CA2449479A1 - Map3ks as modifiers of the p53 pathway and methods of use - Google Patents

Map3ks as modifiers of the p53 pathway and methods of use Download PDF

Info

Publication number
CA2449479A1
CA2449479A1 CA002449479A CA2449479A CA2449479A1 CA 2449479 A1 CA2449479 A1 CA 2449479A1 CA 002449479 A CA002449479 A CA 002449479A CA 2449479 A CA2449479 A CA 2449479A CA 2449479 A1 CA2449479 A1 CA 2449479A1
Authority
CA
Canada
Prior art keywords
map3k
ser
assay
leu
agent
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002449479A
Other languages
French (fr)
Inventor
Lori Friedman
Gregory D. Plowman
Marcia Belvin
Helen Francis-Lang
Danxi Li
Roel P. Funke
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Exelixis Inc
Original Assignee
Exelixis, Inc.
Lori Friedman
Gregory D. Plowman
Marcia Belvin
Helen Francis-Lang
Danxi Li
Roel P. Funke
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., Lori Friedman, Gregory D. Plowman, Marcia Belvin, Helen Francis-Lang, Danxi Li, Roel P. Funke filed Critical Exelixis, Inc.
Publication of CA2449479A1 publication Critical patent/CA2449479A1/en
Abandoned legal-status Critical Current

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

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • General Health & Medical Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Organic Chemistry (AREA)
  • Oncology (AREA)
  • Hospice & Palliative Care (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

Human MAP3K genes are identified as modulators of the p53 pathway, and thus are therapeutic targets for disorders associated with defective p53 function . Methods for identifying modulators of p53, comprising screening for agents that modulate the activity of MAP3K are provided.

Description

MAP3Ks AS MODIFIERS OF THE p53 PATHWAY AND METHODS OF USE
REFERENCE TO RELATED APPLICATIONS
This application claims priority to U.S. provisional patent applications 60/296,076 filed 6/5/2001, 60/328,605 filed 10/10/2001, 60/357,253 filed 2/15/2002, and 60/361,196 filed 3/1/2002. The contents of the prior applications are hereby incorporated in their entirety.
BACKGROUND OF THE INVENTION
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.
Aberrant forms of human p53 are associated with poor prognosis, more aggressive tumors, metastasis, and short survival rates (Mitsudomi et al., Clin Cancer Res 2000 Oct;
6(10):4055-63; Koshland, Science (1993) 262:1953).
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).
The biochemical mechanisms and pathways through which p53 functions in normal and cancerous cells are not fully understood, but one clearly important aspect of p53 function is its activity as a gene-specific transcriptional activator. Among the 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).
Protein kinases (PKs) play a crucial role in regulating cellular processes, including growth factor response, cytoskeletal changes, gene expression, and metabolism.
PKs have very similar sequences and can be grouped based on specificity for the acceptor amino acid. Most PKs phosphorylate either serinelthreonine or tyrosine. However, some PKs, referred to as mixed-lineage kinases, have features of both serine/threonine and tyrosine PKs. All PKs have Src homology (SH) domains and can also be grouped as receptors or nonreceptors. Receptor PKs have a transmembrane region, an extracellular ligand-binding domain, and an intracellular catalytic domain.
Mitogen activated protein kinase kinase kinase 12 (MAP3K12), is a dual leucine zipper-bearing kinase, and a member of the mixed lineage protein kinase (MLK) (Reddy, U. and Pleasure, D., (1994) Biochem. Biophys. Res. Commun. 202: 613-620).
MAP3Kl2 contains a COOH-terminal and NH2-terminal proline-rich domains suggestive of src homology 3 (SH3) domain binding regions, and can be autophosphorylated on serine and threonine residues (Holzman, L. et al., (1994) J. Biol. Chem. 269: 30808-30817). This kinase activates the SAPK/JNK signaling pathway, and may play a role in neuronal differentiation (Hirai, S., (1996) Oncogene 12: 641-650).
MAP3K13 protein, also called LZK (leucine zipper-bearing kinase) contains double leucine/isoleucine zippers, has no apparent signal sequence or transmembrane region but does contain a kinase catalytic domain, and an acidic domain at its C-terminal end Sakuma, H. et al.,( 1997) J. Biol. Chem. 272: 28622-28629). MAP3K13 shares 86.4%
amino acid identity with MAP3K12 and like MAP3K12 it is also a member of the mixed-lineage kinase family of proteins which contain similarities to both serine/threonine and tyrosine kinases ( Sakuma, H. et al.,( 1997) J. Biol. Chem. 272: 28622-28629).
These kinases activate the phosphorylation event of c-Jun and turn on JNK-1 (Sakuma, H. et al.,( 1997) J. Biol. Chem. 272: 28622-28629).
MAP3K12 and MAP3K13 are both highly conserved genes that have been found in organisms from yeast to man. MAP3K12 has been implicated in neuronal Bell death (Xu, Z. et al. (2001) Mol Cell Biol 21:4713-24).
The ability to manipulate the genomes of model organisms such as Drosoplaila provides a powerful means to analyze biochemical processes that, due to significant evolutionary conservation, has direct relevance to more complex vertebrate organisms.
Due to a high level of gene and pathway conservation, the strong similarity of cellular processes, and the functional conservation of genes between these model organisms and mammals, identification of the involvement of novel genes in particular pathways and their functions in such model organisms can directly contribute to the understanding of the correlative pathways and methods of modulating them in mammals (see, for example, Mechler BM et al., 1985 EMBO J 4:1551-1557; Gateff E. 1982 Adv. Cancer Res.
37: 33-74; Watson KL., et al., 1994 J Cell Sci. 18: 19-33; Miklos GL, and Rubin GM.
1996 Cell 86:521-529; Wassarman DA, et al., 1995 Curr Opin Gen Dev 5: 44-50; and Booth DR.
1999 Cancer Metastasis Rev. 18: 261-284). For example, 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. When the genetic entry point is an ortholog of a human gene implicated in a disease pathway, such as p53, modifier genes can be identified that may be attractive candidate targets for novel therapeutics.
All references cited herein, including sequence information in referenced Genbank identifier numbers and website references, are incorporated herein in their entireties.
SUMMARY OF THE INVENTION
We have discovered genes that modify the p53 pathway in Drosophila, and identified their human orthologs, hereinafter referred to as MAP3Ks. The invention provides methods for utilizing these p53 modifier genes and polypeptides to identify candidate therapeutic agents that can be used in the treatment of disorders associated with defective p53 function. Preferred MAP3K-modulating agents specifically bind to MAP3K
polypeptides and restore p53 function. Other preferred MAP3K-modulating agents are nucleic acid modulators such as antisense oligomers and RNAi that repress MAP3K gene expression or product activity by, for example, binding to and inhibiting the respective nucleic acid (i.e. DNA or mRNA).
MAP3K-specific modulating agents may be evaluated by any convenient in vitro or if2 viv~ assay for molecular interaction with a MAP3K polypeptide or nucleic acid.
In one embodiment, candidate p53 modulating agents are tested with an assay system comprising a MAP3K 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. MAP3K-modulating agents include MAP3K related proteins (e.g. dominant negative mutants, and biotherapeutics);
MAP3K-specific antibodies; MAP3K-specific antisense oligomers and other nucleic acid modulators; and chemical agents that specifically bind MAP3K or compete with binding target. In one specific embodiment, a small molecule modulator is identified using a kinase assay. In specific embodiments, 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.
S In another embodiment, 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. In specific embodiments, 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 MAP3K 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.
DETAILED DESCRIPTION OF THE INVENTION
Genetic screens were designed to identify modifiers of the p53 pathway in and Dr~sophila in which p53 was overexpressed in the wing (Ollmann M, et al., Cell 101: 91-101). The CG8789 gene was identified as a modifier of the p53 pathway.
Accordingly, vertebrate orthologs of these modifiers, and preferably the human orthologs, MAP3K genes (i.e., nucleic acids and polypeptides) are attractive drug targets for the treatment of pathologies associated with a defective p53 signaling pathway, such as cancer.
In vitro and in vivo methods of assessing MAP3K function are provided herein.
Modulation of the MAP3K or their respective binding partners is useful for understanding the association of the p53 pathway and its members in normal and disease conditions and for developing diagnostics and therapeutic modalities for p53 related pathologies.
MAP3K-modulating agents that act by inhibiting or enhancing MAP3K expression, directly or indirectly, for example, by affecting a MAP3K function such as enzymatic (e.g., catalytic) or binding activity, can be identified using methods provided herein.
MAP3K modulating agents are useful in diagnosis, therapy and pharmaceutical development.
Nucleic acids and nolypentides of the invention Sequences related to MAP3K nucleic acids and polypeptides that can be used in the invention are disclosed in Genbank (referenced by Genbank identifier (GI) number) as GI#s 5454183 (SEQ ID NO:1), 13645287 (SEQ ID N0:4), and 4758695 (SEQ ID N0:5) for nucleic acid, and GI#s 5454184 (SEQ ID N0:8) and 4758696 (SEQ ID N0:9) for polypeptides. Further, sequences of SEQ ID NOs: 2, 3, 6, and 7 can also be used in the invention.
MAP3Ks are kinase proteins with kinase domains. The term "MAP3K polypeptide"
refers to a full-length MAP3K protein or a functionally active fragment or derivative thereof. A "functionally active" MAP3K fragment or derivative exhibits one or more functional activities associated with a full-length, wild-type MAP3K protein, such as antigenic or immunogenic activity, enzymatic activity, ability to bind natural cellular substrates, etc. The functional activity of MAP3K 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. For purposes herein, functionally active fragments also include those fragments that comprise one or more structural domains of a MAP3K, such as a kinase 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:l/pfam.wustl.edul. For example, the kinase domain of MAP3K from GI#

(SEQ ID N0:8) is located at approximately amino acid residues 125-366 (PFAM
00069).
Likewise, the kinase domain of MAP3K from GI# 4758696 (SEQ ~ N0:9) is located at approximately amino acid residues 168-409. Methods for obtaining MAP3K
polypeptides are also further described below. In some embodiments, 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:B or 9 (a MAP3K). In further preferred embodiments, the fragment comprises the entire kinase (functionally active) domain.
The term "MAP3K nucleic acid" refers to a DNA or RNA molecule that encodes a MAP3K polypeptide. Preferably, the MAP3K 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 MAP3K.
Normally, 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, such as CLUSTAL (Thompson JD et al, 1994, Nucleic Acids Res 22:4673-4680) may be used to highlight conserved regions and/or residues of orthologous proteins and to generate phylogenetic trees. In a phylogenetic tree representing multiple homologous sequences from diverse species (e.g., retrieved through BLAST analysis), 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 (e.g., using software by ProCeryon, Biosciences, Salzburg, Austria) may also identify potential orthologs. In evolution, when a gene duplication event follows speciation, a single gene in one species, such as Drosophila, may correspond to multiple genes (paralogs) in another, such as human. As used herein, the term "orthologs" encompasses paralogs. As used herein, "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.Oa19 (Altschul et al., J. Mol. Biol.
(1997) 215:403-410; http://blast.wustl.edu/blast/README.html) with all the search parameters set to default values. The 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.
A conservative amino acid substitution is one in which an amino acid is substituted for another amino acid having similar properties such that the folding or activity of the protein is not significantly affected. 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.
Alternatively, an alignment for nucleic acid sequences is 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, or 7The 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)). In some embodiments, 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, or 7 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) (1X SSC is 0.15 M NaCI, 0.015 M Na citrate;
pH 7.0), 5X
Denhardt's solution, 0.05% sodium pyrophosphate and 100 ,uglml herring sperm DNA;
hybridization for 18-20 hours at 65° C in a solution containing 6X SSC, 1X Denhardt's solution, 100 ~.g/ml yeast tRNA and 0.05% sodium pyrophosphate; and washing of filters at 65° C for 1h in a solution containing 0.2X SSC and 0.1% SDS (sodium dodecyl sulfate).
In other embodiments, moderately stringent hybridization conditions are used that 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 ~Cg/ml salmon sperm DNA, and 10% (wdvol) dextrin sulfate; followed by washing twice for 1 hour at 55° C in a solution containing 2X SSC and 0.1% SDS.
Alternatively, 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, IO% dextrin sulfate, and 20 ~Cglml denatured sheared salmon sperm DNA; hybridization in the same buffer for 18 to hours; and washing of filters in 1 x SSC at about 37° C for I hour.
Isolation, Production, Expression, and Mis-expression of MAP3K Nucleic Acids and Polyneptides MAP3I~ nucleic acids and polypeptides, useful for identifying and testing agents that modulate MAP3K function and for other applications related to the involvement of MAP3I~ in the p53 pathway. MAP3K 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 polymerise chain reaction (PCR) are well known in the art. In general, 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. Expression of proteins to be purified for screening or antibody production may require the addition of specific tags (e.g., generation of fusion proteins).
Overexpression of a MAP3K protein for assays used to assess MAP3K function, such as involvement in cell cycle regulation or hypoxic response, may require expression in eukaryotic cell lines capable of these cellular activities. Techniques for the expression, production, and purification of proteins are well known in the art; any suitable means therefore may be , used (e.g., Higgins SJ and Hames BD (eds.) Protein Expression: A Practical Approach, Oxford University Press Inc., New York 1999; Stanbury PF et al., Principles of Fermentation Technology, 2nd edition, Elsevier Science, New York, 1995; Doonan S (ed.) Protein Purification Protocols, Humana Press, New Jersey, 1996; Coligan JE et al, Current Protocols in Protein Science (eds.), 1999, John Wiley & Sons, New York). In particular embodiments, recombinant MAP3K 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 American Type Culture Collection (ATCC), Manassas, VA). The recombinant cells are used in cell-based screening assay systems of the invention, as described further below.
The nucleotide sequence encoding a MAP3K polypeptide can be inserted into any appropriate expression vector. The necessary transcriptional and translational signals, including promoter/enhancer element, can derive from the native MAP3K 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. baculovirus);
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.
To detect expression of the MAP3K gene product, the expression vector can comprise a promoter operably linked to a MAP3K gene nucleic acid, one or more origins of replication, and, one or more selectable markers (e.g. thymidine kinase activity, resistance to antibiotics, etc.). Alternatively, recombinant expression vectors can be identified by assaying for the expression of the MAP3K gene product based on the physical or functional properties of the MAP3K protein in in vitro assay systems (e.g.
immunoassays).
The MAP3K protein, fragment, or derivative may be optionally expressed as a fusion, or chimeric protein product (i.e. it is joined via a peptide bond to a heterologous protein sequence of a different protein), for example to facilitate purification or detection. 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).
Once a recombinant cell that expresses the MAP3K gene sequence is identified, 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). Alternatively, native MAP3K
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 MAP3K or other genes associated with the pS3 pathway.
As used herein, 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).
Genetically modified animals Animal models that have been genetically modified to alter MAP3K 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 MAP3K in a p53 pathway process such as apoptosis or cell proliferation. Preferably, the altered MAP3K expression results in a detectable phenotype, such as decreased or increased levels of cell proliferation, angiogenesis, or apoptosis compared to control animals having normal MAP3K 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 Z~rosophila. 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.

