at 4 °C with gentle rocking. After extensive washing with lysis buffer, proteins bound to the beads are solubilized by boiling in SDS sample buffer, fractionated by SDS-polyacrylamide gel electrophoresis, transferred to poly vinylidene difluoride membrane and blotted with the indicated antibodies. The reactive bands are visualized with horseradish peroxidase coupled to the appropriate secondary antibodies and the enhanced chemiluminescence (ECL) Western blotting detection system (Amersham Pharmacia Biotech).
  • ECL enhanced chemiluminescence
  • NCI National Cancer Institute
  • ATCC American Type Culture Collection, Manassas, VA 20110-2209
  • Normal and tumor tissues were obtained from Impath, UC Davis, Clontech, Stratagene, and Ambion.
  • CA CA
  • Taqman reactions were carried out following manufacturer's protocols, in 25 ⁇ l total volume for 96-well plates and 10 ⁇ l total volume for 384-well plates, using 300nM primer and 250 nM probe, and approximately 25ng of cDNA.
  • the standard curve for result analysis was prepared using a universal pool of human cDNA samples, which is a mixture of cDNAs from a wide variety of tissues so that the chance that a target will be present in appreciable amounts is good.
  • the raw data were normalized using 18S rRNA (universally expressed in all tissues and cells).
  • tumor tissue samples were compared with matched normal tissues from the same patient.
  • a gene was considered overexpressed in a tumor when the level of expression of the gene was 2 fold or higher in the tumor compared with its matched normal sample.
  • a universal pool of cDNA samples was used instead.
  • a gene was considered overexpressed in a tumor sample when the difference of expression levels between a tumor sample and the average of all normal samples from the same tissue type was greater than 2 times the standard deviation of all normal samples (i.e., Tumor - average(all normal samples) > 2 x STDEV(all normal samples) ).
  • Results are shown in Table 1. Data presented in bold indicate that greater than 50% of tested tumor samples of the tissue type indicated in row 1 exhibited over expression of the gene listed in column 1, relative to normal samples. Underlined data indicates that between 25% to 49% of tested tumor samples exhibited over expression.
  • a modulator identified by an assay described herein can be further validated for therapeutic effect by administration to a tumor in which the gene is overexpressed. A decrease in tumor growth confirms therapeutic utility of the modulator.
  • the likelihood that the patient will respond to treatment can be diagnosed by obtaining a tumor sample from the patient, and assaying for expression of the gene targeted by the modulator.
  • the expression data for the gene(s) can also be used as a diagnostic marker for disease progression.
  • the assay can be performed by expression analysis as described above, by antibody directed to the gene target, or by any other available detection method.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • General Health & Medical Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Pathology (AREA)
  • Biotechnology (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biophysics (AREA)
  • General Engineering & Computer Science (AREA)
  • Public Health (AREA)
  • Toxicology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • General Chemical & Material Sciences (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

On a identifié des gènes SLC13A humains en tant que modulateurs de la voie p53 qui sont donc des cibles thérapeutiques pour les troubles associés à un dysfonctionnement de p53. Cette invention concerne également des méthodes qui permettent d'identifier des modulateurs de p53 et qui comprennent le criblage d'agents modulant l'activité de SLC13A.
PCT/US2002/017460 2001-06-05 2002-06-03 Slc13a utiles en tant que modificateurs de la voie p53 et methodes d'utilisation WO2002098468A1 (fr)

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US29607601P 2001-06-05 2001-06-05
US60/296,076 2001-06-05
US32860501P 2001-10-10 2001-10-10
US60/328,605 2001-10-10
US35725302P 2002-02-15 2002-02-15
US60/357,253 2002-02-15
US36119602P 2002-03-01 2002-03-01
US60/361,196 2002-03-01

Publications (1)

Publication Number Publication Date
WO2002098468A1 true WO2002098468A1 (fr) 2002-12-12

Family

ID=27501675

Family Applications (4)