Methods of making transgenic animals are well-known in the art (for 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. No., 4,945,050, by Sandford et al.; for transgenic Drosophila see Rubin arid Spradling, Science (1982) 218:348-53 and U.S. Pat. No. 4,670,388; for transgenic insects see Berghammer A.J. et al., A Universal Marker for Transgenic Insects (1999) Nature 402:370-371; for transgenic Zebrafish see Lin S., Transgenic Zebrafish, Methods Mol Biol. (2000);136:375-3830); for microinjection procedures for fish, amphibian eggs and birds see Houdebine and Chourrout, Experientia (1991) 47:897-905; for transgenic rats see Hammer et al., Cell (1990) 63:1099-1112; and for culturing of embryonic stem (ES) cells and the subsequent production of transgenic animals by the introduction of DNA into ES cells using methods such as electroporation, calcium phosphatelDNA precipitation and direct injection see, e.g., Teratocarcinomas and Embryonic Stem Cells, A Practical Approach, E. J.
Robertson, ed., IRL Press (1987)). 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).
In one embodiment, the transgenic animal is a "knock-out" animal having a heterozygous or homozygous alteration in the sequence of an endogenous MAP3K
gene that results in a decrease of MAP3K function, preferably such that MAP3K
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. For example, a mouse MAP3K gene is used to construct a homologous recombination vector suitable for altering an endogenous MAP3K 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). In a preferred embodiment, 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).
In another embodiment, 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 MAP3K gene, e.g., by introduction of additional copies of MAP3K, or by operatively inserting a regulatory sequence that provides for altered expression of an endogenous copy of the MAP3K gene. Such regulatory sequences 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. One example of such a system that may be produced is the cre/loxP recombinase system of bacteriophage P1 (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. Another example of 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). In a preferred embodiment, 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 MAP3K function and phenotypic changes are compared with appropriate control animals such as genetically modified animals that receive placebo treatment, and/or animals with unaltered MAP3K expression that receive candidate therapeutic agent.
In addition to the above-described genetically modified animals having altered MAP3K function, animal models having defective p53 function (and otherwise normal MAP3K function), can be used in the methods of the present invention. For example, 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). Preferably, 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.
ModuIatin~ Agents The invention provides methods to identify agents that interact with and/or modulate the function of MAP3K 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 MAP3K 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 MAP3K activity by administering a MAP3K-interacting or -modulating agent.
In a preferred embodiment, MAP3K-modulating agents inhibit or enhance MAP3K
activity or otherwise affect normal MAP3K function, including transcription, protein expression, protein localization, and cellular or extra-cellular activity. In a further preferred embodiment, the candidate p53 pathway- modulating agent specifically modulates the function of the MAP3K. The phrases "specific modulating agent", "specifically modulates", etc., are used herein to refer to modulating agents that directly bind to the MAP3K polypeptide or nucleic acid, and preferably inhibit, enhance, or otherwise alter, the function of the MAP3K. The term also encompasses modulating agents that alter the interaction of the MAP3K with a binding partner or substrate (e.g. by binding to a binding partner of a MAP3K, or to a protein/binding partner complex, and inhibiting function).
Preferred MAP3K-modulating agents include small molecule compounds; MAP3K-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~
edition.
Small molecule modulators 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 MAP3K protein or may be identified by screening compound libraries. Alternative appropriate 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 MAP3K-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. Additionally, candidate clinical compounds are generated with specific regard to clinical and pharmacological properties. For example, the reagents may be derivatized and re-screened using in vitro and an vivo assays to optimize activity and minimize toxicity for pharmaceutical development.
Protein Modulators Specific MAP3K-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 MAP3K-modulating agents. In a preferred embodiment, MAP3K-interacting proteins affect normal MAP3K function, including transcription, protein expression, protein localization, and cellular or extra-cellular activity. In another embodiment, MAP3K-interacting proteins are useful in detecting and providing information about the function of MAP3K proteins, as is relevant to p53 related disorders, such as cancer (e.g., for diagnostic means).
An MAP3K-interacting protein may be endogenous, i.e. one that naturally interacts genetically or biochemically with a MAP3K, such as a member of the MAP3K
pathway that modulates MAP3K expression, localization, and/or activity. MAP3K-modulators include dominant negative forms of MAP3K-interacting proteins and of MAP3K
proteins themselves. Yeast two-hybrid and variant screens offer preferred methods for identifying endogenous MAP3K-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. 169-203; Fashema SF et al., Gene (2000) 250:1-14; Drees BL Curr Opin Chem Biol (1999) 3:64-70; Vidal M and Legrain P Nucleic Acids Res (1999) 27:919-29; and U.S. Pat. No. 5,928,868). 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 3rd, Trends Genet (2000) 16:5-8).
An MAP3K-interacting protein may be an exogenous protein, such as a MAP3K-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). MAP3K antibodies are further discussed below.
In preferred embodiments, a MAP3K-interacting protein specifically binds a protein. In alternative preferred embodiments, a MAP3K-modulating agent binds a MAP3K substrate, binding partner, or cofactor.
Antibodies In another embodiment, the protein modulator is a MAP3K specific antibody agonist or antagonist. The antibodies have therapeutic and diagnostic utilities, and can be used in screening assays to identify MAP3K modulators. The antibodies can also be used in dissecting the portions of the MAP3K pathway responsible for various cellular responses and in the general processing and maturation of the MAP3K.
Antibodies that specifically bind MAP3K polypeptides can be generated using known methods. Preferably the antibody is specific to a mammalian ortholog of MAP3K
polypeptide, and more preferably, to human MAP3K. Antibodies may be polyclonal, monoclonal (mAbs), humanized or chimeric antibodies, single chain antibodies, Fab fragments, F(ab')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 MAP3K which are particularly antigenic can be selected, for example, by routine screening of MAP3K 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:8 or 9. Monoclonal antibodies with affinities of 108 M-1 preferably 10~ M-1 to 101° 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 MAP3K or substantially purified fragments thereof. If MAP3K fragments are used, they preferably comprise at least 10, and more preferably, at least 20 contiguous amino acids of a MAP3K
protein. In a particular embodiment, MAP3K-specific antigens and/or immunogens are coupled to carrier proteins that stimulate the immune response. For example, 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. An appropriate immune system such as a laboratory rabbit or mouse is 2Q immunized according to conventional protocols.
The presence of MAP3K-specific antibodies is assayed by an appropriate assay such as a solid phase enzyme-linked immunosorbant assay (ELISA) using immobilized corresponding MAP3K polypeptides. Other assays, such as radioimmunoassays or fluorescent assays might also be used.
Chimeric antibodies specific to MAP3K polypeptides can be made that contain different portions from different animal species. For instance, 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).
MAP3K-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).
Other suitable techniques for antibody production involve in vitro exposure of lymphocytes to the antigenic polypeptides or alternatively to selection of libraries of antibodies in phage or similar vectors (Huse et al., Science (1989) 246:1275-1281). As used herein, T-cell antigen receptors are included within the scope of antibody modulators (Harlow and Lane, 1988, supra).
The 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). A
wide variety of 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. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437;
4,275,149;
and 4,366,241). Also, recombinant immunoglobulins may be produced (U.S. Pat.
No.
4,816,567). Antibodies to cytoplasmic polypeptides may be delivered and reach their targets by conjugation with membrane-penetrating toxin proteins (U.S. Pat. No.
6,086,900).
When used therapeutically in a patient, 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.
Typically, the amount of antibody administered is in the range of about 0.1 mg/kg -to about 10 mg/kg of patient weight. For parenteral administration, the antibodies are formulated in a unit dosage injectable form (e.g., solution, suspension, emulsion) in association with a pharmaceutically acceptable vehicle. Such vehicles 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 aboutl0 mg/ml.
Immunotherapeutic methods are further described in the Literature (US Pat. No.
5,859,206;
W00073469).
Nucleic Acid Modulators Other preferred MAP3K-modulating agents comprise nucleic acid molecules, such as antisense oligomers or double stranded RNA (dsRNA), which generally inhibit activity. Preferred nucleic acid modulators interfere with the function of the nucleic acid such as DNA replication, transcription, translocation of the MAP3K RNA to the site of protein translation, translation of protein from the MAP3K RNA, splicing of the MAP3K RNA to yield one or more mRNA species, or catalytic activity which may be engaged in or facilitated by the MAP3K RNA.
In one embodiment, the antisense oligomer is an oligonucleotide that is sufficiently complementary to a MAP3K mRNA to bind to and prevent translation, preferably by binding to the 5' untranslated region. MAP3K-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.
In another embodiment, 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 Drug Dev. :7:187-95; US Pat. No.
5,235,033; and US Pat No. 5,378,841).
Alternative preferred MAP3K nucleic acid modulators are double-stranded RNA
species mediating RNA interference (RNAi). 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. Methods relating to the use of RNAi to silence genes in C. elegar2s, l~rosophila, plants, and humans are known in the art (Fire A, et al., 1998 Nature 391:806-811; Fire, A. Trends Genet.
15, 3S8-363 (1999); Sharp, P. A. RNA interference 2001. Genes Dev. 15, 485-490 (2001);
Hammond, S. M., et al., Nature Rev. Genet. 2, 110-1119 (2001); Tuschl, T. Chem.
Biochem. 2, 239-245 (2001); Hamilton, A. et al., Science 286, 950-952 (1999); Harnmond, S. M., et al., Nature 404, 293-296 (2000); Zamore, P. D., et al., Cell 101, 25-33 (2000);
Bernstein, E., et al., Nature 409, 363-366 (2001); Elbashir, S. M., et al., Genes Dev. 15, (2001); W00129058; WO99326I9; Elbashir SM, et aL, 2001 Nature 411:494-498).
Nucleic acid modulators are commonly used as research reagents, diagnostics, and therapeutics. For example, antisense oligonucleotides, which are able to inhibit gene expression with exquisite 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.
For example, antisense oligomers have been employed as therapeutic moieties in the treatment of disease states in animals and man and have been demonstrated in numerous clinical trials to be safe and effective (Milligan JF, et al, Current Concepts in Antisense Drug Design, J Med Chem. (1993) 36:1923-1937; Tonkinson JL et al., Antisense Oligodeoxynucleotides as Clinical Therapeutic Agents, Cancer Invest. (1996) 14:54-65).
Accordingly, in one aspect of the invention, a MAP3K-specific nucleic acid modulator is used in an assay to further elucidate the role of the MAP3K in the p53 pathway, and/or its relationship to other members of the pathway. In another aspect of the invention, a MAP3K-specific antisense oligomer is used as a therapeutic agent for treatment of p53-related disease states.