Application Number Title Priority Date Filing Date
PCT/US2002/017457 WO2002098889A2 (fr) 2001-06-05 2002-06-03 Map3ks utiles en tant que modificateurs de la voie p53 et methodes d'utilisation
PCT/US2002/017465 WO2002098891A2 (fr) 2001-06-05 2002-06-03 Gad utiles en tant que modificateurs de la voie p53 et methodes d'utilisation
PCT/US2002/017739 WO2002099068A2 (fr) 2001-06-05 2002-06-03 Lce utilises comme modificateurs de la voie p53 et procede d'utilisation
PCT/US2002/017460 WO2002098468A1 (fr) 2001-06-05 2002-06-03 Slc13a utiles en tant que modificateurs de la voie p53 et methodes d'utilisation

Family Applications Before (3)

Application Number Title Priority Date Filing Date
PCT/US2002/017457 WO2002098889A2 (fr) 2001-06-05 2002-06-03 Map3ks utiles en tant que modificateurs de la voie p53 et methodes d'utilisation
PCT/US2002/017465 WO2002098891A2 (fr) 2001-06-05 2002-06-03 Gad utiles en tant que modificateurs de la voie p53 et methodes d'utilisation
PCT/US2002/017739 WO2002099068A2 (fr) 2001-06-05 2002-06-03 Lce utilises comme modificateurs de la voie p53 et procede d'utilisation

Country Status (5)

Country Link
EP (2) EP1402262A4 (fr)
JP (2) JP2005514906A (fr)
AU (1) AU2002314887A1 (fr)
CA (2) CA2449479A1 (fr)
WO (4) WO2002098889A2 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005083425A2 (fr) * 2004-02-26 2005-09-09 Bayer Healthcare Ag Agents de diagnostic et de therapie destines aux maladies associees a la proteine kinase 13 activee par un mitogene (map3k13)
EP2399998A3 (fr) * 2002-11-22 2012-07-25 Ganymed Pharmaceuticals AG Produits géniques d'expression différentielle dans les tumeurs et leur utilisation
AU2012233044B2 (en) * 2002-11-22 2015-09-03 Ganymed Pharmaceuticals Ag Genetic products differentially expressed in tumors and the use thereof

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2006517788A (ja) * 2002-12-20 2006-08-03 ミレニアム・ファーマシューティカルズ・インコーポレイテッド 15986、2188、20743、9148、9151、9791、44252、14184、42461、8204、7970、25552、21657、26492、2411、15088、1905、28899、63380、33935、10480、12686、25501、17694、15701、53062、49908、21612、38949、6216、46863、9235、2201、6985、9883、12238、18057、21617、39228、49928、54476、62113、64316、12264、32362、58198、2887、3205、8557、9600、9693、44867、53058、55556、57658、2208、10252、10302、14218、33877、10317、10485、25964、14815、1363、1397、14827、21708、3801、64698、2179または13249を使用する癌を処置するための方法および組成物
DE10316701A1 (de) * 2003-04-09 2004-11-04 Hinzmann, Bernd, Dr. Humane Nukleinsäuresequenzen aus Bronchialkarzinomen
CA2532150A1 (fr) * 2003-07-11 2005-01-20 Banyu Pharmaceutical Co., Ltd. Methode d'evaluation de compose efficace dans le traitement de l'obesite
WO2005023186A2 (fr) * 2003-09-04 2005-03-17 Immusol Inc. Methodes permettant d'identifier des agents qui inhibent la croissance des cellules cancereuses
WO2005122285A2 (fr) 2004-06-04 2005-12-22 The Board Of Trustees Of The University Of Illinois Procedes et dispositifs permettant de fabriquer et d'assembler des elements a semi-conducteur imprimables
KR101519038B1 (ko) 2007-01-17 2015-05-11 더 보오드 오브 트러스티스 오브 더 유니버시티 오브 일리노이즈 프린팅­기반 어셈블리에 의해 제조되는 광학 시스템
EP2430652B1 (fr) 2009-05-12 2019-11-20 The Board of Trustees of the University of Illionis Ensembles imprimés de diodes électroluminescentes inorganiques microscopiques ultraminces pour dispositifs d'affichage déformables et semi-transparents
US9442285B2 (en) 2011-01-14 2016-09-13 The Board Of Trustees Of The University Of Illinois Optical component array having adjustable curvature
WO2012158709A1 (fr) 2011-05-16 2012-11-22 The Board Of Trustees Of The University Of Illinois Barrettes de del à gestion thermique assemblées par impression
EP2713863B1 (fr) 2011-06-03 2020-01-15 The Board of Trustees of the University of Illionis Réseau d'électrodes de surface conformables, multiplexées de manière active et à haute densité, pour un interfaçage avec le cerveau
KR101979354B1 (ko) 2011-12-01 2019-08-29 더 보오드 오브 트러스티스 오브 더 유니버시티 오브 일리노이즈 프로그램 변형을 실행하도록 설계된 과도 장치
CN114181944B (zh) * 2020-09-14 2023-10-03 中国科学院动物研究所 突变基因及构建短肢性侏儒症小型猪模型的方法和用途