Assay Systems The invention provides assay systems and screening methods for identifying specific modulators of MAP3K activity. As used herein, an "assay system" encompasses all the components required for performing and analyzing results of an assay that detects and/or measures a particular event. In general, primary assays are used to identify or confirm a modulator's specific biochemical or molecular effect with respect to the MAP3K
nucleic acid or protein. In general, secondary assays further assess the activity of a modulating agent identified by a primary assay and may confirm that the modulating agent affects MAP3K in a manner relevant to the p53 pathway. In some cases, MAP3K
modulators will be directly tested in a secondary assay.
In a preferred embodiment, the screening method comprises contacting a suitable assay system comprising a MAP3K polypeptide with a candidate agent under conditions whereby, but for the presence of the agent, the system provides a reference activity (e.g.
kinase activity), which is based on the particular molecular event the screening method detects. A statistically significant difference between the agent-biased activity and the reference activity indicates that the candidate agent modulates MAP3K
activity, and hence the p53 pathway.
Primary Assays The type of modulator tested generally determines the type of primary assay.
Primary assays for sfnall »zoleczsle modulators For small molecule modulators, 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). As used herein 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. The term "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 MAP3K 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 MAP3K-interacting proteins are used in screens to identify small molecule modulators, the binding specificity of the interacting protein to the MAP3K protein may be assayed by various known methods such as substrate processing (e.g. ability of the candidate MAP3K-specific binding agents to function as negative effectors in MAP3K-expressing cells), binding equilibrium constants (usually at least about 107 M-1, preferably at least about 10$1V1-1, more preferably at least about 109 M-1), and immunogenicity (e.g. ability to elicit MAP3K
specific antibody in a heterologous host such as a mouse, rat, goat or rabbit). For enzymes and receptors, 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 MAP3K polypeptide, a fusion protein thereof, or to cells or membranes bearing the polypeptide or fusion protein. The MAP3K polypeptide can be full length or a fragment thereof that retains functional MAP3K activity. The polypeptide may be fused to another polypeptide, such as a peptide tag for detection or anchoring, or to another tag. The MAP3K polypeptide is preferably human MAP3K, or is an ortholog or derivative thereof as described above. In a preferred embodiment, the screening assay detects candidate agent-based modulation of MAP3K interaction with a binding target, such as an endogenous or exogenous protein or other substrate that has MAP3K-specific binding activity, and can be used to assess normal MAP3K gene function.
Suitable assay formats that may be adapted to screen for MAP3K modulators are known in the art. Preferred 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). In one preferred embodiment, screening assays uses fluorescence technologies, including fluorescence polarization, time-resolved fluorescence, and fluorescence resonance energy transfer. These systems offer means to monitor protein-protein or DNA-protein interactions in which the intensity of the signal emitted from dye-labeled molecules depends upon their interactions with partner molecules (e.g., Selvin PR, Nat Struct Biol (2000) 7:730-4; Fernandes PB, supra;
Hertzberg RP and Pope AJ, Curr Opin Chem Biol (2000) 4:445-451).
A variety of suitable assay systems may be used to identify candidate MAP3K
and p53 pathway modulators (e.g. U.S. Pat. No. 6,165,992 (kinase assays); U.S. Pat.
Nos.
5,550,019 and 6,133,437 (apoptosis assays); and U.S. Pat. No. 6,020,135 (p53 modulation), among others). Specific preferred assays are described in more detail below.
Kinase assays. In some preferred embodiments the screening assay detects the ability of the test agent to modulate the kinase activity of a MAP3K polypeptide. In further embodiments, a cell-free kinase assay system is used to identify a candidate p53 modulating agent, and a secondary, cell-based assay, such as an apoptosis or hypoxic induction assay (described below), may be used to further characterize the candidate p53 modulating agent. Many different assays for kinases have been reported in the literature and are well known to those skilled in the art (e.g. U.S. Pat. No. 6,165,992;
Zhu et aL, Nature Genetics (2000) 26:283-289; and W00073469). Radioassays, which monitor the transfer of a gamma phosphate are frequently used. For instance, a scintillation assay for p56 (lck) kinase activity monitors the transfer of the gamma phosphate from gamma -33P
ATP to a biotinylated peptide substrate; the substrate is captured on a streptavidin coated bead that transmits the signal (Beveridge M et al., J Biomol Screen (2000) 5:205-212).
This assay uses the scintillation proximity assay (SPA), in which only radio-ligand bound to receptors tethered to the surface of an SPA bead are detected by the scintillant immobilized within it, allowing binding to be measured without separation of bound from free ligand.
Apoptosis assays. Assays for apoptosis may be performed by terminal deoxynucleotidyl transferase-mediated digoxigenin-11-dUTP nick end labeling (TUNEL) assay. The TUNEL assay is used to measure nuclear DNA fragmentation characteristic of apoptosis ( Lazebnik 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 a MAP3K, 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. In some embodiments of the invention, 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 MAP3K function plays a direct role in apoptosis.
For example, an apoptosis assay may be performed on cells that over- or under-express MAP3K relative to wild type cells. Differences in apoptotic response compared to wild type cells suggests that the MAP3K plays a direct role in the apoptotic response.
Apoptosis assays are described further in US Pat. No. 6,133,437.
Cell proliferation and cell cycle assays. Cell proliferation may be assayed via bromodeoxyuridine (BRDU) incorporation. 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 [3H]-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. In this assay, cells synthesizing DNA will incorporate [3H]-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 MAP3K 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 cytometxy (Gray JW et al. (1986) Int J Radiat Biol Relat Stud Phys Chem Med 49:237-55). Cells transfected with a MAP3K may be stained with propidium iodide and evaluated in a flow cytometer (available from Becton Dickinson).
Accordingly, a cell proliferation or cell cycle assay system may comprise a cell that expresses a MAP3K, 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. In some embodiments of the invention, 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 kinase assay system. A cell proliferation assay may also be used to test whether MAP3K function plays a direct role in cell proliferation or cell cycle. For example, a cell proliferation or cell cycle assay may be performed on cells that over- or under-express MAP3K relative to wild type cells. Differences in proliferation or cell cycle compared to wild type cells suggests that the MAP3K plays a direct role in cell proliferation or cell cycle.
Angiogenesis. 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). Accordingly, an angiogenesis assay system may comprise a cell that expresses a MAP3K, 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. In some embodiments of the invention, 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 MAP3K function plays a direct role in cell proliferation. For example, an angiogenesis assay may be performed on cells that over- or under-express MAP3K relative to wild type cells.
Differences in angiogenesis compared to wild type cells suggests that the MAP3K plays a direct role in angiogenesis.
Hypoxic induction. The alpha subunit of the transcription factor, hypoxia inducible factor-1 (HIF-1), is upregulated in tumor cells following exposure to hypoxia in vitro.
Under hypoxic conditions, HIF-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 MAP3K in hypoxic conditions (such as with 0.1% 02, 5% C02, and balance N2, generated in a Napco 7001 incubator (Precision Scientific)) and normoxic conditions, followed by assessment of gene activity or expression by Taqman~.
For example, a hypoxic induction assay system may comprise a cell that expresses a MAP3K, and that optionally has a mutated p53 (e.g. p53 is over-expressed ox 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. In some embodiments of the invention, 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 MAP3K function plays a direct role in the hypoxic response. For example, a hypoxic induction assay may be performed on cells that over- or under-express MAP3K relative to wild type cells. Differences in hypoxic response compared to wild type cells suggests that the MAP3K plays a direct role in hypoxic induction.
Cell adhesion. 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.Sg/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.
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. In an exemplary assay, 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. In one such assay, 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. In 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;l2(3):346-53).
Primary assays for antibody modulators For antibody modulators, appropriate primary assays test is a binding assay that tests the antibody's affinity to and specificity for the MAP3K 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 MAP3K-specific antibodies; others include FACS assays, radioimmunoassays, and fluorescent assays.
Primary assays for nucleic acid modulators For nucleic acid modulators, primary assays may test the ability of the nucleic acid modulator to inhibit or enhance MAP3K gene expression, preferably mRNA
expression.
In general, expression analysis comprises comparing MAP3K expression in like populations of cells (e.g., two pools of cells that endogenously or reeombinantly express MAP3K) in the presence and absence of the nucleic acid modulator. Methods for analyzing mRNA and protein expression are well known in the art. 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 MAP3K mRNA expression is reduced in cells treated with the nucleic acid modulator (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 DH and Guiseppi-Elie, A Curr Opin Biotechnol 2001, 12:41-47). Protein expression may also be monitored. Proteins are most commonly detected with specific antibodies or antisera directed against either the MAP3K
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 Secondary assays may be used to further assess the activity of MAP3K-modulating agent identified by any of the above methods to confirm that the modulating agent affects MAP3K in a manner relevant to the p53 pathway. As used herein, MAP3K-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 MAP3K.
Secondary assays generally compare like populations of cells or animals (e.g., two pools of cells or animals that endogenously or recombinantly express MAP3K) in the presence and absence of the candidate modulator. In general, such assays test whether treatment of cells or animals with a candidate MAP3K-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.
Animal Assays A variety of non-human animal models of normal or defective p53 pathway may be used to test candidate MAP3K modulators. 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.
In a preferred embodiment, 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 MAP3K 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 MAP3K. The mixture is then injected subcutaneously(SC) into 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.
In another preferred embodiment, the effect of the candidate modulator on MAP3K is assessed via tumorigenicity assays. In one example, 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 ifi vitro culture. The tumors which express the MAP3K
endogenously are injected in the flank, 1 x 105 to 1 x 107 cells per mouse in a volume of 100 p.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. Depending upon the pharmacokinetics of each unique candidate modulator, 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. At the end of the experiment, the excised tumors maybe utilized for biomarker identification or further analyses. For imrnunohistochemistry staining, xenograft tumors are fixed in 4%
paraformaldehyde, O.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.
Diagnostic and therapeutic uses Specific MAP3K-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.
Accordingly, 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 MAP3K activity. Preferably, 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.
The discovery that MAP3K 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 MAP3K
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 a MAP3K, are identified as amenable to treatment with a MAP3K modulating agent.
In a preferred application, the p53 defective tissue overexpresses a MAP3K relative to normal tissue. 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 MAP3K cDNA sequences as probes, can determine whether particular tumors express or overexpress MAP3K. Alternatively, the TaqMan~ is used for quantitative RT-PCR analysis of MAP3K expression in cell lines, normal tissues and tumor samples (PE
Applied Biosystems).
Various other diagnostic methods may be performed, fox example, utilizing reagents such as the MAP3K oligonucleotides, and antibodies directed against a MAP3K, as described above for: (1) the detection of the presence of MAP3K gene mutations, or the detection of either over- or under-expression of MAP3K mRNA relative to the non-disorder state; (2) the detection of either an over- or an under-abundance of MAP3K gene product relative to the non-disorder state; and (3) the detection of perturbations or abnormalities in the signal transduction pathway mediated by MAP3K.
Thus, in a specific embodiment, 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 MAP3K expression; c) comparing results from step (b) with a control; and d) determining whether step (c) indicates a likelihood of disease. Preferably, the disease is cancer, most preferably a cancer as shown in TABLE 1. The probe may be either DNA or protein, including an antibody.
EXAMPLES
The following experimental section and examples are offered by way of illustration and not by way of limitation.
I. Drosophila p53 screen 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. In a screen designed to identify enhancers and suppressors of Drosophila p53, homozygous females carrying two copies of p53 were crossed to 5663 males carrying random insertions of a piggyBac transposon (Eraser M et al., Virology (1985) 145:356-361). Progeny containing insertions were compared to non-insertion-bearing sibling progeny for enhancement or suppression of the p53 phenotypes. Sequence information surrounding the piggyBac insertion site was used to identify the modifier genes. Modifiers of the wing phenotype were identified as members of the p53 pathway. CG8789 was an enhancer of the wing phenotype.
Human orthologs of the modifiers, are referred to herein as MAP3K.
BLAST analysis (Altschul et al., supra) was employed to identify Targets from Drosophila modifiers. [For example, representative sequences from MAP3K, GI#5454184 (SEQ ID N0:8) and GI#4758696 (SEQ ID N0:9) share 52% and 37% amino acid identity, respectively, with the Drosophila.CG8789.
Various domains, signals, and functional subunits in proteins were analyzed using the PSORT (Nakai K., and Horton P., Trends Biochem Sci, 1999, 24:34-6; Kenta Nakai, Protein sorting signals and prediction of subcellular localization, Adv.
Protein Chem. 54, 277-344 (2000)), PFAM (Bateman A., et al., Nucleic Acids Res, 1999, 27:260-2;
htt~//pfam.wustl.edu), SMART (Ponting CP, et al., SMART: identification and annotation of domains from signaling and extracellular protein sequences. Nucleic Acids Res. 1999 Jan 1;27(1):229-32), TM-HMM (Erik L.L. Sonnhammer, Gunnar von Heijne, and Anders Krogh: A hidden Markov model for predicting transmembrane helices in protein sequences. In Proc. of Sixth Int. Conf. on Intelligent Systems for Molecular Biology, p 175-182 Ed J. Glasgow, T. Littlejohn, F. Major, R. Lathrop, D. Sankoff, and C.
Sensen Menlo Park, CA: AAAI Press, 1998), and clust (Remm M, and Sonnhammer E.
Classification of transmembrane protein families in the Caenorhabditis elegans genome and identification of human orthologs. Genome Res. 2000 Nov;lO(11):1679-89) programs.
Using PFAM, the kinase domain of MAP3K from GI# 5454184 (SEQ ID N0:8) is located at approximately amino acid residues 125-366 (PFAM 00069). Likewise, the kinase domain of MAP3K from GI# 4758696 (SEQ ID N0:9) is located at approximately amino acid residues 168-409.
II. High-Throughput In Vitro Fluorescence Polarization Assay Fluorescently-labeled MAP3K 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
NaCI, 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 MAP3K activity.

III. High-Throu.~hput In Vitro BindingAssay.
s3P-labeled MAP3K peptide is added in an assay buffer (100 mM KCI, 20 mM HEPES
pH 7.6, 1 mM MgCl2, 1% glycerol, 0.5% NP-40, 50 mM beta-mercaptoethanol, 1 mg/ml BSA, cocktail of protease inhibitors) along with a test agent to the wells of a Neutralite-avidin coated assay plate and incubated at 25°C for 1 hour.
Biotinylated substrate is then added to each well and incubated for 1 hour. Reactions are stopped by washing with PBS, and counted in a scintillation counter. Test agents that cause a difference in activity relative to control without test agent are identified as candidate p53 modulating agents.
IV. Immunopreci~itations and Immunoblottin~
For coprecipitation of transfected proteins, 3 x 106 appropriate recombinant cells containing the MAP3K proteins are plated on 10-cm dishes and transfected on the following day with expression constructs. The total amount of DNA is kept constant in each transfection by adding empty vector. After 24 h, cells are collected, washed once with phosphate-buffered saline and lysed for 20 min on ice in 1 ml of lysis buffer containing 50 mM Hepes, pH 7.9, 250 mM NaCI, 20 mM -glycerophosphate, 1 mM
sodium orthovanadate, 5 mM p-nitrophenyl phosphate, 2 mM dithiothreitol, protease inhibitors (complete, Roche Molecular Biochemicals), and 1 % Nonidet P-40.
Cellular debris is removed by centrifugation twice at 15,000 x g for 15 min. The cell lysate is incubated with 25 ~,1 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 polyvinylidene 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).
V. Kinase assay A purified or partially purified MAP3K is diluted in a suitable reaction buffer, e.g., 50 mM Hepes, pH 7.5, containing magnesium chloride or manganese chloride (1-20 mM) and a peptide or polypeptide substrate, such as myelin basic protein or casein (1-10 ~,g/ml).
The final concentration of the kinase is 1-20 nM. The enzyme reaction is conducted in microtiter plates to facilitate optimization of reaction conditions by increasing assay throughput. A 96-well microtiter plate is employed using a final volume 30-100 ~,1. The reaction is initiated by the addition of 33P-gamma-ATP (0.5 ~,Ci/ml) and incubated for 0.5 to 3 hours at room temperature. Negative controls are provided by the addition of EDTA, which chelates the divalent cation (Mg2+ or Mn2+) required for enzymatic activity.
Following the incubation, the enzyme reaction is quenched using EDTA. Samples of the reaction are transferred to a 96-well glass fiber filter plate (MultiScreen, Millipore). The filters are subsequently washed with phosphate-buffered saline, dilute phosphoric acid (0.5°Io) or other suitable medium to remove excess radiolabeled ATP.
Scintillation cocktail is added to the filter plate and the incorporated radioactivity is quantitated by scintillation counting (Wallac/Perkin Elmer). Activity is defined by the amount of radioactivity detected following subtraction of the negative control reaction value (EDTA
quench).
VI. Expression analysis All cell lines used in the following experiments are NCI (National Cancer Institute) lines, and are available from ATCC (American Type Culture Collection, Manassas, VA
20110-2209). Normal and tumor tissues were obtained from Impath, UC Davis, Clontech, Stratagene, and Ambion.
TaqMan analysis was used to assess expression levels of the disclosed genes in various samples.
RNA was extracted from each tissue sample using Qiagen (Valencia, CA) RNeasy kits, following manufacturer's protocols, to a final concentration of 50ng1~,l. Single stranded cDNA was then synthesized by reverse transcribing the RNA samples using random hexamers and 500ng of total RNA per reaction, following protocol 4304965 of Applied Biosystems (Foster City, CA, httn:/lwww.appliedbiosystems.com/ ).
Primers for expression analysis using TaqMan assay (Applied Biosystems, Foster City, CA) were prepared according to the TaqMan protocols, and the following criteria: a) primer pairs were designed to span introns to eliminate genomic contamination, and b) each primer pair produced only one product.
Taqman reactions were carried out following manufacturer's protocols, in 25 ~.1 total volume for 96-well plates and 10 ~1 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).
For each expression analysis, 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. In cases where normal tissue was not available, a universal pool of cDNA samples was used instead. In these cases, 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. Results from various batches of mRNA are represented for each batch. 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. Prior to treating a patient with 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 genes) 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.
Table 1 breast. . . . . .
colon lung ovary GI#5454183 (SEQ TaqExp_1005012 11. 30 . 13 . 7 ID NO:1) 4 1 1 GI#13645287 (SEQTaqExp_1005013 11. 30 . 13 . 7 D7 N0:4) 1 2 3 SEQUENCE LISTING
<110> EXELIXIS, INC.

<120> MAP3Ks AS p53 PATHWAY
MODIFIERS OF AND METHODS
THE OF USE

<130> EX02-072C-PC

<150> US 60/296,076 <151> 2001-06-05 <150> US 60/328,605 <151> 2001-10-10 <150> US 60/357,253 <151> 2002-02-15 <150> US 60/361,196 <151> 2002-03-O1 <160> 9 <170> Patentln version 3.1 <210> 1 <211> 3365 <212> DNA

<213> Homo sapiens <400> 1 agcatccgga gcggagctgcagcagcgccgccttttgtgctgcggccgcggagcccccga60 gggcccagtg ttcaccatcataccaggggccagaggcgatggcttgcctccatgagaccc120 gaacaccctc tccttcctttgggggctttgtgtctaccctaagtgaggcatccatgcgca180 agctggaccc agacacttctgactgcactcccgagaaggacctgacgcctacccatgtcc240 tgcagctaca tgagcaggatgcagggggcccagggggagcagctgggtcacctgagagtc300 gggcatccag agttcgagctgacgaggtgcgactgcagtgccagagtggcagtggcttcc360 ttgagggcct ctttggctgcctgcgccctgtctggaccatgattggcaaagcctactcca420 ctgagcacaa gcagcagcaggaagacctttgggaggtcccctttgaggaaatcctggacc480 tgcagtgggt gggctcaggggcccagggtgctgtcttcctggggcgcttccacggggagg540 aggtggctgt gaagaaggtgcgagacctcaaagaaaccgacatcaagcacttgcgaaagc600 tgaagcaccc caacatcatcactttcaagggtgtgtgcacccaggctccctgctactgca660 tcctcatgga gttctgcgcccagggccagctgtatgaggtactgcgggctggccgccctg720 tcaccccctc cttactggttgactggtccatgggcatcgctggtggcatgaactacctgc780 acctgcacaa gattatccacagggatctcaagtcacccaacatgctaatcacctacgacg840 atgtggtgaa gatctcagattttggcacttccaaggagctgagtgacaagagcaccaaga900 tgtcctttgc agggacagtagcctggatggcccctgaggtgatccgcaatgaacctgtgt960 ctgagaaggtcgacatctggtcctttggcgtggtgctatgggaactgctgactggtgaga1020 tcccctacaaagacgtagattcctcagccattatctggggtgtgggaagcaacagtctcc1080 atctgcccgtgccctccagttgcccagatggtttcaagatcctgcttcgccagtgctgga1140 atagcaaaccacgaaatcgcccatcattccgacagatcctgctgcatctggacattgcct1200 cagctgatgtactctccacaccccaggagacttactttaagtcccaggcagagtggcggg1260 aagaagtaaaactgcactttgaaaagattaagtcagaagggacctgtctgcaccgcctag1320 aagaggaactggtgatgaggaggagggaggagctcagacacgccctggacatcagggagc1380 actatgaaaggaagctggagagagccaacaacctgtatatggaacttaatgccctcatgt1440 tgcagctggaactcaaggagagggagctgctcaggcgagagcaagctttagagcggaggt1500 gcccaggcctgctgaagccacacccttcccggggcctcctgcatggaaacacaatggaga1560 agcttatcaagaagaggaatgtgccacagaatctgtcaccccatagccaaaggccagata1620 tcctcaaggcggagtctttgctccctaaactagatgcagccctgagtggggtggggcttc1680 ctgggtgtcctaaggcccccccctcaccaggacggagtcgccgtggcaagacccgtcacc1740 gcaaggccagcgccaaggggagctgtggggacctgcctgggcttcgtacagctgtgccac1800 cccatgaacctggaggaccaggaagcccagggggcctaggagggggaccctcagcctggg1860 aggcctgccctcccgccctccgtgggcttcatcatgacctcctgctccgcaaaatgtctt1920 catcgtccccagacctgctgtcagcagcactagggtcccggggccggggggccacaggcg1980 gagctggggatcctggctcaccacctccggcccggggtgacaccccaccaagtgagggct2040 cagcccctggctccaccagcccagattcacctgggggagccaaaggggaaccacctcctc2100 cagtagggcctggtgaaggtgtggggcttctgggaactggaagggaagggacctcaggcc2160 ggggaggaagccgggctgggtcccagcacttgaccccagctgcactgctgtacagggctg2220 ccgtcacccgaagtcagaaacgtggcatctcatcggaagaggaggaaggagaggtagaca2280 gtgaagtagagctgacatcaagccagaggtggcctcagagcctgaacatgcgccagtcac2340 tatctaccttcagctcagagaatccatcagatggggaggaaggcacagctagtgaacctt2400 cccccagtggcacacctgaagttggcagcaccaacactgatgagcggccagatgagcggt2460 ctgatgacatgtgctcccagggctcagaaatcccactggacccacctccttcagaggtca2520 tccctggccctgaacccagctccctgcccattccacaccaggaacttctcagagagcggg2580 gccctcccaattctgaggactcagactgtgacagcactgaattggacaactccaacagcg2640 ttgatgccttgcgccccccagcttccctccctccatgaaagccactcgtattccttgtac2700 atagagaaatatttatatggattatatatatatacatatatatatatatatgcgccacat2760 aatcaacagaaagatggggctgtcccagccgtaagtcaggctcgagggagactgatcccc2820 tgaccaattcacctgataaactctagggacactggcagctgtggaaatgaatgaggcaca2880 gccgtagagctgtggctaagggcaagccccttcctgccccaccccattccttatattcag2940 caagcaacaaggcaatagaaaagccagggttgtctttatattctttatccccaaataata3000 gggggtggggggaggggcggtgggaggggcaggagagaaaaccacttagactgcactttt3060 ctgttccgtttactctgtttacacattttgcacttgggaggagggaggctaaggctgggt3120 cctcccctctgaggtttctcaggtggcaatgtaactcatttttttgtcccaccatttatc3180 ttctctgcccaagccctgtcttaaggcccagggggaggttaggagactgatagcatgtga3240 tggctcaggctgaagaaccggggttctgtttaagtccctgcttttatcctggtgcctgat3300 tggggtggggactgtcctactgtaacccctgtgaaaaaccttgaaaaataacactccatg3360 cagga 3365 <210> 2 <211> 2830 <212> DNA
<213> Homo Sapiens <220>
<221> misc_feature <222> (33) .(33) <223> "n" is A, C, G, or T
<400>

cgagggcccagtgttcaccatcataccaggggncagaggcgatggcttgcctccatgaga60 cccgaacaccctctccttcctttgggggctttgtgtctaccctaagtgaggcatccatgc120 gcaagctggacccagacacttctgactgcactcccgagaaggacctgacgcctacccagt180 gtgtacttcgagatgtggtaccccttggtgggcagggtgggggagggcccagcccctccc240 caggtggagagccgccccctgagccttttgccaacagtgtcctgcagctacatgagcagg300 atgcagggggcccagggggagcagctgggtcacctgagagtcgggcatccagagttcgag360 ctgacgaggtgcgactgcagtgccagagtggcagtggcttccttgagggcctctttggct420 gcctgcgccctgtctggaccatgattggcaaagcctactccactgagcacaagcagcagc480 aggaagacctttgggaggtcccctttgaggaaatcctggacctgcagtgggtgggctcag540 gggcccagggtgctgtcttcctggggcgcttccacggggaggaggtggctgtgaagaagg600 tgcgagacctcaaagaaaccgacatcaagcacttgcgaaagctgaagcaccccaacatca660 tcactttcaagggtgtgtgcacccaggctccctgctactgcatcctcatggagttctgcg720 cccagggccagctgtatgaggtactgcgggctggccgccctgtcaccccctccttactgg780 ttgactggtc catgggcatc gctggtggca tgaactacct gcacctgcac aagattatcc 840 acagggatct caagtcaccc aacatgctaa tcacctacga cgatgtggtg aagatctcag 900 attttggcacttccaaggagctgagtgacaagagcaccaagatgtcctttgcagggacag960 tagcctggatggcccctgaggtgatccgcaatgaacctgtgtctgagaaggtcgacatct1020 ggtcctttggcgtggtgctatgggaactgctgactggtgagatcccctacaaagacgtag1080 attcctcagccattatctggggtgtgggaagcaacagtctccatctgcccgtgccctcca1140 gttgcccagatggtttcaagatcctgcttcgccagtgctggaatagcaaaccacgaaatc1200 gcccatcattccgacagatcctgctgcatctggacattgcctcagctgatgtactctcca1260 caccccaggagacttactttaagtcccaggcagagtggcgggaagaagtaaaactgcact1320 ttgaaaagattaagtcagaagggacctgtctgcaccgcctagaagaggaactggtgatga1380 ggaggagggaggagctcagacacgccctggacatcagggagcactatgaaaggaagctgg1440 agagagccaacaacctgtatatggaacttaatgccctcatgttgcagctggaactcaagg1500 agagggagctgctcaggcgagagcaagctttagagcggaggtgcccaggcctgctgaagc1560 cacacccttcccggggcctcctgcatggaaacacaatggagaagcttatcaagaagagga1620 atgtgccacagaagctgtcaccccatagcaaaaggccagatatcctcaagacggagtctt1680 tgctccctaaactagatgcagccctgagtggggtggggcttcctgggtgtcctaagggcc1740 ccccctcaccaggacggagtcgccgtggcaagacccgtcaccgcaaggccagcgccaagg1800 ggagctgtggggacctgcctgggcttcgtacagctgtgccaccccatgaacctggaggac1860 caggaagcccagggggcctaggagggggaccctcagcctgggaggcctgccctcccgccc1920 tccgtgggcttcatcatgacctcctgctccgcaaaatgtcttcatcgtccccagacctgc1980 tgtcagcagcactagggtcccggggccggggggccacaggcggagctggggatcctggct2040 caccacctccggcccggggtgacaccccaccaagtgagggctcagcccctggctccacca2100 gcccagattcacctgggggagccaaaggggaaccacctcctccagtagggcctggtgaag2160 gtgtggggcttctgggaactggaagggaagggacctcaggccggggaggaagccgggctg2220 ggtcccagcacttgaccccagctgcactgctgtacagggctgccgtcacccgaagtcaga2280 aacgtggcatctcatcggaagaggaggaaggagaggtagacagtgaagtagagctgacat2340 caagccagaggtggcctcagagcctgaacatgcgccagtcactatctaccttcagctcag2400 agaatccatcagatggggaggaaggcacagctagtgaaccttcccccagtggcacacctg2460 aagttggcagcaccaacactgatgagcggccagatgagcggtctgatgacatgtgctccc2520 agggctcagaaatcccactggacccacctccttcagaggtcatccctggccctgaaccca2580 gctccctgcccattccacaccaggaacttctcagagagcggggccctcccaattctgagg2640 actcagactgtgacagcactgaattggacaactccaacagcgttgatgccttgcggcccc2700 cagcttccctccctccatgaaagccactcgtattccttgtacatagagaaatatttatat2760 aaattatata tatatacata tatatatata tatgcgccac ataatcaaca gaaagatggg 2820 gctgtcccag 2830 <210>

<211>

<212>
DNA

<213> sapiens Homo <400>

cgagggcccagtgttcaccatcataccaggggccagaggcgatggcttgcctccatgaga60 cccgaacaccctctccttcctttgggggctttgtgtctaccctaagtgaggcatccatgc120 gcaagctggacccagacacttctgactgcactcccgagaaggacctgacgcctacccatg180 tcctgcagctacatgagcaggatgcagggggcccagggggagcagctgggtcacctgaga240 gtcgggcatccagagttcgagctgacgaggtgcgactgcagtgccagagtggcagtggct300 tccttgagggcctctttggctgcctgcgccctgtctggaccatgattggcaaagcctact360 ccactgagcacaagcagcagcaggaagacctttgggaggtcccctttgaggaaatcctgg420 acctgcagtgggtgggctcaggggcccagggtgctgtcttcctggggcgcttccacgggg480 aggaggtggctgtgaagaaggtgcgagacctcaaagaaaccgacatcaagcacttgcgaa540 agctgaagcaccccaacatcatcactttcaagggtgtgtgcacccaggctccctgctact600 gcatcctcatggagttctgcgcccagggccagctgtatgaggtactgcgggctggccgcc660 ctgtcaccccctccttactggttgactggtccatgggcatcgctggtggcatgaactacc720 tgcacctgcacaagattatccacagggatctcaagtcacccaacatgctaatcacctacg780 acgatgtggtgaagatctcagattttggcacttccaaggagctgagtgacaagagcacca840 agatgtcctttgcagggacagtagcctggatggcccctgaggtgatccgcaatgaacctg900 tgtctgagaaggtcgacatctggtcctttggcgtggtgctatgggaactgctgactggtg960 agatcccctacaaagacgtagattcctcagccattatctggggtgtgggaagcaacagtc1020 tccatctgcccgtgccctccagttgcccagatggtttcaagatcctgcttcgccagtgct1080 ggaatagcaaaccacgaaatcgcccatcattccgacagatcctgctgcatctggacattg1140 cctcagctgatgtactctccacaccccaggagacttactttaagtcccaggcagagtggc1200 gggaagaagtaaaactgcactttgaaaagattaagtcagaagggacctgtctgcaccgcc1260 tagaagaggaactggtgatgaggaggagggaggagctcagacacgccctggacatcaggg1320 agcactatgaaaggaagctggagagagccaacaacctgtatatggaacttaatgccctca1380 tgttgcagctggaactcaaggagagggagctgctcaggcgagagcaagctttagagcgga1440 ggtgcccaggcctgctgaagccacacccttcccggggcctcctgcatggaaacacaatgg1500 agaagcttatcaagaagaggaatgtgccacagaagctgtcaccccatagcaaaaggccag1560 atatcctcaagacggagtctttgctccctaaactagatgcagccctgagtggggtggggc1620 ttcctgggtgtcctaagggccccccctcaccaggacggagtcgccgtggcaagacccgtc1680 accgcaaggccagcgccaaggggagctgtggggacctgcctgggcttcgtacagctgtgc1740 caccccatgaacctggaggaccaggaagcccagggggcctaggagggggaccctcagcct1800 gggaggcctgccctcccgccctccgtgggcttcatcatgacctcctgctccgcaaaatgt1860 cttcatcgtccccagacctgctgtcagcagcactagggtcccggggccggggggccacag1920 gcggagctggggatcctggctcaccacctccggcccggggtgacaccccaccaagtgagg1980 gctcagcccctggctccaccagcccagattcacctgggggagccaaaggggaaccacctc2040 ctccagtagggcctggtgaaggtgtggggcttctgggaactggaagggaagggacctcag2100 gccggggaggaagccgggctgggtcccagcacttgaccccagctgcactgctgtacaggg2160 ctgccgtcacccgaagtcagaaacgtggcatctcatcggaagaggaggaaggagaggtag2220 acagtgaagtagagctgacatcaagccagaggtggcctcagagcctgaacatgcgccagt2280 cactatctaccttcagctcagagaatccatcagatggggaggaaggcacagctagtgaac2340 cttcccccagtggcacacctgaagttggcagcaccaacactgatgagcggccagatgagc2400 ggtctgatgacatgtgctcccagggctcagaaatcccactggacccacctccttcagagg2460 tcatccctggccctgaacccagctccctgcccattccacaccaggaacttctcagagagc2520 ggggccctcccaattctgaggactcagactgtgacagcactgaattggacaactccaaca2580 gcgttgatgccttgcggcccccagcttccctccctccatgaaagccactcgtattccttg2640 tacatagagaaatatttatataaattatatatatatacatatatatatatatatgcgcca2700 cataatcaacagaaagatggggctgtccagcc 2732 <210>

<211>

<212>
DNA

<213> sapiens Homo <400>

gttttggagccctctcttaagtcagaactctgtcccaaaaatcttctgagtgtcatctca60 ggactttggttatactcatggcacgatggccaactttcaggagcacctgagctgctcctc120 ttctccacacttacccttcagtgaaagcaaaaccttcaatggactacaagatgagctcac180 agctatggggaaccacccttctcccaagctgctcgaggaccagcaggaaaaggggatggt240 acgaacagagctaatcgagagcgtgcacagccccgtcaccacaacagtgttgacgagcgt300 aagtgaggattccagggaccagtttgagaacagcgttcttcagctaagggaacacgatga360 atcagagacggcggtgtctcaggggaacagcaacacggtggacggagagagcacaagcgg420 aactgaagacataaagattcagttcagcaggtcaggcagtggcagtggtgggtttcttga480 aggactatttggatgcttaaggcctgtatggaatatcattgggaaggcatattccactga540 ttacaaattgcagcagcaagatacttgggaagtgccatttgaggagatctcagagctgca600 gtggctgggtagtggagcccaaggagcggtcttcttgggcaagttccgggcggaagaggt660 ggccatcaagaaagtgagagaacagaatgagacggatatcaagcatttgaggaagttgaa720 gcaccctaacatcatcgcattcaagggtgtttgtactcaggccccatgttattgtattat780 catggaatactgtgcccatggacaactctacgaggtcttacgagctggcaggaagatcac840 acctcgattgctagtagactggtccacaggaattgcaagtggaatgaattatttgcacct900 ccataaaattattcatcgtgatctcaaatcacctaatgttttagtgacccacacagatgc960 ggtaaaaatttcagattttggtacatctaaggaactcagtgacaaaagtaccaagatgtc1020 atttgctggcacggtcgcatggatggcgccagaggtgatacggaatgaacctgtctctga1080 aaaagttgatatatggtcttttggagtggtgctttgggagctgctgacaggagagatccc1140 ttacaaagatgtagattcttcagccattatctggggtgttggaagcaacagcctccacct1200 tccagttccttccacttgccctgatggattcaaaatccttatgaaacagacgtggcagag1260 taaacctcgaaaccgaccttcttttcggcagacactcatgcatttagacattgcctctgc1320 agatgtacttgccaccccacaagaaacttacttcaagtctcaggctgaatggagagaaga1380 agtgaaaaaacattttgagaagatcaaaagtgaaggaacttgtatacaccggttagatga1440 agaactgattcgaaggcgcagagaagagctcaggcatgcgctggatattcgtgaacacta1500 tgagcggaagcttgagcgggcgaataatttatacatggaattgagtgccatcatgctgca1560 gctagaaatgcgggagaaggagctcattaagcgtgagcaagcagtggaaaagaagtatcc1620 tgggacctacaaacgacaccctgttcgtcctatcatccatcccaatgccatggagaaact1680 catgaaaaggaaaggagtgcctcacaaatctgggatgcagaccaaacggccagacttgtt1740 gagatcagaagggatccccaccacagaagtggctcccactgcatcccctttgtccggaag1800 tcccaaaatgtccacttctagcagcaagagccgatatcgaagcaaaccacgccaccgccg1860 agggaatagcagaggcagccatagtgactttgccgcaatcttgaaaaaccagccagccca1920 ggaaaattcaccccatcccacttacctgcaccaagctcaatcccaatacccttctcttca1980 tcaccataattctctgcagcagcaataccagcagccccctcctgccatgtcccagagtca2040 ccatcccagactcaatatgcacggacaggacatagcaacctgcgccaacaacctgaggta2100 tttcggcccagcagcagccctgcggagcccactcagcaaccatgCtcagagacagctgcc2160 cggctcgagccctgacctcatctccacagccatggctgcagactgctggagaagttctga2220 gcctgacaagggccaagctggtccctggggctgttgccaggctgacgcttatgacccctg2280 ccttcagtgcaggccagaacagtatgggtccttagacataccctctgctgagccagtggg2340 gaggagccctgacctttccaagtcaccagcacataatcctctcttggaaaacgcccagag2400 ttctgagaaaacggaagaaaatgaattcagcggctgtaggtctgagtcatccctcggcac2460 ctctcatctcggcacccctccagcgctacctcgaaaaacaaggcctctgcagaagagtgg2520 agatgactcctcagaagaggaagaaggggaagtagatagtgaagttgaatttccacgaag2580 acagaggccccatcgctgtatcagcagctgccagtcatattcaacctttagctctgagaa2640 tttctctgtgtctgatggagaagagggaaataccagtgaccactcaaacagtcctgatga2700 gttagctgataaacttgaagaccgcttggcagagaagctagacgacctgctgtcccagac2760 gccagagattcccattgacatatcctcacactcggatgggctctctgacaaggagtgtgc2820 cgtgcgccgtgtgaagactcagatgtctctgggcaagctgtgtgtggaggaacgtggcta2880 tgagaaccccatgcagtttgaagaatcggactgtgactcttcagatggggagtgttctga2940 tgccacagttaggaccaataaacactacagctctgctacctggtaatgaaggaatacaca3000 tcctgaagatctcgtgactatactggcatttcagatccaccccacccccagactcatccc3060 actctctcccagcattttgtctgggaagagagactaccccatctttaccaccccctagaa3120 atgagctgcaataacaggaacatgagacttcgcaaatctctggaaaataatatccaaatg3180 aaattaagtctcactgaacatttcaatcaagaatggcagggatctattttattgaatatt3240 ctagctactgtaacattgatatttatttttgtttgacattttaacactttgtactgcaaa3300 gagtgaactatatatgagatagagagacaataatttcttgcaaaaaaaaaaagagataaa3360 agaaagaaca gaaaaaaa 3378 <210> 5 <211> 3569 <212> DNA
<213> Homo sapiens <220>
<221> misc_feature <222> (3283)..(3283) <223> "n" is A, C, G, or T
<400> 5 aatttacatc cattcatgaa tctgtgacgt cagcaagcct ttgggctcct ttgcggtggg 60 ctggaggattgtgtgggtggaatccccctcccctttatttttccaattctgcaaggcttt120 taaaattcaccttacatcttttcaaagcaagaaaatggaacagcatgtgtaggaattctt180 cgttgttgttttggagccctctcttaagtcagaactctgtcccaaaaatcttctgagtgt240 catctcaggactttggttatactcatggcacgatggccaactttcaggagcacctgagct300 g gctcctcttctccacacttacccttcagtgaaagcaaaaccttcaatggactacaagatg360 agctcacagctatggggaaccacccttctcccaagctgctcgaggaccagcaggaaaagg420 ggatggtacgaacagagctaatcgagagcgtgcacagccccgtcaccacaacagtgttga480 cgagcgtaagtgaggattccagggaccagtttgagaacagcgttcttcagctaagggaac540 acgatgaatcagagacggcggtgtctcaggggaacagcaacacggtggacggagagagca600 caagcggaactgaagacataaagattcagttcagcaggtcaggcagtggcagtggtgggt660 ttcttgaaggactatttggatgcttaaggcctgtatggaatatcattgggaaggcatatt720 ccactgattacaaattgcagcagcaagatacttgggaagtgccatttgaggagatctcag780 agctgcagtggctgggtagtggagcccaaggagcggtcttcttgggcaagttccgggcgg840 aagaggtggccatcaagaaagtgagagaacagaatgagacggatatcaagcatttgagga900 agttgaagcaccctaacatcatcgcattcaagggtgtttgtactcaggccccatgttatt960 gtattatcatggaatactgtgcccatggacaactctacgaggtcttacgagctggcagga1020 agatcacacctcgattgctagtagactggtccacaggaattgcaagtggaatgaattatt1080 tgcacctccataaaattattcatcgtgatctcaaatcacctaatgttttagtgacccaca1140 cagatgcggtaaaaatttcagattttggtacatctaaggaactcagtgacaaaagtacca1200 agatgtcatttgctggcacggtcgcatggatggcgccagaggtgatacggaatgaacctg1260 tctctgaaaaagttgatatatggtcttttggagtggtgctttgggagctgctgacaggag1320 agatcccttacaaagatgtagattcttcagccattatctggggtgttggaagcaacagcc1380 tccaccttccagttccttccacttgccctgatggattcaaaatccttatgaaacagacgt1440 ggcagagtaaacctcgaaaccgaccttcttttcggcagacactcatgcatttagacattg1500 cctctgcagatgtacttgccaccccacaagaaacttacttcaagtctcaggctgaatgga1560 gagaagaagtgaaaaaacattttgagaagatcaaaagtgaaggaacttgtatacaccggt1620 tagatgaagaactgattcgaaggcgcagagaagagctcaggcatgcgctggatattcgtg1680 aacactatgagcggaagcttgagcgggcgaataatttatacatggaattgagtgccatca1740 tgctgcagctagaaatgcgggagaaggagctcattaagcgtgagcaagcagtggaaaaga1800 agtatcctgggacctacaaacgacaccctgttcgtcctatcatccatcccaatgccatgg1860 agaaactcatgaaaaggaaaggagtgcctcacaaatctgggatgcagaccaaacggccag1920 acttgttgagatcagaagggatccccaccacagaagtggctcccactgcatcccctttgt1980 ccggaagtcccaaaatgtccacttctagcagcaagagccgatatcgaagcaaaccacgcc2040 accgccgagggaatagcagaggcagccatagtgactttgccgcaatcttgaaaaaccagc2100 cagcccaggaaaattcaccccatcccacttacctgcaccaagctcaatcccaataccctt2160 ctcttcatca ccataattct ctgcagcagc aataccagca gccccctcct gccatgtccc 2220 agagtcacca tcccagactc aatatgcacg gacaggacat agcaacctgc gccaacaacc 2280 tgaggtattt cggcccagca gcagccctgc ggagcccact cagcaaccat gctcagagac 2340 agctgcccgg ctcgagccct gacctcatct ccacagccat ggctgcagac tgctggagaa 2400 gttctgagcc tgacaagggc caagctggtc cctggggctg ttgccaggct gacgcttatg 2460 acccctgcct tcagtgcagg ccagaacagt atgggtcctt agacataccc tctgctgagc 2520 cagtggggag gagccctgac ctttccaagt caccagcaca taatcctctc ttggaaaacg 2580 cccagagttctgagaaaacggaagaaaatgaattcagcggctgtaggtctgagtcatccc2640 tcggcacctctcatctcggcacccctccagcgctacctcgaaaaacaaggcctctgcaga2700 agagtggagatgactcctcagaagaggaagaaggggaagtagatagtgaagttgaatttc2760 cacgaagacagaggccccatcgctgtatcagcagctgccagtcatattcaacctttagct2820 ctgagaatttctctgtgtctgatggagaagagggaaataccagtgaccactcaaacagtc2880 ctgatgagttagctgataaacttgaagaccgcttggcagagaagctagacgacctgctgt2940 cccagacgccagagattcccattgacatatcctcacactcggatgggctctctgacaagg3000 agtgtgccgtgcgccgtgtgaagactcagatgtctctgggcaagctgtgtgtggaggaac3060 gtggctatgagaaccccatgcagtttgaagaatcggactgtgactcttcagatggggagt3120 gttctgatgccacagttaggaccaataaacactacagctctgctacctggtaatgaagga3180 atacacatcctgaagatctcgtgactatactggcatttcagatccaccccacccccagac3240 tcatcccactctctcccagcattttgtctgggaagagagactnacccatctttacccacc3300 ccctagaaatgagctgcaataacaggaacatgagacttcgcaaatctctggaaaataata3360 tccaaatgaaattaagtctcactgaacatttcaatcaagaatggcagggatctattttat3420 tgaatattctagctactgtaacattgatatttatttttgtttgacattttaacactttgt3480 actgcaaagagtgaactatatatgagatagagagacaataatttcttgcaaaaaaaaaaa3540 gagataaaagaaagaacaaaaaaaaaaaa 3569 <210> 6 <211> 2910 <212> DNA
<213> Homo sapiens <400> 6 acgatggcca actttcagga gcacctgagc tgctcctctt ctccacactt acccttcagt 60 gaaagcaaaa ccttcaatgg actacaagat gagctcacag ctatggggaa ccacccttct 120 cccaagctgc tcgaggacca gcaggaaaag gggatggtac gaacagagct aatcgagagc 180 gtgcacagcc ccgtcaccac aacagtgttg acgagcgtaa gtgaggattc cagggaccag 240 1~

tttgagaacagcgttcttcagctaagggaacacgatgaatcagagacggcggtgtctcag300 gggaacagcaacacggtggacggagagagcacaagtggaactgaagacataaagattcag360 ttcagcaggtcaggcagtggcagtggtgggtttcttgaaggactatttggatgcttaagg420 cctgtatggaatatcattgggaaggcatattccactgattacaaattgcagcagcaagat480 acttgggaagtgccatttgaggagatctcagagctgcagtggctgggtagtggagcccaa540 ggagcggtcttcttgggcaagttccgggcggaagaggtggccatcaagaaagtgagagaa600 cagaatgagacggatatcaagcatttgaggaagttgaagcaccctaacatcatcgcattc660 aagggtgtttgtactcaggccccatgttattgtattatcatggaatactgtgcccatgga720 caactctacgaggtcttacgagctggcaggaagatcacacctcgattgctagtagactgg780 tccacaggaattgcaagtggaatgaattatttgcacctccataaaattattcatcgtgat840 ctcaaatcacctaatgttttagtgacccacacagatgcggtaaaaatttcagattttggt900 acatctaaggaactcagtgacaaaagtaccaagatgtcatttgctggcacggtcgcatgg960 atggcgccagaggtgatacggaatgaacctgtctctgaaaaagttgatatatggtctttt1020 ggagtggtgctttgggagctgctgacaggagagatcccttacaaagatgtagattcttca1080 gccattatctggggtgttggaagcaacagcctccaccttccagttccttccacttgccct1140 gatggattcaaaatccttatgaaacagacgtggcagagtaaacctcgaaaccgaccttct1200 tttcggcagacactcatgcatttagacattgcctctgcagatgtacttgccaccccacaa1260 gaaacttacttcaagtctcaggctgaatggagagaagaagtgaaaaaacattttgagaag1320 atcaaaagtgaaggaacttgtatacaccggttagatgaagaactgattcgaaggcgcaga1380 gaagagctcaggcatgcgctggatattcgtgaacactatgagcggaagcttgagcgggcg1440 aataatttatacatggaattgagtgccatcatgctgcagctagaaatgcgggagaaggag1500 ctcattaagcgtgagcaagcagtggaaaagaagtatcctgggacctacaaacgacaccct1560 gttcgtcctatcatccatcccaatgccatggagaaactcatgaaaaggaaaggagtgcct1620 cacaaatctgggatgcagaccaaacggccagacttgttgagatcagaagggatccccacc1680 acagaagtggctcccactgcatcccctttgtccggaagtcccaaaatgtccacttctagc1740 agcaagagccgatatcgaagcaaaccacgccaccgccgagggaatagcagaggcagccat1800 agtgactttgccgcaatcttgaaaaaccagccagcccaggaaaattcaccccatcccact1860 tacctgcaccaagctcaatcccaatacccttctcttcatcaccataattctctgcagcag1920 caataccagcagccccctcctgccatgtcccagagtcaccatcccagactcaatatgcac1980 ggacaggacatagcaacctgcgccaacaacctgaggtatttcggcccagcagcagccctg2040 cggagcccactcagcaaccatgctcagagacagctgcccggctcgagccctgacctcatc2100 tccacagccatggctgcagactgctggagaagttctgagcctgacaagggccaagctggt2160 ccctggggctgttgccaggctgacgcttatgacccctgccttcagtgcaggccagaacag2220 tatgggtccttagacataccctctgctgagccagtggggaggagccctgacctttccaag2280 tcaccagcacataatcctctcttggaaaacgcccagagttctgagaaaacggaagaaaat2340 gaattcagcggctgtaggtctgagtcatccctcggcacctctcatctcggcacccctcca2400 gcgctacctcgaaaaacaaggcctctgcagaagagtggagatgactcctcagaagaggaa2460 gaaggggaagtagatagtgaagttgaatttccacgaagacagaggccccatcgctgtatc2520 agcagctgccagtcatattcaacctttagctctgagaatttctctgtgtctgatggagaa2580 gagggaaataccagtgaccactcaaacagtcctgatgagttagctgataaacttgaagac2640 cgcttggcagagaagctagacgacctgctgtcccagacgccagagattcccattgacata2700 tcctcacactcggatgggctctctgacaaggagtgtgccgtgcgccgtgtgaagactcag2760 atgtctctgggcaagctgtgtgtggaggaacgtggctatgagaaccccatgcagtttgaa2820 gaatcggactgtgactcttcagatggggagtgttctgatgccacagttaggaccaataaa2880 cactacagctctgctacctggtaatgaagg 2910 <210> 7 <211> 333 <212> DNA
<213> Homo sapiens <400> 7 tacctataca tggagtattg tgcccatgga caactctacg aggtcttacg agctggcagg 60 aagatcacac ctcgattgct agtagactgg tccacaggaa ttgcaagtgg aatgaattat 120 ttgcacctcc ataaaattat tcatcgtgat ctcaaatcac ctaatgtttt agtgacccac 180 acagatgcggtaaaaatttcagattttggtacatctaagg aactcagtga caaaagtacc240 aagatgtcatttgctggcacggtcgcatggatggcgccag aggtgatacg gaatgaacct300 gtctctgaaaaagttgatatctggtctatggta 333 <210> 8 <211> 859 <212> PRT
<213> Homo sapiens <400> 8 Met Ala Cys Leu His Glu Thr Arg Thr Pro Ser Pro Ser Phe Gly Gly Phe Val Ser Thr Leu Ser Glu Ala Ser Met Arg Lys Leu Asp Pro Asp Thr Ser Asp Cys Thr Pro Glu Lys Asp Leu Thr Pro Thr His Val Leu Gln Leu His Glu Gln Asp Ala Gly Gly Pro Gly Gly Ala Ala Gly Ser Pro Glu Ser Arg Ala Ser Arg Val Arg Ala Asp Glu Val Arg Leu Gln Cys Gln Ser Gly Ser Gly Phe Leu Glu Gly Leu Phe Gly Cys Leu Arg Pro Val Trp Thr Met Ile Gly Lys Ala Tyr Ser Thr Glu His Lys Gln Gln Gln Glu Asp Leu Trp Glu Val Pro Phe Glu Glu Ile Leu Asp Leu Gln Trp Val Gly Ser Gly Ala Gln Gly Ala Val Phe Leu Gly Arg Phe His Gly Glu Glu Val Ala Val Lys Lys Val Arg Asp Leu Lys Glu Thr Asp Ile Lys His Leu Arg Lys Leu Lys His Pro Asn Ile Ile Thr Phe Lys Gly Val Cys Thr Gln Ala Pro Cys Tyr Cys Ile Leu Met Glu Phe Cys Ala Gln Gly Gln Leu Tyr Glu Val Leu Arg Ala Gly Arg Pro Val Thr Pro Ser Leu Leu Val Asp Trp Ser Met Gly Ile Ala Gly Gly Met Asn Tyr Leu His Leu His Lys Ile Ile His Arg Asp Leu Lys Ser Pro Asn Met Leu Ile Thr Tyr Asp Asp Val Val Lys Ile Ser Asp Phe Gly Thr Ser Lys Glu Leu Ser Asp Lys Ser Thr Lys Met Ser Phe Ala Gly Phe Val Ser Thr Leu Ser Glu Thr Val Ala Trp Met Ala Pro Glu Val Ile Arg Asn Glu Pro Val Ser Glu Lys Val Asp Ile Trp Ser Phe Gly Val Val Leu Trp Glu Leu Leu Thr Gly Glu Ile Pro Tyr Lys Asp Val Asp Ser Ser Ala Ile Ile Trp Gly Val Gly Ser Asn Ser Leu His Leu Pro Val Pro Ser Ser Cys Pro Asp Gly Phe Lys Ile Leu Leu Arg Gln Cys Trp Asn Ser Lys Pro Arg Asn Arg Pro Ser Phe Arg Gln Ile Leu Leu His Leu Asp Ile Ala Ser Ala Asp Val Leu Ser Thr Pro Gln Glu Thr Tyr Phe Lys Ser Gln Ala Glu Trp Arg Glu Glu Val Lys Leu His Phe Glu Lys Ile Lys Ser Glu Gly Thr Cys Leu His Arg Leu Glu Glu Glu Leu Val Met Arg Arg Arg Glu Glu Leu Arg His Ala Leu Asp Ile Arg Glu His Tyr Glu Arg Lys Leu Glu Arg Ala Asn Asn Leu Tyr Met Glu Leu Asn Ala Leu Met Leu Gln Leu Glu Leu Lys Glu Arg Glu Leu Leu Arg Arg Glu Gln Ala Leu Glu Arg Arg Cys Pro Gly Leu Leu Lys Pro His Pro Ser Arg Gly Leu Leu His Gly Asn Thr Met Glu Lys Leu Ile Lys Lys Arg Asn Val Pro Gln Asn Leu Ser Pro His Ser Gln Arg Pro Asp Ile Leu Lys Ala Glu Ser Leu Leu Pro Lys Leu Asp Ala Ala Leu Ser Gly Val Gly Leu Pro Gly Cys Pro Lys Ala Pro Pro Ser Pro Gly Arg Ser Arg Arg Gly Lys Thr Arg His Arg Lys Ala Ser Ala Lys Gly Ser Cys Gly Asp Leu Pro Gly Leu Arg Thr Ala Val Pro Pro His Glu Pro Gly Gly Pro Gly Ser Pro Gly Gly Leu Gly Gly Gly Pro Ser Ala Trp Glu Ala Cys Pro Pro Ala Leu Arg Gly Leu His His Asp Leu Leu Leu Arg Lys Met Ser Ser Ser Ser Pro Asp Leu Leu Ser Ala Ala Leu Gly Ser Arg Gly Arg Gly Ala Thr Gly Gly Ala Gly Asp Pro Gly Ser Pro Pro Pro Ala Arg Gly Asp Thr Pro Pro Ser Glu Gly Ser Ala Pro Gly Ser Thr Ser Pro Asp Ser Pro Gly Gly Ala Lys Gly Glu Pro Pro Pro Pro Val Gly Pro Gly Glu Gly Val Gly Leu Leu Gly Thr Gly Arg Glu Gly Thr Ser Gly Arg Gly Gly Ser Arg Ala Gly Ser Gln His Leu Thr Pro Ala Ala Leu Leu Tyr Arg Ala Ala Val Thr Arg Ser Gln Lys Arg Gly Ile Ser Ser Glu Glu Glu Glu Gly Glu Val Asp Ser Glu Val Glu Leu Thr Ser Ser Gln Arg Trp Pro Gln Ser Leu Asn Met Arg Gln Ser Leu Ser Thr Phe Ser Ser Glu Asn Pro Ser Asp Gly Glu Glu Gly Thr Ala Ser Glu Pro Ser Pro Ser Gly Thr Pro Glu Val Gly Ser Thr Asn Thr Asp Glu Arg Pro Asp Glu Arg Ser Asp Asp Met Cys Ser Gln Gly Ser Glu Ile Pro Leu Asp Pro Pro Pro Ser Glu Val Ile Pro Gly Pro Glu Pro Ser Ser Leu Pro Ile Pro His Gln Glu Leu Leu Arg Glu Arg Gly Pro Pro Asn Ser Glu Asp Ser Asp Cys Asp Ser Thr Glu Leu Asp Asn Ser Asn Ser Val Asp Ala Leu Arg Pro Pro Ala Ser Leu Pro Pro <210> 9 <211> 966 <212> PRT
<213> Homo Sapiens <400> 9 Met Ala Asn Phe Gln Glu His Leu Ser Cys Ser Ser Ser Pro His Leu Pro Phe Ser Glu Ser Lys Thr Phe Asn Gly Leu Gln Asp Glu Leu Thr Ala Met Gly Asn His Pro Ser Pro Lys Leu Leu Glu Asp Gln Gln Glu Lys Gly Met Val Arg Thr Glu Leu Ile Glu Ser Val His Ser Pro Val Thr Thr Thr Val Leu Thr Ser Val Ser Glu Asp Ser Arg Asp Gln Phe Glu Asn Ser Val Leu Gln Leu Arg Glu His Asp Glu Ser Glu Thr Ala Val Ser Gln G1y Asn Ser Asn Thr Val Asp Gly Glu Ser Thr Ser Gly Thr Glu Asp Ile Lys Ile Gln Phe Ser Arg Ser Gly Ser Gly Ser Gly Gly Phe Leu Glu Gly Leu Phe Gly Cys Leu Arg Pro Val Trp Asn Ile Ile Gly Lys Ala Tyr Ser Thr Asp Tyr Lys Leu Gln Gln Gln Asp Thr Trp Glu Val Pro Phe Glu Glu Ile Ser Glu Leu Gln Trp Leu Gly Ser Gly Ala Gln Gly Ala Val Phe Leu Gly Lys Phe Arg Ala Glu Glu Val Ala Ile Lys Lys Val Arg Glu Gln Asn Glu Thr Asp Ile Lys His Leu Arg Lys Leu Lys His Pro Asn Ile Ile Ala Phe Lys Gly Val Cys Thr Gln Ala Pro Cys Tyr Cys Ile Ile Met Glu Tyr Cys Ala His Gly Gln Leu Tyr Glu Val Leu Arg Ala Gly Arg Lys Ile Thr Pro Arg Leu Leu Val Asp Trp Ser Thr Gly Ile Ala Ser Gly Met Asn Tyr Leu His Leu His Lys Ile Ile His Arg Asp Leu Lys Ser Pro Asn Val Leu Val Thr His Thr Asp Ala Val Lys Ile Ser Asp Phe Gly Thr Ser Lys Glu Leu Ser Asp Lys Ser Thr Lys Met Ser Phe Ala Gly Thr Val Ala Trp Met Ala Pro Glu Val Ile Arg Asn Glu Pro Val Ser Glu Lys Val Asp Ile Trp Ser Phe Gly Val Val Leu Trp Glu Leu Leu Thr Gly Glu Ile Pro Tyr Lys Asp Val Asp Ser Ser Ala Ile Ile Trp Gly Val Gly Ser Asn Ser Leu His Leu Pro Val Pro Ser Thr Cys Pro Asp Gly Phe Lys Ile 1~

Leu Met Lys Gln Thr Trp Gln Ser Lys Pro Arg Asn Arg Pro Ser Phe Arg Gln Thr Leu Met His Leu Asp Ile Ala Ser Ala Asp Val Leu Ala Thr Pro Gln Glu Thr Tyr Phe Lys Ser Gln Ala Glu Trp Arg Glu Glu Val Lys Lys His Phe Glu Lys Ile Lys Ser Glu Gly Thr Cys Ile His Arg Leu Asp Glu Glu Leu Ile Arg Arg Arg Arg Glu Glu Leu Arg His Ala Leu Asp Ile Arg Glu His Tyr Glu Arg Lys Leu Glu Arg Ala Asn Asn Leu Tyr Met Glu Leu Ser Ala Ile Met Leu Gln Leu Glu Met Arg Glu Lys Glu Leu Ile Lys Arg Glu Gln Ala Val Glu Lys Lys Tyr Pro Gly Thr Tyr Lys Arg His Pro Val Arg Pro Ile Ile His Pro Asn Ala Met Glu Lys Leu Met Lys Arg Lys Gly Val Pro His Lys Ser Gly Met Gln Thr Lys Arg Pro Asp Leu Leu Arg Ser Glu Gly Ile Pro Thr Thr Glu Val Ala Pro Thr Ala Ser Pro Leu Ser Gly Ser Pro Lys Met Ser Thr Ser Ser Ser Lys Ser Arg Tyr Arg Ser Lys Pro Arg His Arg Arg Gly Asn Ser Arg Gly Ser His Ser Asp Phe Ala Ala Ile Leu Lys Asn Gln Pro Ala Gln Glu Asn Ser Pro His Pro Thr Tyr Leu His Gln Ala Gln Ser Gln Tyr Pro Ser Leu His His His Asn Ser Leu Gln Gln Gln Tyr Gln Gln Pro Pro Pro Ala Met Ser Gln Ser His His Pro Arg Leu Asn Met His Gly Gln Asp Ile Ala Thr Cys Ala Asn Asn Leu Arg Tyr Phe Gly Pro Ala Ala Ala Leu Arg Ser Pro Leu Ser Asn His Ala Gln Arg Gln Leu Pro Gly Ser Ser Pro Asp Leu Ile Ser Thr Ala Met Ala Ala Asp Cys Trp Arg Ser Ser Glu Pro Asp Lys Gly Gln Ala Gly Pro Trp Gly Cys Cys Gln Ala Asp Ala Tyr Asp Pro Cys Leu Gln Cys Arg Pro Glu Gln Tyr Gly Ser Leu Asp Ile Pro Ser Ala Glu Pro Val Gly Arg Ser Pro Asp Leu Ser Lys Ser Pro Ala His Asn Pro Leu Leu Glu Asn Ala Gln Ser Ser Glu Lys Thr Glu Glu Asn Glu Phe Ser Gly Cys Arg Ser Glu Ser Ser Leu Gly Thr Ser His Leu Gly Thr Pro Pro Ala Leu Pro Arg Lys Thr Arg Pro Leu Gln Lys Ser Gly Asp Asp Ser Ser Glu Glu Glu Glu Gly Glu Val Asp Ser Glu Val Glu Phe Pro Arg Arg Gln Arg Pro His Arg Cys Ile Ser Ser Cys Gln Ser Tyr Ser Thr Phe Ser Ser Glu Asn Phe Ser Val Ser Asp Gly Glu Glu Gly Asn Thr Ser Asp His Ser Asn Ser Pro Asp Glu Leu Ala Asp Lys Leu Glu Asp Arg Leu Ala Glu Lys Leu Asp Asp Leu Leu Ser Gln Thr Pro Glu Ile Pro Ile Asp Ile Ser Ser His Ser Asp Gly Leu Ser Asp Lys Glu Cys Ala Val Arg Arg Val Lys Thr Gln Met Ser Leu Gly Lys Leu Cys Val Glu Glu Arg Gly Tyr Glu Asn Pro Met Gln Phe Glu Glu Ser Asp Cys Asp Ser Ser Asp Gly Glu Cys Ser Asp Ala Thr Val Arg Thr Asn Lys His Tyr Ser Ser Ala Thr Trp

Claims (25)

  1. WHAT IS CLAIMED IS:

    A method of identifying a candidate p53 pathway modulating agent, said method comprising the steps of:

    (a) providing an assay system comprising a purified MAP3K polypeptide or nucleic acid or a functionally active fragment or derivative thereof;
    (b) contacting the assay system with a test agent under conditions whereby, but for the presence of the test agent, the system provides a reference activity; and (c) detecting a test agent-biased activity of the assay system, wherein a difference between the test agent-biased activity and the reference activity identifies the test agent as a candidate p53 pathway modulating agent.
  2. 2. The method of Claim 1 wherein the assay system comprises cultured cells that express the MAP3K polypeptide.
  3. 3. The method of Claim 2 wherein the cultured cells additionally have defective p53 function.
  4. 4. The method of Claim 1 wherein the assay system includes a screening assay comprising a MAP3K polypeptide, and the candidate test agent is a small molecule modulator.
  5. 5. The method of Claim 4 wherein the assay is a kinase assay.
  6. 6. The method of Claim 1 wherein the assay system is selected from the group consisting of an apoptosis assay system, a cell proliferation assay system, an angiogenesis assay system, and a hypoxic induction assay system.
  7. 7. The method of Claim 1 wherein the assay system includes a binding assay comprising a MAP3K polypeptide and the candidate test agent is an antibody.
  8. 8. The method of Claim 1 wherein the assay system includes an expression assay comprising a MAP3K nucleic acid and the candidate test agent is a nucleic acid modulator.
  9. 9. The method of claim 8 wherein the nucleic acid modulator is an antisense oligomer.
  10. 10. The method of Claim 8 wherein the nucleic acid modulator is a PMO.
  11. 11. The method of Claim 1 additionally comprising:

    (d) administering the candidate p53 pathway modulating agent identified in (c) to a model system comprising cells defective in p53 function and, detecting a phenotypic change in the model system that indicates that the p53 function is restored.
  12. 12. The method of Claim 11 wherein the model system is a mouse model with defective p53 function.
  13. 13. A method for modulating a p53 pathway of a cell comprising contacting a cell defective in p53 function with a candidate modulator that specifically binds to a MAP3K
    polypeptide comprising an amino acid sequence selected from group consisting of SEQ ID
    NOs: 8 and 9, whereby p53 function is restored.
  14. 14. The method of claim 13 wherein the candidate modulator is administered to a vertebrate animal predetermined to have a disease or disorder resulting from a defect in p53 function.
  15. 15. The method of Claim 13 wherein the candidate modulator is selected from the group consisting of an antibody and a small molecule.
  16. 16. The method of Claim 1, comprising the additional steps of:

    (d) providing a secondary assay system comprising cultured cells or a non-human animal expressing MAP3K, (e) contacting the secondary assay system with the test agent of (b) or an agent derived therefrom under conditions whereby, but for the presence of the test agent or agent derived therefrom, the system provides a reference activity; and (f) detecting an agent-biased activity of the second assay system, wherein a difference between the agent-biased activity and the reference activity of the second assay system confirms the test agent or agent derived therefrom as a candidate p53 pathway modulating agent, and wherein the second assay detects an agent-biased change in the p53 pathway.
  17. 17. The method of Claim 16 wherein the secondary assay system comprises cultured cells.
  18. 18. The method of Claim 16 wherein the secondary assay system comprises a non-human animal.
  19. 19. The method of Claim 18 wherein the non-human animal mis-expresses a p53 pathway gene.
  20. 20. A method of modulating p53 pathway in a mammalian cell comprising contacting the cell with an agent that specifically binds a MAP3K polypeptide or nucleic acid.
  21. 21. The method of Claim 20 wherein the agent is administered to a mammalian animal predetermined to have a pathology associated with the p53 pathway.
  22. 22. The method of Claim 20 wherein the agent is a small molecule modulator, a nucleic acid modulator, or an antibody.
  23. 23. A method for diagnosing a disease in a patient comprising:
    (a) obtaining a biological sample from the patient;
    (b) contacting the sample with a probe for MAP3K expression;
    (c) comparing results from step (b) with a control;
    (d) determining whether step (c) indicates a likelihood of disease.
  24. 24. The method of claim 23 wherein said disease is cancer.
  25. 25. The method according to claim 24, wherein said cancer is a cancer as shown in Table 1 as having >25% expression level.
CA002449479A 2001-06-05 2002-06-03 Map3ks as modifiers of the p53 pathway and methods of use Abandoned CA2449479A1 (en)

Applications Claiming Priority (9)

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
PCT/US2002/017457 WO2002098889A2 (en) 2001-06-05 2002-06-03 MAP3Ks AS MODIFIER OF THE p53 PATHWAY AND METHODS OF USE

Publications (1)

Publication Number Publication Date
CA2449479A1 true CA2449479A1 (en) 2002-12-12

Family

ID=27501675

Family Applications (2)

Application Number Title Priority Date Filing Date
CA002449479A Abandoned CA2449479A1 (en) 2001-06-05 2002-06-03 Map3ks as modifiers of the p53 pathway and methods of use
CA002448112A Abandoned CA2448112A1 (en) 2001-06-05 2002-06-03 Lces as modifiers of the p53 pathway and methods of use

Family Applications After (1)

Application Number Title Priority Date Filing Date
CA002448112A Abandoned CA2448112A1 (en) 2001-06-05 2002-06-03 Lces as modifiers of the p53 pathway and methods of use

Country Status (5)

Country Link
EP (2) EP1402262A4 (en)
JP (2) JP2004532638A (en)
AU (1) AU2002314887A1 (en)
CA (2) CA2449479A1 (en)
WO (4) WO2002098889A2 (en)

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE10254601A1 (en) * 2002-11-22 2004-06-03 Ganymed Pharmaceuticals Ag Gene products differentially expressed in tumors and their use
AU2012233044B2 (en) * 2002-11-22 2015-09-03 Ganymed Pharmaceuticals Ag Genetic products differentially expressed in tumors and the use thereof
EP1572118A4 (en) * 2002-12-20 2010-07-14 Millennium Pharm Inc Methods and compositions for treating cancer using 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, 216
DE10316701A1 (en) * 2003-04-09 2004-11-04 Hinzmann, Bernd, Dr. New nucleic acid, and derived proteins, useful for diagnosis of bronchial cancer and in screening for therapeutic and diagnostic agents
WO2005005665A1 (en) 2003-07-11 2005-01-20 Banyu Pharmaceutical Co., Ltd. Method of evaluating compound efficacious in treating obesity
US20070087984A1 (en) * 2003-09-04 2007-04-19 Xiuyuan Hu Method of identifying agents that inhibit the growth of cancer cells
WO2005083425A2 (en) * 2004-02-26 2005-09-09 Bayer Healthcare Ag Diagnostics and therapeutics for diseases associated with mitogen-activated protein kinase kinase kinase 13 (map3k13)
US7557367B2 (en) 2004-06-04 2009-07-07 The Board Of Trustees Of The University Of Illinois Stretchable semiconductor elements and stretchable electrical circuits
CN102176486B (en) 2007-01-17 2015-06-24 伊利诺伊大学评议会 Optical systems fabricated by printing-based assembly
KR101870690B1 (en) 2009-05-12 2018-06-25 더 보드 오브 트러스티즈 오브 더 유니버시티 오브 일리노이 Printed assemblies of ultrathin, microscale inorganic light emitting diodes for deformable and semitransparent displays
US9442285B2 (en) 2011-01-14 2016-09-13 The Board Of Trustees Of The University Of Illinois Optical component array having adjustable curvature
WO2012158709A1 (en) 2011-05-16 2012-11-22 The Board Of Trustees Of The University Of Illinois Thermally managed led arrays assembled by printing
EP2713863B1 (en) 2011-06-03 2020-01-15 The Board of Trustees of the University of Illionis Conformable actively multiplexed high-density surface electrode array for brain interfacing
CN104472023B (en) 2011-12-01 2018-03-27 伊利诺伊大学评议会 It is designed to undergo the transient state device of programmable transformation
CN114181944B (en) * 2020-09-14 2023-10-03 中国科学院动物研究所 Mutant gene, method for constructing miniature pig model of short-limb dwarfism and application

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0572478B1 (en) * 1991-02-22 2005-09-07 The Walter And Eliza Hall Institute Of Medical Research Hbgad and higad polypeptides and nucleic acids and their use for the diagnosis and treatment of gad autoantigen associated diseases
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 (en) * 1998-02-13 1999-08-19 Cadus Pharmaceutical Corporation Mekk1 proteins and fragments thereof for use in regulating apoptosis
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
JP2002538826A (en) * 1999-03-18 2002-11-19 ザ ユニバーシティ オブ ブリストル Polyunsaturated fatty acid (PUFA) elongase from Kaenorabuditis elegans
JP2002542782A (en) * 1999-04-23 2002-12-17 インサイト・ゲノミックス・インコーポレイテッド Human membrane-related protein
CA2342831A1 (en) * 1999-07-22 2001-02-01 The University Of British Columbia A plant long chain fatty acid biosynthetic enzyme
AU1472801A (en) * 1999-11-12 2001-06-06 Human Genome Sciences, Inc. 24 human secreted proteins
US6436703B1 (en) * 2000-03-31 2002-08-20 Hyseq, Inc. Nucleic acids and polypeptides
JP2004500870A (en) * 2000-06-20 2004-01-15 インサイト・ゲノミックス・インコーポレイテッド Secreted protein
JP2004506422A (en) * 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 Novel mitogen-activated kinase
US7297523B2 (en) * 2000-11-29 2007-11-20 Xenon Pharmaceuticals Inc. Human elongase genes and uses thereof
WO2002063006A2 (en) * 2001-02-06 2002-08-15 Incyte Genomics, Inc. Receptors and membrane-associated proteins
WO2002062974A2 (en) * 2001-02-08 2002-08-15 Bayer Aktiengesellschaft Regulation of human elongase hselo1-like protein
WO2002062975A2 (en) * 2001-02-08 2002-08-15 Bayer Aktiengesellschaft Regulation of human elongase hselo1-like protein
WO2002096943A1 (en) * 2001-05-25 2002-12-05 Asahi Kasei Kabushiki Kaisha Stat6-activating genes
AU2003231531A1 (en) * 2002-04-24 2003-11-10 Incyte Corporation Kinases and phosphatases

Also Published As

Publication number Publication date
EP1402051A4 (en) 2005-08-17
CA2448112A1 (en) 2002-12-12
WO2002098891A3 (en) 2003-05-30
WO2002098468A1 (en) 2002-12-12
JP2004532638A (en) 2004-10-28
WO2002099068A3 (en) 2003-12-04
WO2002098889A2 (en) 2002-12-12
JP2005514906A (en) 2005-05-26
EP1402262A2 (en) 2004-03-31
EP1402262A4 (en) 2005-05-11
AU2002314887A1 (en) 2002-12-16
WO2002098889A3 (en) 2003-03-27
EP1402051A2 (en) 2004-03-31
WO2002098891A2 (en) 2002-12-12
WO2002099068A2 (en) 2002-12-12

Similar Documents

Publication Publication Date Title
US20060160764A1 (en) LIMKs as modifiers of p53 pathway and methods of use
US20050170344A1 (en) Chds as modifiers of the p53 pathway and methods of use
CA2449479A1 (en) Map3ks as modifiers of the p53 pathway and methods of use
US8268548B2 (en) MAP3Ks as modifiers of the p53 pathway and methods of use
AU2002314883A1 (en) MAP3Ks as modifier of the p53 pathway and methods of use
AU2002310283A1 (en) PIBs as modifiers of the p53 pathway and methods of use
AU2002312253A1 (en) LIMKs as modifiers of the p53 pathway and methods of use
AU2002320051A1 (en) CHDs as modifiers of the p53 pathway and methods of use
AU2002312271A1 (en) MARKs as modifiers of the p53 pathway and methods of use
AU2002305777A1 (en) CADs as modifiers of the p53 pathway and methods of use
AU2002312284A1 (en) DGKS as modifiers of the p53 pathway and methods of use
AU2002310273A1 (en) SLC2As as modifiers of the P53 pathway and methods of use

Legal Events

Date Code Title Description
FZDE Dead