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0572478B1 (fr) * 1991-02-22 2005-09-07 The Walter And Eliza Hall Institute Of Medical Research Polypeptides et acides nucleiques de hbgad et higad et leur utilisation dans le traitement et le diagnostic de maladies associees a l'autoantigene gad.
US5405941A (en) * 1993-04-15 1995-04-11 National Jewish Center For Immunology And Respiratory Medicine MEKK protein, capable of phosphorylating MEK
US5554523A (en) * 1994-03-01 1996-09-10 Children's Hospital Of Philadelphia Nucleic acid sequences encoding human leucine-zipper protein-kinase
WO1999041385A1 (fr) * 1998-02-13 1999-08-19 Cadus Pharmaceutical Corporation Proteines mekk1 et leurs fragments destines a etre utilises dans la regulation d'une apoptose
WO2001034643A1 (fr) * 1999-11-12 2001-05-17 Human Genome Sciences, Inc. 24 proteines humaines secretees
US6312934B1 (en) * 1998-03-16 2001-11-06 National Jewish Center For Immunology And Respiratory Medicine Human MEKK proteins, corresponding nucleic acid molecules, and uses therefor
WO2000055330A1 (fr) * 1999-03-18 2000-09-21 The University Of Bristol Elongase d'acides gras polyinsatures tires de caenorhabiditis elegans
EP1173566A2 (fr) * 1999-04-23 2002-01-23 Incyte Genomics, Inc. Proteines humaines associees aux membranes
CA2342831A1 (fr) * 1999-07-22 2001-02-01 The University Of British Columbia Enzyme biosynthetique d'acides gras a chaines longues de plantes
US6436703B1 (en) * 2000-03-31 2002-08-20 Hyseq, Inc. Nucleic acids and polypeptides
AU2001270045A1 (en) * 2000-06-20 2002-01-02 Incyte Genomics, Inc. Secreted proteins
JP2004506422A (ja) * 2000-08-11 2004-03-04 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング 新規マイトジェン活性化キナーゼ
JP2004513662A (ja) * 2000-11-29 2004-05-13 キセノン ジェネティックス インコーポレイテッド ヒトエロンガーゼ遺伝子、その使用およびそれを調節する化合物
JP2005504503A (ja) * 2001-02-06 2005-02-17 インサイト・ゲノミックス・インコーポレイテッド 受容体および膜関連タンパク質
WO2002062975A2 (fr) * 2001-02-08 2002-08-15 Bayer Aktiengesellschaft Regulation de la proteine humaine du type elongase hselo1
AU2002247691A1 (en) * 2001-02-08 2002-08-19 Bayer Aktiengesellschaft Regulation of human elongase hselo1-like protein
WO2002096943A1 (fr) * 2001-05-25 2002-12-05 Asahi Kasei Kabushiki Kaisha Genes activant le stat6
AU2003231531A1 (en) * 2002-04-24 2003-11-10 Incyte Corporation Kinases and phosphatases

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
KARNISKI L.: "Mutations in the diastrophic dysplasia sulfate transporter (DTDST) gene: correlation between sulfate transport activity and chondrodysplasia phenotype.", HUMAN MOLECULAR GENETICS, vol. 10, no. 14, 2001, pages 1485 - 1490, XP002954319 *
LEE ET AL: "The human renal sodium sulfate cotransporter (SLC13A1;hNaSi-1) cDNA and gene: Organization, Chromosomal localization, and Functional characterization.", GENOMICS, vol. 70, 2000, pages 354 - 363, XP002954317 *
LEVINE: "p53, the cellular gatekeeper for growth and division.", CELL, vol. 88, 7 February 1997 (1997-02-07), pages 323 - 331, XP002930312 *
OLLMANN ET AL: "Drosophila p53 is a structural and functional homolog of the tumor suppressor p53", CELL, vol. 101, 31 March 2000 (2000-03-31), pages 91 - 101, XP002954318 *
ROGINA ET AL: "Extended life-span conferred by cotransporter gene mutations in Drosophila.", SCIENCE, vol. 290, 15 December 2000 (2000-12-15), pages 2137 - 2140, XP002954320 *

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2399998A3 (fr) * 2002-11-22 2012-07-25 Ganymed Pharmaceuticals AG Produits géniques d'expression différentielle dans les tumeurs et leur utilisation
EP2399997A3 (fr) * 2002-11-22 2012-08-08 Ganymed Pharmaceuticals AG Produits géniques d'expression différentielle dans les tumeurs et leur utilisation
AU2012233044B2 (en) * 2002-11-22 2015-09-03 Ganymed Pharmaceuticals Ag Genetic products differentially expressed in tumors and the use thereof
WO2005083425A2 (fr) * 2004-02-26 2005-09-09 Bayer Healthcare Ag Agents de diagnostic et de therapie destines aux maladies associees a la proteine kinase 13 activee par un mitogene (map3k13)
WO2005083425A3 (fr) * 2004-02-26 2005-10-06 Bayer Healthcare Ag Agents de diagnostic et de therapie destines aux maladies associees a la proteine kinase 13 activee par un mitogene (map3k13)

Also Published As

Publication number Publication date
WO2002098891A2 (fr) 2002-12-12
EP1402262A4 (fr) 2005-05-11
WO2002099068A3 (fr) 2003-12-04
JP2005514906A (ja) 2005-05-26
WO2002098889A3 (fr) 2003-03-27
AU2002314887A1 (en) 2002-12-16
CA2449479A1 (fr) 2002-12-12
CA2448112A1 (fr) 2002-12-12
EP1402262A2 (fr) 2004-03-31
WO2002098891A3 (fr) 2003-05-30
JP2004532638A (ja) 2004-10-28
EP1402051A4 (fr) 2005-08-17
EP1402051A2 (fr) 2004-03-31
WO2002099068A2 (fr) 2002-12-12
WO2002098889A2 (fr) 2002-12-12

Similar Documents

Publication Publication Date Title
US20050170344A1 (en) Chds as modifiers of the p53 pathway and methods of use
EP1404374A1 (fr) Slc2as identifies comme des modificateurs de la voie p53 et leurs procedes d'utilisation
AU2002320264A1 (en) GFATs as modifiers of the p53 pathway and methods of use
WO2002098468A1 (fr) Slc13a utiles en tant que modificateurs de la voie p53 et methodes d'utilisation
US20110159508A1 (en) GFATS as Modifiers of the P53 Pathway and Methods of Use
US20060024673A1 (en) Slc2as as modifiers of the p53 pathway and methods of use
AU2002310273A1 (en) SLC2As as modifiers of the P53 pathway and methods of use
AU2002346246A1 (en) SLC7s as modifiers of the p53 pathway and methods of use
AU2002320051A1 (en) CHDs as modifiers of the p53 pathway and methods of use
AU2002310270A1 (en) HS2STs as modifiers of the p53 pathway and methods of use
AU2002305777A1 (en) CADs as modifiers of the p53 pathway and methods of use
AU2002314883A1 (en) MAP3Ks as modifier of the p53 pathway and methods of use
AU2002312253A1 (en) LIMKs as modifiers of the p53 pathway and methods of use

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP