WO2003101283A2 - Diagnostics markers for lung cancer - Google Patents

Diagnostics markers for lung cancer Download PDF

Info

Publication number
WO2003101283A2
WO2003101283A2 PCT/US2003/017409 US0317409W WO03101283A2 WO 2003101283 A2 WO2003101283 A2 WO 2003101283A2 US 0317409 W US0317409 W US 0317409W WO 03101283 A2 WO03101283 A2 WO 03101283A2
Authority
WO
WIPO (PCT)
Prior art keywords
protein
antibody
cdna
cdnas
proteins
Prior art date
Application number
PCT/US2003/017409
Other languages
French (fr)
Other versions
WO2003101283A3 (en
Inventor
Paula K. Rickert
Randi Krasnow
Original Assignee
Incyte Corporation
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 Incyte Corporation filed Critical Incyte Corporation
Priority to AU2003240495A priority Critical patent/AU2003240495A1/en
Publication of WO2003101283A2 publication Critical patent/WO2003101283A2/en
Publication of WO2003101283A3 publication Critical patent/WO2003101283A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Definitions

  • the present invention relates to a combination comprising a plurality of cDNAs or a plurality of proteins which are differentially expressed in lung cancer and which may be used to diagnose, to stage, to treat, or to monitor the progression or treatment of respiratory disorders and in particular, lung cancer.
  • Bronchogenic carcinoma is a leading cause of cancer death in the United States affecting men (32%) and women (25%) each year. Cigarette smoking is the largest contributory factor in the diagnosis of males (90%) and females (85%) while occupational hazards, such as asbestos, arsenic, nickel and chloromethyl esters account for another small proportion of these cancers (15% in men and 5% in women). Damage to the DNA, activation of cellular oncogenes, and stimulation by growth factors are of primary importance in disease development. In nearly 80% of patients diagnosed with lung cancer, metastasis to the pleura, brain, bone, pericardium, and liver has already occurred. The decision to treat with surgery, radiation therapy, or chemotherapy is made on the basis of tumor histology, response to hormones or growth factors, and sensitivity to inhibitors or drugs. With current treatments, most patients die within one year of diagnosis.
  • the three most common types of bronchogenic carcinoma are squamous cell, small cell, and adenocarcinoma. These cancers progress through a series of morphologically distinct stages from hyperplasia to invasive carcinoma.
  • Malignant lung cancers are divided into small cell and non-small cell groups.
  • the non small cell carcinoma group accounts for about 70% of all lung cancer cases and includes adenocarcinomas, squamous cell carcinomas, and large cell carcinomas.
  • Adenocarcinomas typically arise in the peripheral airways and often form mucin secreting glands.
  • Squamous cell carcinomas typically arise in proximal airways in apparent response to chronic inflammation and injury of the bronchial epithelium.
  • the small cell carcinomas account for about 20% of lung cancers, typically arise in proximal airways, and inappropriately produce adrenocorticotropin and anti-diuretic hormone.
  • Lung cancer cells accumulate numerous genetic lesions, many of which are associated with cytologically visible chromosomal aberrations.
  • the high frequency of chromosomal deletions associated with lung cancer may reflect the roles of multiple tumor suppressor loci in the etiology of this disease.
  • Deletion of the short arm of chromosome 3 is found in over 90% of cases and represents one of the earliest genetic lesions leading to lung cancer. Deletions at chromosome arms 9p and 17p are also common.
  • telomeres activation of oncogenes such as K-ras and c-myc
  • tumor suppressor genes such as retinoblastoma, p53 and cyclin-dependent kinase inhibitors such as CDKN2.
  • thrombospondin-1, f ⁇ bronectin, intercellular adhesion molecule 1, and cytokeratins 6 and 18 had been observed previously to be differentially expressed in lung cancers.
  • Wang et al. 2000, Oncogene 19:1519-1528
  • Keratin isoform 6, connexin 26, plakofillin 1 and cytokeratin 13 were identified among the known genes.
  • Array technologies provide a simple way to explore the expression profile of a large number of related or unrelated genes.
  • arrays provide a platform for examining which genes are tissue-specific, carrying out housekeeping functions, parts of a signaling cascade, or specifically related to a particular genetic predisposition, condition, disease, or disorder.
  • the potential application of gene expression profiling is particularly relevant to improving diagnosis, prognosis, and treatment of disease.
  • the present invention satisfies a need in the art by providing a plurality of cDNAs, proteins or antibodies that can be used to diagnose, to stage, to treat, or to monitor the progression or treatment of a subject with a respiratory disorder and in particular, lung cancer.
  • the present invention provides a combination comprising a plurality of cDNAs wherein the cDNAs are SEQ ID NOs: 1-170 as presented in the Sequence Listing that are differentially expressed in lung cancers and the complements of SEQ ID NOs: 1-170.
  • the invention also provides a combination comprising a plurality of cDNAs wherein the cDNAs are SEQ ID NOs: 1-37 that are differentially expressed at least 16-fold in lung cancers and the complements of SEQ ID NOs: 1-37.
  • the invention further provides a combination comprising a plurality of cDNAs wherein the cDNAs are SEQ ID NOs:38-170 that are differentially expressed at least 4-fold in lung cancers and the complements of SEQ ID NOs: 38- 170.
  • the combination is useful to diagnose a respiratory disorder such as lung cancer.
  • the combination is immobilized on a substrate.
  • the invention also provides a high throughput method to detect differential expression of one or more of the cDNAs of the combination.
  • the method comprises hybridizing the substrate comprising the combination with the nucleic acids of a sample, thereby forming one or more hybridization complexes, detecting the hybridization complexes, and comparing the hybridization complexes with those of a standard, wherein differences in the size and signal intensity of each 5 hybridization complex indicates differential expression of nucleic acids in the sample.
  • the nucleic acids of the sample are amplified prior to hybridization, hi another aspect, the sample is from a subject with lung cancer and differential expression identifies the type of lung cancer or stages the lung cancer.
  • the invention provides a high throughput method of screening a library or a plurality of
  • the method comprises combining the substrate comprising the combination with a library or a plurality of molecules or compounds under conditions to allow specific binding and detecting specific binding, thereby identifying a ligand.
  • the library or plurality of molecules or compounds are selected from DNA molecules, enhancers, mimetics, peptide nucleic acids, proteins, repressors, regulatory proteins, RNA molecules, and transcription factors.
  • the invention also provides a method for purifying a ligand, the method comprising combining a cDNA of the invention with a sample under conditions which allow specific binding, recovering the bound cDNA, and separating the cDNA from the ligand, thereby obtaining purified ligand.
  • the invention provides an isolated cDNA comprising a nucleic acid selected from SEQ ID NOs:2, 3, 32, 34, 35, 39, 43, 44, 45, 80, 81, 82, 139, 140, 146, 147, 148, 149, 151, 152, 154, 155, 157,
  • the invention also provides a vector comprising the cDNA, a host cell comprising the vector, and a method for producing a protein comprising culturing the host cell under conditions for the expression of a protein and recovering the protein from the host cell culture.
  • the present invention also provides a combination of purified proteins comprising the amino acids
  • the invention also provides a purified protein comprising the amino acid sequence of SEQ ID NO: 174 encoded by a cDNA of the invention.
  • the invention also provides a high-throughput method for using a protein to screen a library or a plurality of molecules or compounds to identify a ligand. The method comprises
  • the library or plurality of molecules or compounds is selected from agonists, antagonists, antibodies, DNA molecules, small molecule drugs, immunoglobulins, inhibitors, mimetics, peptide nucleic acids, peptides, pharmaceutical agents, proteins, RNA
  • an antibody which specifically binds a protein of the invention is selected from a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a recombinant antibody, a humanized antibody, single chain antibodies, a Fab fragment, an F(ab'> 2 fragment, an Fv fragment; and an antibody-peptide fusion protein.
  • the invention further provides a method for using a protein to purify a ligand. The method comprises combining the protein or a portion thereof with a sample under conditions to allow specific binding, recovering the bound protein, and separating the protein from the ligand, thereby obtaining purified ligand.
  • the invention still further provides method for using the protein to produce a polyclonal or a monoclonal antibody.
  • the method for producing a polyclonal antibody comprises immunizing a animal with a protein under conditions to elicit an antibody response, isolating animal antibodies, attaching the protein to a substrate, contacting the substrate with isolated antibodies under conditions to allow specific binding to the protein, dissociating the antibodies from the protein, thereby obtaining purified polyclonal antibodies.
  • the method for preparing a monoclonal antibody comprises immunizing a animal with a protein under conditions to elicit an antibody response, isolating antibody producing cells from the animal, fusing the antibody producing cells with immortalized cells in culture to form monoclonal antibody producing hybridoma cells, culturing the hybridoma cells, and isolating a monoclonal antibody from culture.
  • the method comprises immunizing an animal with the protein or an antigenic determinant thereof under conditions to elicit an antibody response, isolating animal antibodies, and screening the isolated antibodies with the protein to identify an antibody which specifically binds the protein.
  • the invention yet still further provides a method for using the protein to purify antibodies which bind specifically to the protein.
  • the invention provides a purified antibody which specifically binds a protein of the invention.
  • the invention also provides a method of using an antibody to detect the expression of a protein in a sample, the method comprising contacting the antibody with a sample under conditions for the formation of an antibody:protein complex and detecting complex formation wherein the formation of the complex indicates the expression of the protein in the sample.
  • complex formation is compared to standards and is diagnostic of a respiratory disorder such as lung cancer.
  • the invention further provides using an antibody to immunopurify a protein comprising combining the antibody with a sample under conditions to allow formation of an antibody:protein complex, and separating the antibody from the protein, thereby obtaining purified protein.
  • the invention provides a composition comprising a cDNA, a protein, an antibody, or a ligand which has agonistic or antagonistic activity.
  • Sequence Listing provides SEQ ID NOs: 1-170, exemplary polynucleotides of the invention.
  • the Sequence Listing also provides SEQ ID NOs: 171-296, exemplary proteins of the invention. Each sequence is identified by a sequence identification number (SEQ ID NO) and by INCYTE ID.
  • Figures 1 A-4A show the amino acid sequence of SEQ ID NO: 174 and a nucleic acid sequence of its encoding cDNA (SEQ ID NO:43). The alignment was produced using MACDNASIS PRO software (Hitachi Software Engineering, South San Francisco CA).
  • Table 1 lists the cDNAs whose transcripts are differentially up-regulated by at least 16-fold in lung tumors as determined by microarray analysis.
  • the column descriptions for Tables 1-4 are: Column 1 shows SEQ ID NO: (SEQ ID ); column 2, Incyte ID; column 3, the number of samples showing greater than 16-fold (Tables 1-2) or 4-fold (Tables 3-4) expression levels; column 4, the closest GenBank homolog; column 5, the E-value for the comparison with the GenBank homolog; and column 6, the description of the GenBank homolog.
  • Table 2 lists the cDNAs whose transcripts are differentially down-regulated by at least 16- fold in lung tumors as determined by microarray analysis.
  • Table 3 lists-the cDNAs whose transcripts are differentially up-regulated by at least 4-fold and less than 16-fold in lung tumors as determined by microarray analysis.
  • Table 4 lists the cDNAs whose transcripts are differentially down-regulated by at least 4-fold and less than 16-fold in lung tumors as determined by microarray analysis.
  • Table 5 lists the proteins comprising the amino acid sequences of SEQ ID NOs: 171-296 as shown in the Sequence Listing by SEQ ID NO (columns 1, 3, and 5) and Incyte ID (columns 2, 4, and
  • Antibody refers to intact immunoglobulin molecule, a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a recombinant antibody, a humanized antibody, single chain antibodies, a Fab fragment, an F(ab') 2 fragment, an Fv fragment; and an antibody-peptide fusion protein.
  • Antigenic determinant refers to an antigenic or immunogenic epitope, structural feature, or region of an oligopeptide, peptide, or protein which is capable of inducing formation of an antibody which specifically binds the protein. Biological activity is not a prerequisite for immunogenicity.
  • Array refers to an ordered arrangement of at least two cDNAs, proteins, or antibodies on a substrate. At least one of the cDNAs, proteins, or antibodies represents a control or standard, and the other cDNA, protein, or antibody is of diagnostic or therapeutic interest. The arrangement of at least two and up to about 40,000 cDNAs, proteins, or antibodies on the substrate assures that the size and signal intensity of each labeled complex, formed between each cDNA and at least one nucleic acid, each protein and at least one ligand or antibody, or each antibody and at least one protein to which the antibody specifically binds, is individually distinguishable.
  • a “combination” refers to at least two cDNAs, and up to 170 cDNAs selected from SEQ ID
  • the "complement" of a cDNA of the Sequence Listing refers to a nucleic acid molecule which is completely complementary over its full length and which will hybridize to a nucleic acid molecule under conditions of high stringency.
  • cDNA refers to an isolated polynucleotide, nucleic acid, or a fragment thereof, that contains from about 400 to about 12,000 nucleotides. It may have originated recombinantly or synthetically, may be double-stranded or single-stranded, represents coding and noncoding 3 ' or 5' sequence, generally lacks introns and may be purified or combined with carbohydrate, lipids, protein or inorganic elements or substances.
  • cDNA encoding a protein refers to a nucleic acid whose sequence closely aligns with sequences that encode conserved regions, motifs or domains identified by employing analyses well known in the art. These analyses include BLAST (Altschul (1993) J Mol Evol 36:290-300; Altschul et al. (1990) J Mol Biol 215:403-410; and BLAST2 (Altschul (1997) supra) which provide identity within the conserved region. Brenner et al.
  • composition refers to the polynucleotide and a vector or a labeling moiety; a purified protein and a pharmaceutical carrier or a heterologous, labeling, or purification moiety; an antibody and a labeling moiety or pharmaceutical agent; and the like.
  • “Differential expression” refers to an increased, upregulated or present, or decreased, downregulated or absent, gene expression as detected by the absence, presence, or at least two-fold change in the amount of transcribed messenger RNA or translated protein in a sample.
  • “Disorder” refers to conditions, diseases or syndromes of the respiratory system including, but not limited to, lung cancer, particularly adenocarcinomas and squamous cell carcinomas; chronic obstructive pulmonary disease, emphysema, or asthma.
  • An "expression profile” is a representation of gene expression in a sample.
  • a nucleic acid expression profile is produced using sequencing, hybridization, or amplification technologies and resort , _ _ __ ⁇
  • a protein expression profile although time delayed, mirrors the nucleic acid expression profile and uses two-dimensional polyacrylamide electrophoresis (2D-PAGE, mass spectrophotometry (MS), enzyme-linked immunosorbent assays (ELISAs), radioimmunoassays (RIAs), and fluorescence activated cell sorting (FACS) or arrays and labeling moieties or antibodies to detect expression in a sample.
  • the nucleic acids, proteins, or antibodies may be used in solution or attached to a substrate, and their detection is based on methods and labeling moieties well known in the art.
  • a "hybridization complex” is formed between a polynucleotide of the invention and a nucleic acid of a sample when the purines of one molecule hydrogen bond with the pyrimidines of the complementary molecule, e.g., 5'-A-G-T-C-3'base pairs with its complete complement, 3 -T-C-A-G- 5'.
  • the degree of complementarity and the use of nucleotide analogs affect the efficiency and stringency of hybridization reactions.
  • Identity refers to the quantification (usually percentage) of nucleotide or residue matches between at least two sequences aligned using a standardized algorithm such as Smith-Waterman alignment (Smith and Waterman (1981) J Mol Biol 147:195-197), CLUSTALW (Thompson et al- (1994) Nucleic Acids Res 22:4673-4680), or BLAST2 (Altschul (1997) supra).
  • BLAST2 may be used in a standardized and reproducible way to insert gaps in one of the sequences in order to optimize alignment and to achieve a more meaningful comparison between them.
  • Similarity as applied to proteins uses the same algorithms but takes into account conservative substitutions of nucleotides or residues.
  • isolated or “purified” refers to any molecule or compound that is separated from its natural environment and is from about 60% free to about 90% free from other components with which it is naturally associated.
  • Labeling moiety refers to any reporter molecule whether a visible or radioactive label, stain or dye that can be attached to or incorporated into a cDNA or protein.
  • Visible labels and dyes include but are not limited to anthocyanins, ⁇ glucuronidase, BIODIPY, Coomassie blue, Cy3 and Cy5, digoxigenin, FITC, green fluorescent protein, luciferase, spyro red, silver, and the like.
  • Radioactive markers include radioactive forms of hydrogen, iodine, phosphorous, sulfur, and the like.
  • Ligand refers to any agent, molecule, or compound which will bind specifically to a complementary site on a cDNA molecule or polynucleotide, or to an epitope or a protein. Such ligands stabilize or modulate the activity of polynucleotides or proteins and may be composed of inorganic or organic substances including nucleic acids, proteins, carbohydrates, fats, and lipids.
  • Oligomer refers a single stranded molecule from about 18 to about 60 nucleotides in length which may be used in hybridization or amplification technologies or in regulation of replication, transcription or translation. Equivalent terms are amplimer, primer, and oligomer.
  • Portion refers to any part of a protein used for any purpose which retains at least one biological or antigenic characteristic of a native protein, but especially, to an epitope for the screening of ligands or for the production of antibodies.
  • Post-translational modification of a protein can involve lipidation, glycosylation, phosphorylation, acetylation, racemization, proteolytic cleavage, and the like. These processes may occur synthetically or biochemically. Biochemical modifications will vary by cellular location, cell type, pH, enzymatic milieu, and the like.
  • Probe refers to a cDNA that hybridizes to at least one nucleic acid molecule in a sample. Probes can be labeled for use in hybridization reactions including Southern, northern, in situ, dot blot, array, and like technologies or in screening assays.
  • Protein refers to a polypeptide or peptide comprising an amino acid sequence selected from SEQ ID NOs: 171-296.
  • An "oligopeptide” is an amino acid sequence from about five residues to about 15 residues that is used as part of a fusion protein to produce an antibody.
  • sample is used in its broadest sense as containing nucleic acids, proteins, antibodies, and the like.
  • a sample may comprise a bodily fluid such as ascites, blood, lymph, semen, spinal fluid, sputum, tears, urine, and the like; the soluble fraction of a cell preparation, or an aliquot of media in which cells were grown; a chromosome, an organelle, or membrane isolated or extracted from a cell; genomic DNA, RNA, or cDNA in solution or bound to a substrate; a cell; a tissue, tissue biopsy or tissue print; buccal cells, skin, a hair or its follicle; and the like.
  • Specific binding refers to a special and precise interaction between two molecules which is dependent upon their structure, particularly their molecular side groups. Examples include the intercalation of a regulatory protein into the major groove of a DNA molecule, the hydrogen bonding along the backbone between two single stranded nucleic acids, or the binding between an epitope of a protein and an agonist, antagonist, or antibody.
  • Substrate refers to any rigid or semi-rigid support to which cDNAs or proteins are bound and includes membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, capillaries or other tubing, plates, polymers, and microparticles with a variety of surface forms including wells, trenches, pins, channels and pores.
  • a "transcript image” is a profile of gene transcription activity in a particular tissue at a particular time. TI provides assessment of the relative abundance of expressed polynucleotides in the cDNA libraries of an EST database as described in USPN 5,840,484, incorporated herein by reference.
  • “Variant” refers to molecules that are recognized variations of a cDNA or a protein encoded by the cDNA. Splice variants may be determined by BLAST score, wherein the score is at least 100, and most preferably at least 400. Allelic variants have high percent identity to the cDNAs of the invention and may differ by about three bases per hundred bases. "Single nucleotide polymorphism” (SNP) refers to a change in a single base as a result of a substitution, insertion or deletion. The change may be conservative (purine for purine) or non-conservative (purine to pyrimidine) and may or may not result in a change in an encoded amino acid.
  • SNP single nucleotide polymorphism
  • the present invention provides a combination comprising a plurality of cDNAs, wherein the cDNAs are SEQ ID NOs: 1-170 of the Sequence Listing and the complete complements of SEQ ID NOs: 1-170, that are differentially expressed in lung cancers.
  • the invention also provides a combination comprising a plurality of proteins, wherein the proteins are SEQ ID NOs: 171-296 of the Sequence Listing, that are differentially expressed in lung cancers.
  • compositions when immobilized on an array can be used in a clinically relevant manner with subject tissues to detect changes in expression associated with respiratory disorders, particularly adenocarcinomas, neuroendocrine carcinoids, and squamous cell carcinomas of the lung, chronic obstructive pulmonary disease, emphysema, and asthma.
  • the invention further provides a subcombinations comprising a plurality of cDNAs, wherein the cDNAs are SEQ ID NOs: 1-18 and the complete complements of SEQ ID NOs: 1-18 that are up- regulated by at least 16-fold in lung cancers.
  • the invention still further provides a subcombination comprising a plurality of cDNAs, wherein the cDNAs are SEQ ID NOs: 19-37 and the complete complements of SEQ ID NOs: 19-37 that are down-regulated by at least 16-fold in lung cancers.
  • the invention further provides a subcombination comprising a plurality of cDNAs, wherein the cDNAs are SEQ ID NOs:38-83 and the complete complements of SEQ ID NOs:38-83 that are up-regulated between 4- and 16-fold in lung cancers.
  • the invention still further provides a subcombination comprising a plurality of cDNAs, wherein the cDNAs are SEQ ID NOs: 84-170 and the complete complements of SEQ ID NOs:84-170 that are down-regulated between 4- and 16-fold in lung cancers.
  • the invention yet further provides cDNAs comprising the nucleic acid sequence of SEQ ID NOs: 84-170 and the complete complements of SEQ ID NOs:84-170 that are down-regulated between 4- and 16-fold in lung cancers.
  • the invention yet further provides cDNAs comprising the nucleic acid sequence of SEQ ID NOs: 84-170 and the complete complements of SEQ ID NOs:84-170 that are down-regulated between 4- and 16-fold in lung cancers.
  • the invention yet further provides cDNAs comprising the nucleic acid sequence of SEQ ID NOs: 84-170 and the complete complements of SEQ ID NOs:84-170 that are down-regulated between 4- and 16-fold in lung cancers
  • the invention still yet further provides a purified protein comprising the amino acid sequence of SEQ ID NO: 174.
  • the novel protein is 285 amino acids in length and identical, from residue M52 to residue N285, to human thymidine kinase (g339719) as determined using BLAST.
  • SEQ ID NO: 174 has one potential cAMP- protein kinase phosphorylation site at residue K242; three potential casein kinase II phosphorylation sites at residues S27, T37, and T219; six potential protein kinase C phosphorylation sites at residues S2, S27, S41, S81, T214, and T219; two potential tyrosine kinase phosphorylation site at residues Y34 and Yl 12; two potential ATP/GTP-binding site motif A (P-loop) at residues S84 and T169; and one thymidine kinase cellular-type signature from residues G227 to Y240.
  • SEQ ID NO: 174 also contains a thymidine kinase domain from residues G70 to Y240 as determined by searching for statistically significant matches in the hidden Markov model (HMM)- based PFAM database of conserved protein family domains. Data from BLIMPS, PROFILESCAN, and MOTIFS provide further evidence that SEQ ID NO: 174 is a member of the thymidine kinase family.
  • HMM hidden Markov model
  • Figures 1A-4A show the amino acid sequence of SEQ ID NO: 174 and a nucleic acid sequence of its encoding cDNA (SEQ ID NO:43). The alignment was produced using MACDNASIS PRO software (Hitachi Software Engineering, South San Francisco CA).
  • Table 1 shows those cDNAs of the present invention having the nucleic acid sequences of SEQ ID NOs: 1-18 that showed at least 16-fold up-regulation in lung tumors.
  • Columns 1 and 2 show the SEQ ID NO and Incyte ID, respectively.
  • Column 3 shows the number of tumors in which transcripts are up-regulated by at least 16-fold.
  • Columns 4 and 6 show the GenBank identification number and description of the nearest GenBank homolog identified using BLAST2 analysis with default parameters.
  • Column 5 shows the probability scores for the matches between each cDNA and its homolog.
  • Table 2 shows those cDNAs of the present invention having the nucleic acid sequences of SEQ ID NOs: 19-37 that showed at least 16-fold down-regulation in lung tumors.
  • Columns 1 and 2 show the SEQ ID NO and Incyte ID, respectively.
  • Column 3 shows the number of tumors in which transcripts are down-regulated by at least 16-fold.
  • Columns 4 and 6 show the GenBank identification number and description of the nearest GenBank homolog identified using BLAST2 analysis with default parameters.
  • Column 5 shows the probability scores for the matches between each cDNA and its homolog.
  • Table 3 shows those cDNAs of the present invention having the nucleic acid sequences of
  • SEQ ID NOs:38-83 that showed between 4- to 16-fold up-regulation in lung tumors.
  • Columns 1 and 2 show the SEQ ID NO and Incyte ID, respectively.
  • Column 3 shows the number of tumors in which transcripts are up-regulated between 4- and 16-fold.
  • Columns 4 and 6 show the GenBank identification number and description of the nearest GenBank homolog identified using BLAST2 analysis with default parameters.
  • Column 5 shows the probability scores for the matches between each cDNA and its homolog.
  • Table 4 shows those cDNAs of the present invention having the nucleic acid sequences of SEQ ID NOs:84-170 that showed between 4- to 16-fold down-regulation in lung tumors.
  • Table 4 lists the differential expression of clones representing cDNAs that are down-regulated by at least 4-fold and less than 16-fold on microarrays. Columns 1 and 2 show the SEQ ID NO and Incyte ID, respectively. Column 3 shows the number of tumors in which transcripts are down-regulated between 4- and 16-fold. Columns 4 and 6 show the GenBank identification number and description of the nearest GenBank homolog identified using BLAST2 analysis with default parameters. Column 5 shows the probability scores for the matches between each cDNA and its homolog. Table 5 lists the proteins comprising the amino acid sequences of SEQ ID NOs: 171-296 as shown in the Sequence Listing by SEQ ID NO (columns 1, 3, and 5) and Incyte ID (columns 2, 4, and 6).
  • the cDNAs of the invention define a differential expression pattern against which to compare the expression profile of biopsied, in vitro or in vivo treated lung samples.
  • the proteins may be arranged on an array and used to screen for therapeutics.
  • the combination of nucleotides or antibodies which specifically bind the proteins may be arrayed and contacted with samples from subjects with undiagnosed respiratory disorders. Expression profiles will provide information useful for identifying and distinguishing among different disorders, or stages of those disorders, identifying those sequences of highest diagnostic and potential therapeutic value for each disorder, or evaluating patient treatment.
  • Such nucleotide, protein and antibody combinations may be useful in the elucidation of pathways which are affected in a particular disorder or to identify new, coexpressed, candidate, therapeutic molecules.
  • Samples for these assays are prepared by methods well known in the art and are derived from mammalian cells or tissues which are in a certain stage of development; have been treated with a known molecule or compound, such as a cytokine, growth factor, drug, or the like; or have been extracted or biopsied from a mammal with a known or unknown condition, disorder, or disease before or after treatment.
  • the sample nucleic acids or protein are contacted with the cDNA or antibody array for the purpose of defining a novel gene profile associated with that developmental stage, treatment, or disorder.
  • cDNAs and Their Uses cDNAs can be prepared by a variety of synthetic or enzymatic methods well known in the art.
  • cDNAs can be synthesized, in whole or in part, using chemical methods well known in the art (Caruthers et al. (1980) Nucleic Acids Symp Ser (7) 215-233). Alternatively, cDNAs can be produced enzymatically or recombinantly, by in vitro or m vivo transcription. Nucleotide analogs can be incorporated into cDNAs by methods well known in the art. The only requirement is that the incorporated analog must base pair with native purines or pyrimidines. For example, 2, 6-diaminopurine can substitute for adenine and form stronger bonds with thymidine than those between adenine and thymidine.
  • cDNAs can include nucleotides that have been derivatized chemically or enzymatically.
  • cDNAs can be synthesized on a substrate. Synthesis on the surface of a substrate may be accomplished using a chemical coupling procedure and a piezoelectric printing apparatus as described by Baldeschweiler et al- (PCT publication WO95/251116). Alternatively, the cDNAs can be synthesized on a substrate surface using a self-addressable electronic device that controls when reagents are added as described by Heller et al. (USPN 5,605,662).
  • cDNAs can be synthesized directly on a substrate by sequentially dispensing reagents for their synthesis on the substrate surface or by dispensing preformed DNA fragments to the substrate surface.
  • Typical dispensers include a micropipette delivering solution to the substrate with a robotic system to control the position of the micropipette with respect to the substrate. There can be a multiplicity of dispensers so that reagents can be delivered to the reaction regions efficiently.
  • cDNAs can be immobilized on a substrate by covalent means such as by chemical bonding procedures or UV irradiation. In one method, a cDNA is bound to a glass surface which has been modified to contain epoxide or aldehyde groups.
  • a cDNA is placed on a polylysine coated surface and UV cross-linked to it as described by Shalon et al. (WO95/35505).
  • a cDNA is actively transported from a solution to a given position on a substrate by electrical means (Heller, supra).
  • cDNAs do not have to be directly bound to the substrate, but rather can be bound to the substrate through a linker group.
  • the linker groups are typically about 6 to 50 atoms long to provide exposure of the attached cDNA.
  • Preferred linker groups include ethylene glycol oligomers, diamines, diacids and the like. Reactive groups on the substrate surface react with a terminal group of the linker to bind the linker to the substrate.
  • polynucleotides, plasmids or cells can be arranged on a filter. In the latter case, cells are lysed, proteins and cellular components degraded, and the DNA is coupled to the filter by UV cross-linking.
  • the cDNAs may be used for a variety of purposes.
  • the combination of the invention may be used on an array.
  • the array in turn, can be used in high-throughput methods for detecting a related polynucleotide in a sample, screening a plurality of molecules or compounds to identify a ligand, diagnosing a respiratory disorder such as chronic obstructive pulmonary disease or lung cancer, or inhibiting or inactivating a therapeutically relevant gene related to the cDNA.
  • the cDNAs of the invention are employed on a microarray, the cDNAs are arranged in an ordered fashion so that each cDNA is present at a specified location. Because the cDNAs are at specified locations on the substrate, the hybridization patterns and intensities, which together create a unique expression profile, can be interpreted in terms of expression levels of particular genes and can be correlated with a particular metabolic process, condition, disorder, disease, stage of disease, or treatment. Hybridization The cDNAs or fragments or complements thereof may be used in various hybridization technologies. The cDNAs may be labeled using a variety of reporter molecules by either PCR , recombinant, or enzymatic techniques.
  • a commercially available vector containing the cDNA is transcribed in the presence of an appropriate polymerase, such as T7 or SP6 polymerase, and at least one labeled nucleotide.
  • an appropriate polymerase such as T7 or SP6 polymerase
  • kits are available for labeling and cleanup of such cDNAs. Radioactive (Amersham Biosciences (APB), Piscataway NJ), fluorescent (Qiagen-Operon, Alameda CA), and chemiluminescent labeling (Promega, Madison WI) are well known in the art.
  • a cDNA may represent the complete coding region of an mRNA or be designed or derived from unique regions of the mRNA or genomic molecule, an intron, a 3' untranslated region, or from a conserved motif.
  • the cDNA is at least 18 contiguous nucleotides in length and is usually single stranded.
  • Such a cDNA may be used under hybridization conditions that allow binding only to an identical sequence, a naturally occurring molecule encoding the same protein, or an allelic variant. Discovery of related human and mammalian sequences may also be accomplished using a pool of degenerate cDNAs and appropriate hybridization conditions.
  • a cDNA for use in Southern or northern hybridizations may be from about 400 to about 6000 nucleotides long.
  • Such cDNAs have high binding specificity in solution-based or substrate-based hybridizations.
  • An oligonucleotide, a fragment of the cDNA may be used to detect a polynucleotide in
  • the stringency of hybridization is determined by G+C content of the cDNA, salt concentration, and temperature. In particular, stringency is increased by reducing the concentration of salt or raising the hybridization temperature. In solutions used for some membrane based hybridizations, addition of an organic solvent such as formamide allows the reaction to occur at a lower temperature.
  • Hybridization may be performed with buffers, such as 5x saline sodium citrate (SSC) with 1% sodium dodecyl sulfate (SDS) at 60°C, that permit the formation of a hybridization complex between nucleic acid sequences that contain some mismatches.
  • buffers such as 5x saline sodium citrate (SSC) with 1% sodium dodecyl sulfate (SDS) at 60°C, that permit the formation of a hybridization complex between nucleic acid sequences that contain some mismatches.
  • washes are performed with buffers such as 0.2xSSC with 0.1% SDS at either 45°C (medium stringency) or 65°C- 68°C (high stringency).
  • buffers such as 0.2xSSC with 0.1% SDS at either 45°C (medium stringency) or 65°C- 68°C (high stringency).
  • formamide may be added to the hybridization solution to reduce the temperature at which hybridization is performed. Background signals may be reduced by the use of detergents such as Sarkosyl or TRITON X-100 (Sigma-Aldrich, St. Louis MO) and a blocking agent such as denatured salmon sperm DNA.
  • the array may be used to monitor the expression level of large numbers of genes simultaneously and to identify genetic variants, mutations, and SNPs. Such information may be used to determine gene function; to 5 understand the genetic basis of a disorder; to diagnose a disorder; and to develop and monitor the activities of therapeutic agents being used to control or cure a disorder. (See, e.g., USPN 5,474,796; WO95/11995; WO95/35505; USPN 5,605,662; and USPN 5,958,342.) Screening and Purification Assays
  • a cDNA may be used to screen a library or a plurality of molecules or compounds for a
  • Ligands may be DNA molecules, RNA molecules, peptide nucleic acid molecules, peptides, proteins such as transcription factors, promoters, enhancers, repressors, and other proteins that regulate replication, transcription, or translation of the polynucleotide in the biological system.
  • the assay involves combining the cDNA or a fragment thereof with the molecules or compounds under conditions that allow specific binding and detecting 5 the bound cDNA to identify at least one ligand that specifically binds the cDNA.
  • the cDNA may be incubated with a library of isolated and purified molecules or compounds and binding activity determined by methods such as a gel-retardation assay (USPN 6,010,849) or a reticulocyte lysate transcriptional assay.
  • the cDNA may be incubated with nuclear extracts from biopsied and/or cultured cells and tissues. Specific 0 binding between the cDNA and a molecule or compound in the nuclear extract is initially determined by gel shift assay. Protein binding may be confirmed by raising antibodies against the protein and adding the antibodies to the gel-retardation assay where specific binding will cause a supershift in the assay.
  • the cDNA may be used to purify a molecule or compound using 5 affinity chromatography methods well known in the art.
  • the cDNA is chemically reacted with cyanogen bromide groups on a polymeric resin or gel. Then a sample is passed over and reacts with or binds to the cDNA. The molecule or compound which is bound to the cDNA may be released from the cDNA by increasing the salt concentration of the flow-through medium and collected. 0
  • the cDNA may be used to purify a ligand from a sample.
  • a method for using a cDNA to purify a ligand would involve combining the cDNA or a fragment thereof with a sample under conditions to allow specific binding, recovering the bound cDNA, and using an appropriate agent to separate the cDNA from the purified ligand.
  • Protein Production and Uses 5 The cDNAs or fragment thereof may be used to produce purified proteins using recombinant DNA technologies described herein and taught in Ausubel (supra; Units 16.1-16.62).
  • One of the advantages of producing proteins by these procedures is the ability to obtain highly-enriched sources of the proteins thereby simplifying purification procedures.
  • the proteins may contain amino acid substitutions, deletions or insertions made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. Such substitutions may be conservative in nature when the substituted residue has structural or chemical properties similar to the original residue (e.g., replacement of leucine with isoleucine or valine) or they may be nonconservative when the replacement residue is radically different (e.g., a glycine replaced by a tryptophan).
  • Such vectors usually contain a promoter and a polylinker useful for cloning, priming, and transcription.
  • An exemplary vector may also contain the promoter for ⁇ -galactosidase, an amino-terminal methionine and the subsequent seven amino acid residues of ⁇ -galactosidase.
  • the vector may be transformed into competent E. coli cells. Induction of the isolated bacterial strain with isopropylthiogalactoside (IPTG) using standard methods will produce a fusion protein that contains an N terminal methionine, the first seven residues of ⁇ -galactosidase, about 15 residues of linker, and the protein encoded by the cDNA.
  • IPTG isopropylthiogalactoside
  • the cDNA may be shuttled into other vectors known to be useful for expression of protein in specific hosts. Oligonucleotides containing cloning sites and fragments of DNA sufficient to hybridize to stretches at both ends of the cDNA may be chemically synthesized by standard methods. These primers may then be used to amplify the desired fragments by PCR. The fragments may be digested with appropriate restriction enzymes under standard conditions and isolated using gel electrophoresis. Alternatively, similar fragments are produced by digestion of the cDNA with appropriate restriction enzymes and filled in with chemically synthesized oligonucleotides. Fragments of the coding sequence from more than one gene may be ligated together and expressed.
  • a chimeric protein may be expressed that includes one or more additional purification-facilitating domains.
  • additional purification-facilitating domains include, but are not limited to, metal-chelating domains that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (I munex, Seattle WA).
  • the inclusion of a cleavable-linker sequence such as ENTEROKESfASEMAX (Invitrogen, San Diego CA) between the protein and the purification domain may also be used to recover the protein.
  • Suitable host cells may include, but are not limited to, mammalian cells such as Chinese Hamster Ovary (CHO) and human 293 cells, insect cells such as Sf9 cells, plant cells such as Nicotiana tabacum yeast cells such as Saccharomvces cerevisiae. and bacteria such as E. coli.
  • a useful vector may also include an origin of replication and one or two selectable markers to allow selection in bacteria as well as in a transformed eukaryotic host.
  • Vectors for use in eukaryotic host cells may require the addition of 3'poly(A) tail if the cDNA lacks poly(A).
  • the vector may contain promoters or enhancers that increase gene expression.
  • Many promoters are known and used in the art. Most promoters are host specific and exemplary promoters includes SV40 promoters for CHO cells; T7 promoters for bacterial hosts; viral promoters and enhancers for plant cells; and PGH promoters for yeast.
  • Adenoviral vectors with the rous sarcoma virus enhancer or retroviral vectors with long terminal repeat promoters may be used to drive protein expression in mammalian cell lines. Once homogeneous cultures of recombinant cells are obtained, large quantities of secreted soluble protein may be recovered from the conditioned medium and analyzed using chromatographic and electrophoretic methods well known in the art.
  • An alternative method for the production of large amounts of secreted protein involves the transformation of mammalian embryos and the recovery of the recombinant protein from milk produced by transgenic cows, goats, sheep, and the like.
  • proteins or portions thereof may be produced manually, using solid-phase techniques (Stewart et al. (1969) Solid-Phase Peptide Synthesis. WH Freeman, San Francisco CA; Merrifield (1963) J Am Chem Soc 5:2149-2154), or using machines such as the 431 A peptide synthesizer (Applied Biosystems (ABI), Foster City CA). Proteins produced by any of the above methods may be used as pharmaceutical compositions to treat disorders associated with null or inadequate expression of the genomic sequence. Protein Identification Several techniques have been developed which permit rapid identification of proteins from complex samples using high performance liquid chromatography and mass spectrometry (MS).
  • MS mass spectrometry
  • proteins are separated using two-dimensional gel electrophoresis (2-DE), 2) selected proteins are excised from the gel and digested with a protease to produce a set of peptides; and 3) the peptides are subjected to mass spectral analysis to derive peptide ion mass and spectral pattern information.
  • MS information is used to identify the protein by comparing it with information in a protein database (Shevenko et al.(1996) Proc Natl Acad Sci 93: 14440-14445).
  • Proteins are separated by 2DE employing isoelectric focusing (IEF) in the first dimension followed by SDS-PAGE in the second dimension.
  • IEF isoelectric focusing
  • an immobilized pH gradient strip is useful to increase reproducibility and resolution of the separation.
  • Alternative techniques may be used to improve resolution of very basic, hydrophobic, or high molecular weight proteins.
  • the separated proteins are detected using a stain or dye such as silver stain, Coomassie blue, or spyro red (Molecular Probes, Eugene OR) that is compatible with MS.
  • Gels may be blotted onto a PVDF membrane for western analysis and optically scanned using a STORM scanner (APB) to produce a computer-readable output which is analyzed by pattern recognition software such as MELANTE (GeneBio, Geneva, Switzerland).
  • the software annotates individual spots by assigning a unique identifier and calculating their respective x,y coordinates, molecular masses, isoelectric points, and signal intensity.
  • Individual spots of interest such as those representing differentially expressed proteins, are excised and proteolytically digested with a site-specific protease such as trypsin or chymotrypsin, singly or in combination, to generate a set of small peptides, preferably in the range of 1-2 kDa.
  • a site-specific protease such as trypsin or chymotrypsin, singly or in combination
  • samples Prior to digestion, samples may be treated with reducing and alkylating agents, and following digestion, the peptides are then separated by liquid chromatography or capillary electrophoresis and analyzed using MS.
  • MS converts components of a sample into gaseous ions, separates the ions based on their mass-to-charge ratio, and determines relative abundance.
  • a MALDI-TOF Microx Assisted Laser Desorption/Ionization-Time of Flight
  • ESI Electronpray Ionization
  • TOF-TOF Time of Flight/Time of Flight
  • analytical programs such as TURBOSEQUEST software (Finnigan, San Jose CA)
  • the MS data is compared against a database of theoretical MS data derived from known or predicted proteins. A minimum match of three peptide masses is usually required for reliable protein identification.
  • Tandem-MS may be used to derive information about individual peptides.
  • a first stage of MS is performed to determine individual peptide masses.
  • selected peptide ions are subjected to fragmentation using a technique such as collision induced dissociation to produce an ion series.
  • the resulting fragmentation ions are analyzed in a second round of MS, and their spectral pattern may be used to determine a short stretch of amino acid sequence (Dancik et al. (1999) J Comput Biol 6:327- 342).
  • Proteins or portions thereof may be produced not only by recombinant methods, but also by using chemical methods well known in the art.
  • Solid phase peptide synthesis may be carried out in a batchwise or continuous flow process which sequentially adds ⁇ -amino- and side chain-protected amino acid residues to an insoluble polymeric support via a linker group.
  • a linker group such as methylamine-derivatized polyethylene glycol is attached to poly(styrene-co-divinylbenzene) to form the support resin.
  • the amino acid residues are N- ⁇ -protected by acid labile Boc (t-butyloxycarbonyl) or base-labile Fmoc (9-fluorenylmethoxycarbonyl).
  • the carboxyl group of the protected amino acid is coupled to the amine of the linker group to anchor the residue to the solid phase support resin.
  • Trifluoroacetic acid or piperidine are used to remove the protecting group in the case of Boc or Fmoc, respectively.
  • Each additional amino acid is added to the anchored residue using a coupling agent or pre-activated amino acid derivative, and the resin is washed.
  • the full length peptide is synthesized by sequential deprotection, coupling of derivitized amino acids, and washing with dichloromethane and/or N, N-dimethylformamide. The peptide is cleaved between the peptide carboxy terminus and the linker group to yield a peptide acid or amide.
  • a protein or portion thereof may be purified by preparative high performance liquid chromatography and its composition confirmed by amino acid analysis or by sequencing (Creighton (1984) Proteins. Structures and Molecular Properties, WH Freeman, New York NY). Screening. Purification, and Immunoassays
  • a protein or a portion thereof encoded by the cDNA may be used to screen a library or a plurality of molecules or compounds for a ligand with specific binding affinity or to purify a molecule or compound from a sample.
  • the protein or portion thereof employed in such screening may be free in solution, affixed to an abiotic or biotic substrate, or located intracellularly.
  • viable or fixed prokaryotic host cells that are stably transformed with recombinant nucleic acids that have expressed and positioned a protein on their cell surface can be used in screening assays. The cells are screened against a library or a plurality of ligands and the specificity of binding or formation of complexes between the expressed protein and the ligand may be measured.
  • the ligands can be agonists, antagonists, antibodies, DNA molecules, enhancers, small drug molecules, immunoglobulins, inhibitors, mimetics, peptide nucleic acid molecules, peptides, pharmaceutical agents, proteins, and regulatory proteins, repressors, RNA molecules, ribozymes, and transcription factors, or any other test molecule or compound that specifically binds the protein.
  • An exemplary assay involves combining the mammalian protein or a portion thereof with the molecules or compounds under conditions that allow specific binding and detecting the bound protein to identify at least one ligand that specifically binds the protein.
  • This invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of binding the protein specifically compete with a test compound capable of binding to the protein or oligopeptide or fragment thereof.
  • One method for high throughput screening using very small assay volumes and very small amounts of test compound is described in USPN 5,876,946. Molecules or compounds identified by screening may be used in a model system to evaluate their toxicity, diagnostic, or therapeutic potential.
  • the protein may be used to purify a ligand from a sample.
  • a method for using a protein to purify a ligand would involve combining the protein or a portion thereof with a sample under conditions to allow specific binding, recovering the bound protein, and using an appropriate chaotropic agent to separate the protein from the purified ligand.
  • Immunoassay procedures may be used to quantify expression of the protein in cell cultures, in subjects with a particular disorder or in model animal systems under various conditions. Increased or decreased production of proteins as monitored by immunoassay may contribute to knowledge of the cellular activities associated with developmental pathways, engineered conditions or diseases, or treatment efficacy.
  • the quantity of a given protein in a given tissue may be determined by performing immunoassays on freeze-thawed detergent extracts of biological samples and comparing the slope of the binding curves to binding curves generated by purified protein.
  • a protein encoded by a cDNA of the invention may be used to produce specific antibodies.
  • Antibodies may be produced using an protein, polypeptide, peptide or oligopeptide with inherent antigenicity. Methods for producing antibodies include: 1) injecting an animal, usually goats, rabbits, or mice, with the protein, or an epitope or oligopeptide thereof, to induce an immune response; 2) engineering hybridomas to produce monoclonal antibodies; 3) inducing in vivo production in the lymphocyte population; or 4) screening libraries of recombinant immunoglobulins. Recombinant immunoglobulins may be produced as taught in USPN 4,816,567.
  • Antibodies produced using the proteins of the invention are useful for the diagnosis of prepathologic disorders as well as the diagnosis of chronic or acute diseases characterized by abnormalities in the expression, amount, or distribution of the protein.
  • a variety of protocols for competitive binding or immunoradiometric assays using either polyclonal or monoclonal antibodies specific for proteins are well known in the art.
  • Immunoassays typically involve the formation of complexes between a protein and its specific binding molecule or compound and the measurement of complex formation.
  • Immunoassays may employ a two-site, monoclonal-based assay that utilizes monoclonal antibodies reactive to two noninterfering epitopes on a specific protein or a competitive binding assay (Pound (1998) Immunochemical Protocols. Humana Press, Totowa NJ). Assays Using Antibodies
  • Antibodies directed against epitopes on a protein encoded by a cDNA of the invention may be used in assays to quantify the amount of protein found in a particular human cell. Such assays include methods utilizing the antibody and a label to detect expression level under normal or disease conditions.
  • the antibodies may be used with or without modification, and labeled by joining them, either covalently or noncovalently, with a labeling moiety.
  • Protocols for detecting and measuring protein expression using either polyclonal or monoclonal antibodies are well known in the art. Examples include ELISA, RIA, FACS, and arrays. Such immunoassays typically involve the formation of complexes between the protein and its specific antibody and the measurement of such complexes. Antibody Arrays
  • an antibody array can be used to study protein-protein interactions and phosphorylation.
  • a variety of protein ligands are immobilized on a membrane using methods well known in the art. The array is incubated in the presence of cell lysate until protenr.antibody complexes are formed.
  • Proteins of interest are identified by exposing the membrane to an antibody specific to the protein of interest.
  • a protein of interest is labeled with digoxigenin (DIG) and exposed to the membrane; then the membrane is exposed to anti-DIG antibody which reveals where the protein of interest forms a complex.
  • DIG digoxigenin
  • the identity of the proteins with which the protein of interest interacts is determined by the position of the protein of interest on the membrane.
  • Antibody arrays can also be used for high-throughput screening of recombinant antibodies. Bacteria containing antibody genes are robotically-picked and gridded at high density (up to 18,342 different double-spotted clones) on a filter. Up to 15 antigens at a time are used to screen for clones to identify those that express binding antibody fragments. These antibody arrays can also be used to identify proteins which are differentially expressed in samples (de Wildt et al. (2000) Nature Biotechnol 18:989-94).
  • Labeling of Molecules for Assay A wide variety of reporter molecules and conjugation techniques are known by those skilled in the art and can be used with cDNAs, polynucleotides, proteins, peptides or antibodies in screening, purification, and diagnostic assays. Synthesis of labeled molecules may be achieved using commercial kits for incorporation of a labeled nucleotide such as 32 P-dCTP, Cy3-dCTP or Cy5-dCTP or amino acid such as 35 S-methionine.
  • a labeled nucleotide such as 32 P-dCTP, Cy3-dCTP or Cy5-dCTP or amino acid such as 35 S-methionine.
  • Polynucleotides, cDNAs, proteins, or antibodies may be directly labeled with a reporter molecule by chemical conjugation to amines, thiols and other groups present in the molecules using reagents such as BIOD-PY or FITC (Molecular Probes, Eugene OR).
  • the proteins and antibodies may be labeled for purposes of assay by joining them, either covalently or noncovalently, with a reporter molecule that provides for a detectable signal.
  • a reporter molecule that provides for a detectable signal.
  • a wide variety of labels and conjugation techniques are known and have been reported in the scientific and patent literature including, but not limited to USPNs 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241.
  • the cDNAs, proteins, or antibodies of the invention may be used to detect and quantify differential gene or protein expression; absence, presence, or excess expression of mRNAs or proteins; or to monitor mRNA or protein levels during therapeutic intervention.
  • Disorders associated with altered expression of the nucleic or amino acid include chronic obstructive pulmonary disease, emphysema, asthma, and in particular, lung cancer.
  • These cDNAs, proteins or antibodies can also be utilized as markers of treatment efficacy against the disorders noted above and other disorders, conditions, and diseases over a period ranging from several days to months.
  • Assays for the presence or transcripts or protein may be hybridization or amplification technologies, ELISAs, RIAs, FACs or any other method used to compare gene or protein expression in a biological sample from a patient to standard samples in order to detect altered expression. Qualitative or quantitative methods for such comparisons are well known in the art.
  • the cDNA may be labeled by standard methods and added to a biological sample from a patient under conditions for hybridization complex formation. After an incubation period, the sample is washed and the amount of label (or signal) associated with hybridization complexes is quantified and compared with a standard value. If the amount of label in the patient sample is significantly altered in comparison to the standard value, then the presence of the associated condition, disease or disorder is indicated.
  • a normal or standard expression profile is established. This may be accomplished by combining a biological sample taken from normal subjects, either animal or human, with a probe under conditions for hybridization or amplification.
  • Standard hybridization may be quantified by comparing the values obtained using normal subjects with values from an experiment in which a known amount of a purified target sequence is used. Standard values obtained in this manner may be compared with values obtained from samples from patients who are symptomatic for a particular condition, disease, or disorder. Deviation from standard values toward those associated with a particular condition is used to diagnose that condition.
  • Such assays may also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies and in clinical trials or to monitor the treatment of an individual patient. Once the presence of a condition is established and a treatment protocol is initiated, diagnostic assays may be repeated on a regular basis to determine if the level of expression in the patient begins to approximate that which is observed in a normal subject. The results obtained from successive assays may be used to show the efficacy of treatment over a period ranging from several days to months.
  • a gene expression profile comprises a plurality of cDNAs and a plurality of detectable hybridization complexes, wherein each complex is formed by hybridization of one or more probes to one or more complementary nucleic acids in a sample.
  • the cDNAs of the invention are used as elements on an array to analyze gene expression profiles.
  • the array is used to monitor the progression of disease.
  • researchers can assess and catalog the differences in gene expression between healthy and diseased tissues or cells. By analyzing changes in patterns of gene expression, disease can be diagnosed at earlier stages before the patient is symptomatic.
  • the invention can be used to formulate a prognosis and to design a treatment regimen.
  • the invention can also be used to monitor the efficacy of treatment. For treatments with known side effects, the array is employed to improve the treatment regimen.
  • expression profiles can also be evaluated by methods including, but not limited to, differential display by spatial immobilization, gel electrophoresis, genome mismatch scanning, representational discriminate analysis, clustering, and transcript imaging, and protein or antibody arrays. Expression profiles produced by these methods may be used alone or in combination. The correspondence between mRNA and protein expression has been discussed by Zweiger (2001, Transducing the Genome. McGraw-Hill, San Francisco, CA) and Glavas et al. (2001, T cell activation upregulates cyclic nucleotide phosphodiesterases 8A1 and 7A3, Proc Natl Acad Sci 98:6319-6342) among others.
  • animal models which mimic a human disease can be used to characterize expression profiles associated with a particular condition, disorder or disease or its treatment. Novel treatment regimens may be tested in these animal models using arrays to establish and then follow expression profiles over time.
  • arrays can be used with cell cultures or tissues removed from animal models to rapidly screen large numbers of candidate drug molecules, looking for ones that produce an expression profile similar to those of known therapeutic drugs, with the expectation that molecules with the same expression profile will likely have similar therapeutic effects.
  • the invention provides the means to rapidly determine the molecular mode of action of a drug. THERAPEUTICS
  • cDNAs and fragments thereof can be used in gene therapy.
  • cDNAs can be delivered ex vivo to target cells, such as cells of bone marrow. Once stable integration and transcription and or translation are confirmed, the bone marrow may be reintroduced into the subject. Expression of the protein encoded by the cDNA may correct a disorder associated with mutation of a normal sequence, reduction or loss of an endogenous target protein, or overexpression of an endogenous or mutant protein.
  • cDNAs may be delivered in vivo using vectors such as retrovirus, adenovirus, adeno-associated virus, herpes simplex virus, and bacterial plasmids.
  • Non-viral methods of gene delivery include cationic liposomes, polylysine conjugates, artificial viral envelopes, and direct injection of DNA (Anderson (1998) Nature 392:25-30; Verbs et al. (1997) Oncol Res 9:313-325; Chu et al- (1998) J Mol Med 76(3-4): 184-192; Weiss et al- (1999) Cell Mol Life Sci 55(3):334-358; Agrawal (1996) Antisense Therapeutics. Humana Press, Totowa NJ; and August et al. (1997) Gene Therapy (Advances in Pharmacology, Vol. 40). Academic Press, San Diego CA).
  • expression of a particular protein can be regulated through the specific binding of a fragment of a cDNA to a genomic sequence or an mRNA which encodes the protein or directs its transcription or translation.
  • the cDNA can be modified or derivatized to any RNA-like or DNA-like material including peptide nucleic acids, branched nucleic acids, and the like.
  • Molecules which regulate the activity of the cDNA or expressed protein are useful as therapeutics for respiratory disorders including chronic obstructive pulmonary disease, emphysema, asthma and in particular, lung cancer.
  • Such molecules include agonists which increase the expression or activity of the polynucleotide or encoded protein, respectively; or antagonists which decrease expression or activity of the polynucleotide or encoded protein, respectively.
  • an antibody which specifically binds the protein may be used directly as an antagonist or indirectly as a delivery mechanism for bringing a pharmaceutical agent to cells or tissues which express the protein.
  • any of the proteins, or their ligands, or complementary nucleic acid sequences may be administered as pharmaceutical compositions or in combination with other appropriate therapeutic agents.
  • the combination of therapeutic agents may act synergistically to affect the treatment or prevention of the conditions and disorders associated with an immune response. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects.
  • the therapeutic agents may be combined with pharmaceutically- acceptable carriers including excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Further details on techniques for formulation and administration used by doctors and pharmacists may be found in the latest edition of Remington's Pharmaceutical Sciences (Mack Publishing, Easton PA). Model Systems
  • Animal models may be used as bioassays where they exhibit a phenotypic response similar to that of humans and where exposure conditions are relevant to human exposures. Mammals are the most common models, and most infectious agent, cancer, drug, and toxicity studies are performed on rodents such as rats or mice because of low cost, availability, lifespan, reproductive potential, and abundant reference literature. Inbred and outbred rodent strains provide a convenient model for investigation of the physiological consequences of underexpression or overexpression of genes of interest and for the development of methods for diagnosis and treatment of diseases. A mammal inbred to overexpress a particular gene (for example, secreted in milk) may also serve as a convenient source of the protein expressed by that gene. Transgenic Animal Models
  • Transgenic rodents that overexpress or underexpress a gene of interest may be inbred and used to model human diseases or to test therapeutic or toxic agents.
  • the introduced gene may be activated at a specific time in a specific tissue type during fetal or postnatal development. Expression of the transgene is monitored by analysis of phenotype, of tissue-specific mRNA expression, or of serum and tissue protein levels in transgenic animals before, during, and after challenge with experimental drug therapies.
  • Embryonic (ES) stem cells isolated from rodent embryos retain the potential to form embryonic tissues.
  • ES cells such as the mouse 129/SvJ cell line are placed in a blastocyst from the C57BL/6 mouse strain, they resume normal development and contribute to tissues of the live-born animal.
  • ES cells are preferred for use in the creation of experimental knockout and knockin animals.
  • the method for this process is well known in the art and the steps are: the cDNA is introduced into a vector, the vector is transformed into ES cells, transformed cells are identified and microinjected into mouse cell blastocysts, blastocysts are surgically transferred to pseudopregnant dams.
  • the resulting chimeric progeny are genotyped and bred to produce heterozygous or homozygous strains.
  • a region of a gene is enzymatically modified to include a non-natural intervening sequence such as the neomycin phosphotransferase gene (neo; Capecchi (1989) Science 244:1288-1292).
  • the modified gene is transformed into cultured ES cells and integrates into the endogenous genome by homologous recombination.
  • the inserted sequence disrupts transcription and translation of the endogenous gene.
  • ES cells can be used to create knockin humanized animals or transgenic animal models of human diseases.
  • knockin technology a region of a human gene is injected into animal ES cells, and the human sequence integrates into the animal cell genome.
  • Transgenic progeny or inbred lines are studied and treated with potential pharmaceutical agents to obtain info ⁇ nation on the progression and treatment of the analogous human condition.
  • cDNAs As described herein, the uses of the cDNAs, provided in the Sequence Listing of this application, and their encoded proteins are exemplary of known techniques and are not intended to reflect any limitation on their use in any technique that would be known to the person of average skill in the art.
  • the cDNAs provided in this application may be used in molecular biology techniques that have not yet been developed, provided the new techniques rely on properties of nucleotide sequences that are currently known to the person of ordinary skill in the art, e.g., the triplet genetic code, specific base pair interactions, and the like.
  • reference to a method may include combining more than one method for obtaining, assembling or expressing cDNAs that will be known to those skilled in the art.
  • RNA was purchased from Clontech Laboratories (Palo Alto CA) or isolated from various tissues. Some tissues were homogenized and lysed in guanidinium isothiocyanate, while others were homogenized and lysed in phenol or in a suitable mixture of denaturants, such as TRIZOL reagent (Invi-rogen). The resulting lysates were centrifuged over CsCl cushions or extracted with chloroform. RNA was precipitated with either isopropanol or ethanol and sodium acetate, or by other routine methods.
  • poly(A) RNA was isolated using oligo d(T)-coupled paramagnetic particles (Promega), OLIGOTEX latex particles (Qiagen, Valencia CA), or an OLIGOTEX mRNA purification kit (Qiagen).
  • poly(A) RNA was isolated directly from tissue lysates using other kits, including the POLY(A)PURE mRNA purification kit (Ambion, Austin TX). hi some cases, Stratagene (La Jolla CA) was provided with RNA and constructed the cDNA libraries.
  • cDNA was synthesized and cDNA libraries were constructed with the UNIZAP vector system (Stratagene) or SUPERSCRIPT plasmid system (Invitrogen) using the recommended procedures or similar methods known in the art. (See Ausubel, supra. Units 5.1 through 6.6.) Reverse transcription was initiated using oligo d(T) or random primers. Synthetic oligonucleotide adapters were ligated to double stranded cDNA, and the cDNA was digested with the appropriate restriction enzyme or enzymes.
  • the cDNA was size-selected (300-1000 bp) using SEPHACRYL S1000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (APB) or preparative agarose gel electrophoresis.
  • cDNAs were ligated into compatible restriction enzyme sites of the polylinker of the pBLUESCRIPT phagemid (Stratagene), pSPORTl plasmid (Invitrogen), or pINCY plasmid (Incyte Genomics). Recombinant plasmids were transformed into XLl-BLUE, XLl-BLUEMRF, or SOLR competent E.
  • the modified Soares normalization procedure was utilized to reduce the repetitive cloning of highly expressed, high abundance cDNAs while maintaining the overall sequence complexity of the library. Modification included significantly longer hybridization times which allowed for increased gene discovery rates by biasing the normalized libraries toward those infrequently expressed, low-abundance cDNAs which are poorly represented in a standard transcript image (Soares, supra).
  • Plasmids were recovered from host cells by in vivo excision using the UNIZAP vector system (Stratagene) or by cell lysis. Plasmids were purified using one of the following: the Magic or WIZARD M-MPREPS DNA purification system (Promega); the AGTC MIN-PREP purification kit (Edge BioSystems, Gaithersburg MD); the QIAWELL 8, QIAWELL 8 Plus, or QIAWELL 8 Ultra plasmid purification systems, or the REAL PREP 96 plasmid purification kit (Qiagen). Following precipitation, plasmids were resuspended in 0.1 ml of distilled water and stored, with or without lyophilization, at 4°C.
  • the Magic or WIZARD M-MPREPS DNA purification system Promega
  • AGTC MIN-PREP purification kit Edge BioSystems, Gaithersburg MD
  • QIAWELL 8, QIAWELL 8 Plus, or QIAWELL 8 Ultra plasmid purification systems or
  • plasmid DNA was amplified from host cell lysates using direct link PCR in a high-throughput format (Rao (1994) Anal Biochem 216: 1-14). Host cell lysis and thermal cycling steps were carried out in a single reaction mixture. Samples were processed and stored in 384-well plates, and the concentration of amplified plasmid DNA was quantified fluorometrically using PICOGREEN dye (Molecular Probes) and a FLUOROSKAN II fluorescence scanner (Labsystems Oy, Helsinki, Finland).
  • cDNA sequencing reactions were processed using standard methods or high-throughput instrumentation such as the CATALYST 800 thermal cycler (ABI) or the DNA ENGINE thermal cycler (MJ Research, Watertown MA) in conjunction with the HYDRA microdispenser (Robbins Scientific, Sunnyvale CA) or the MICROLAB 2200 system (Hamilton, Reno NV).
  • cDNA sequencing reactions were prepared using reagents provided by APB or supplied in sequencing kits such as the PRISM BIGDYE cycle sequencing kit (ABI).
  • Electrophoretic separation of cDNA sequencing reactions and detection of labeled cDNAs were carried out using the MEGABACE 1000 DNA sequencing system (APB); the PRISM 373 or 377 sequencing systems (ABI) in conjunction with standard protocols and base calling software; or other sequence analysis systems known in the art. Reading frames within the cDNA sequences were identified using standard methods and software (reviewed in Ausubel, supra, unit 1.1). Ill Extension of cDNA Sequences
  • Nucleic acid sequences were extended using the cDNA clones and oligonucleotide primers.
  • One primer was synthesized to initiate 5' extension of the known fragment, and the other, to initiate 3' extension of the known fragment.
  • the initial primers were designed using OLIGO primer analysis software (Molecular Biology Insights, Cascade CO), or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the target sequence at temperatures of about 68°C to about 72°C. Any stretch of nucleotides which would result in hairpin structures and primer-primer dimerizations was avoided.
  • Selected human cDNA libraries were used to extend the sequence. If more than one extension was necessary or desired, additional or nested sets of primers were designed. Preferred libraries are ones that have been size-selected to include larger cDNAs. Also, random primed libraries are preferred because they will contain more sequences with the 5' and upstream regions of genes. A randomly primed library is particularly useful if an oligo d(T) library does not yield a full-length cDNA. High fidelity amplification was obtained by PCR using methods well known in the art. PCR was performed in 96-well plates using the DNA ENGINE thermal cycler (MJ Research).
  • the reaction mix contained DNA template, 200 nmol of each primer, reaction buffer containing Mg 2+ , (NH 4 ) 2 S0 4 , and ⁇ -mercaptoethanol, Taq DNA polymerase (APB), ELONGASE enzyme (Invitrogen), and Pfu DNA polymerase (Stratagene), with the following parameters for primer pair PCI A and PCI B (Incyte Genomics): Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 60°C, 1 min; Step 4: 68°C, 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68°C, 5 min; Step 7: storage at 4°C.
  • the parameters for primer pair T7 and SK+ were as follows: Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 57°C, 1 min; Step 4: 68°C, 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68°C, 5 min; Step 7: storage at 4°C.
  • the concentration of DNA in each well was determined by dispensing 100 ⁇ l PICOGREEN reagent (0.25% reagent in lx TE, v/v; Molecular Probes) and 0.5 ⁇ l of undiluted PCR product into each well of an opaque fluorimeter plate (Corning Costar, Acton MA) and allowing the DNA to bind to the reagent.
  • the plate was scanned in a FLUOROSKAN II (Labsystems Oy) to measure the fluorescence of the sample and to quantify the concentration of DNA. A 5 ⁇ l to 10 ⁇ l aliquot of the
  • reaction mixture was analyzed by electrophoresis on a 1% agarose mini-gel to determine which reactions were successful in extending the sequence.
  • the extended nucleic acids were desalted and concentrated, transferred to 384-well plates, digested with CviJI cholera virus endonuclease (Molecular Biology Research, Madison WI), and sonicated or sheared prior to religation into pUC18 vector (APB).
  • CviJI cholera virus endonuclease Molecular Biology Research, Madison WI
  • API pUC18 vector
  • Step 1 94°C, 3 min
  • Step 2 94°C, 15 sec
  • Step 3 60°C, 1 min
  • Step 4 72°C, 2 min
  • Step 5 steps 2, 3, and 4 repeated 29 times
  • Step 6
  • DNA was quantified using PICOGREEN reagent (Molecular Probes) as described above. Samples with low DNA recoveries were reamplified using the same conditions described above. Samples were diluted with 20% dimethylsulfoxide (DMSO; 1:2, v/v), and sequenced using DYENAMIC energy transfer sequencing primers and the DYENAMIC DIRECT cycle sequencing kit (APB) or the PRISM BIGDYE te ⁇ ninator cycle sequencing kit (ABI).
  • DMSO dimethylsulfoxide
  • API DYENAMIC DIRECT cycle sequencing kit
  • ABSI PRISM BIGDYE te ⁇ ninator cycle sequencing kit
  • nucleic acid sequences of the cDNAs presented in the Sequence Listing may contain occasional sequencing errors and unidentified nucleotides (N) that reflect state-of-the-art technology at the time the cDNA was sequenced. Occasional sequencing errors and Ns may be resolved and SNPs verified either by resequencing the cDNA or using algorithms to compare multiple sequences;
  • sequences may be analyzed using a variety of algorithms described in Ausubel (supra, unit 1.1) and in Meyers (1995; Molecular Biology and Biotechnology, Wiley VCH, New York NY, pp. 856-853).
  • sequences from chromatograms were subjected to PHRED analysis (Phil Green, University of Washington, Seattle WA) and assigned a quality score.
  • the sequences 5 having at least a required quality score were subject to various pre-processing algorithms to eliminate low quality 3' ends, vector and linker sequences, polyA tails, Alu repeats, mitochondrial and ribosomal sequences, bacterial contamination sequences, and sequences smaller than 50 base pairs.
  • Sequences were screened using the BLOCK 2 program (Incyte Genomics), a motif analysis program based on sequence information contained in the SWISS-PROT and PROSITE databases (Bairoch et
  • BLASTn (vers.1.4, WashU)
  • CROSSMATCH software (Green, supra).
  • Candidate pairs were identified as all BLAST hits having a quality score greater than or equal to 150. Alignments of at least 82% local identity were accepted into the bin. The component sequences from each bin were assembled using PHRAP (Green, supra). Bins with several overlapping component sequences were assembled using DEEP PHRAP (Green, supra).
  • Bins were compared against each other, and those having local similarity of at least 82% were combined and reassembled. Reassembled bins having templates of insufficient overlap (less than 95% local identity) were re-split. Assembled templates were also subjected to analysis by STITCHER/EXON MAPPER algorithms which analyzed the probabilities of the presence of splice variants, alternatively spliced exons, splice junctions, differential expression of alternative spliced
  • Template sequences were subjected to motif, BLAST, Hidden Markov Model (HMM; Pearson and Lipman (1988) Proc Natl Acad Sci 85:2444-2448; Smith and Waterman (1981) J Mol Biol 147:195-197), and functional analyses, and categorized in protein hierarchies using methods described in USSN 08/812,290, filed March 6, 1997; USSN 08/947,845, filed October 9, 1997; USPN 5,953,727; and USSN 09/034,807, filed March 4, 1998. Template sequences may be further queried against public databases such as the GenBank rodent, mammalian, vertebrate, eukaryote, prokaryote, and human EST databases.
  • Incyte cDNAs represent template sequences derived from the LIFESEQ
  • cDNAs were amplified from bacterial cells using primers complementary to vector sequences flanking the cDNA insert. Thirty cycles of PCR increased the initial quantity of cDNAs from 1-2 ng to a final quantity greater than 5 ⁇ g. Amplified cDNAs were then purified using SEPHACRYL-400 columns (APB). Purified cDNAs were immobilized on polymer-coated glass slides. Glass microscope slides (Corning, Corning NY) were cleaned by ultrasound in 0.1% SDS and acetone, with extensive distilled water washes between and after treatments.
  • Microarrays were UV-crosslinked using a STRATALINKER UV-crosslinker (Stratagene), and then washed at room temperature once in 0.2% SDS and three times in distilled water. Nonspecific binding sites were blocked by incubation of microarrays in 0.2% casein in phosphate buffered saline (Tropix, Bedford MA) for 30 minutes at 60°C followed by washes in 0.2% SDS and distilled water as before.
  • Pulmonary carcinoid, stage IA, and grossly uninvolved lung tissue were removed from a 79 year-old male donor (RCIC).
  • the tumor sample showed 100% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue.
  • Dn 7165 Poorly differentiated, adenocarcinoma, stage IA, and grossly uninvolved lung tissue were removed from a 70 year-old female donor (RCIC).
  • the tumor sample showed 20% overt tumor cells within the tumor.
  • Moderately differentiated, squamous cell carcinoma, stage DA, and grossly uninvolved lung tissue were removed from a 73 year-old male donor (RCIC).
  • the tumor sample showed 60% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • Moderately differentiated, adenocarcinoma, stage IA, and grossly uninvolved lung tissue were removed from a 71 year-old female donor (RCIC).
  • the tumor sample showed 75% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • Dn 5796 Moderately differentiated, squamous cell carcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 66 year-old male donor (RCIC).
  • the tumor sample showed 50% overt tumor cells within the tumor, and less than 10% overt tumor cells in the uninvolved tissue.
  • Moderately differentiated, squamous cell carcinoma, stage IDB, and grossly uninvolved lung tissue were removed from a 70 year-old male donor (RCIC).
  • the tumor sample showed 70% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • Moderately differentiated, squamous cell carcinoma, stage HB, and grossly uninvolved lung tissue were removed from a 73 year-old male donor (RCIC).
  • the tumor sample showed 80% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • adenocarcinoma adenocarcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 67 year-old male donor (RCIC).
  • the tumor sample showed 50% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue.
  • Dn 7176 adenocarcinoma, stage IB, and grossly uninvolved lung tissue was removed from a 67 year-old male donor (RCIC).
  • the tumor sample showed 50% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue.
  • Atypical carcinoid, stage IA, and grossly uninvolved lung tissue were removed from a 61 year-old male donor (RCIC).
  • the tumor sample showed 90% overt tumor cells within the tumor. Dn 5799
  • Moderately differentiated, adenocarcinoma, stage IIB, and grossly uninvolved lung tissue were removed from a 66 year-old female donor (RCIC).
  • the tumor sample showed 80% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue.
  • Dn 7188 Poorly differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 54 year-old male donor (RCIC).
  • the tumor sample showed 90% overt tumor cells within the tumor, and less than 10% overt tumor cells in the uninvolved tissue.
  • Moderately differentiated, squamous cell carcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 50 year-old female donor (RCIC).
  • the tumor sample showed 70% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue.
  • Moderately differentiated, squamous cell carcinoma, stage DB, and grossly uninvolved lung tissue were removed from a 60 year-old female donor (RCIC).
  • the tumor sample showed 50% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • Moderately differentiated, squamous cell carcinoma, stage DB, and grossly uninvolved lung tissue were removed from a 75 year-old female donor (RCIC).
  • the tumor sample showed 70% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • RCIC 53 year-old male donor
  • the tumor sample showed 70% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • Dn 7962 Moderately differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 71 year-old male donor (RCIC).
  • the tumor sample showed 85% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue.
  • the patient had undergone 3 cycles of chemotherapy with partial remission of the tumor prior to surgery.
  • Dn 7963 Poorly differentiated, adenocarcinoma, stage DIA, and grossly uninvolved lung tissue were removed from a 71 year-old male donor (RCIC).
  • the tumor sample showed 60% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • the patient had undergone 3 cycles of chemotherapy with progressive disease in the tumor prior to surgery.
  • Dn 7964 Moderately differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 50 year-old male donor (RCIC).
  • the tumor sample showed 70% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue.
  • Dn 7965 Moderately differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 54 year-old female donor (RCIC).
  • the tumor sample showed 60% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • the patient had undergone 3 cycles of chemotherapy with no change in the tumor prior to surgery.
  • Dn 7966 Moderately differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 39 year-old female donor (RCIC).
  • the tumor sample showed 10% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • the patient had undergone 3 cycles of chemotherapy with complete remission of the tumor prior to surgery.
  • Dn 7967 Moderately differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 57 year-old female donor (RCIC).
  • the tumor sample showed 60% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue.
  • the patient had undergone 3 cycles of chemotherapy with no change in the tumor prior to surgery.
  • Dn 7968 Moderately differentiated, squamous cell carcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 58 year-old male donor (RCIC).
  • the tumor sample showed 10% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • the patient had undergone 3 cycles of chemotherapy with no change in the tumor prior to surgery.
  • Dn 7969 Poorly differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 62 year-old male donor (RCIC).
  • the tumor sample showed 60% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue.
  • the tumor sample showed no overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • the tumor sample showed 10% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • the tumor sample showed 60% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue.
  • the patient had undergone 3 cycles of chemotherapy with no change in the tumor prior to surgery.
  • Dn 7975 Poorly differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 62 year-old male donor (RCIC).
  • the tumor sample showed 10% overt tumor cells within the tumor.
  • the patient had undergone 3 cycles of chemotherapy with partial remission of the tumor prior to surgery.
  • Donor samples Dn 7160, Dn 7165, Dn 7186, and Dn 7192 were matched against a pool of normal/uninvolved tissues from Dn 7173, 7175, 7176, 7178, 7188, 7189, 7190, 7191, 7196, and 7197.
  • Donor sample Dn 7975 was matched against a pool of normal/uninvolved tissues fromDn 7964, 7965, 7966, 7967, and 7968. All other patient samples were matched against normal/uninvolved tissue from the same patient.
  • Each of the lung tissue samples was lysed in 1 ml of TRIZOL reagent (Invitrogen).
  • RNA pellet was washed with 1 ml of 70% ethanol, centrifuged at 15,000 rpm at 4°C, and resuspended in RNAse-free water. The concentration of the total RNA was determined by measuring the optical density at 260 nm.
  • Poly(A) RNA was prepared using an OLIGOTEX mRNA kit (Qiagen) with the following modifications: OLIGOTEX beads were washed in tubes instead of on spin columns, resuspended in elution buffer, and then loaded onto spin columns to recover mRNA. To obtain maximum yield, the mRNA was eluted twice.
  • Each poly(A) RNA sample was reverse transcribed using a cDNA synthesis system (Invitrogen) with Not I-T7-VN primers (5' GCATTAGCGGCCGCGAAATTAATACGACTCACTATAGGGAGAT TTTTTTTTTTTTTTTTVN 3 * ) and 100 units MMLV RNAseH (-) reverse-transcriptase (Progmega) in the first strand reaction.
  • the resulting cDNA was purified on a CHROMASPDSf TE-200 column (Clontech) and lyophilized until dry.
  • the cDNA was amplified 200-400 fold using an AMPLISCRIBE IVT kit (Epicentre Technologies, Madison WI) in a procedure modified from USPN 5,716,785 and USPN 5,891,636.
  • the amplified RNA was purified on a CHROMASPIN DEPC-200 column (Clontech).
  • the reverse transcription reaction was performed in a 25 ml volume containing 200 ng poly(A) RNA using the GEMBRIGHT kit (Incyte Genomics).
  • control poly(A) RNAs (YCFR06, YCFR45, YCFR67, YCFR85, YCFR43, YCFR22, YCFR23, YCFR25, YCFR44, YCFR26) were synthesized by in vitro transcription from non-coding yeast genomic DNA (W. Lei, unpublished).
  • control RNAs (YCFR06, YCFR45, YCFR67, and YCFR85) at 0.002ng, 0.02ng, 0.2 ng, and 2ng were diluted into reverse transcription reaction at ratios of 1:100,000, 1:10,000, 1:1000, 1:100 (w/w) to sample mRNA, respectively.
  • control mRNAs (YCFR43, YCFR22, YCFR23, YCFR25, YCFR44, YCFR26) were diluted into reverse transcription reaction at ratios of 1:3, 3:1, 1:10, 10: 1, 1:25, 25: 1 (w/w) to sample mRNA. Reactions were incubated at 37°C for 2 hr, treated with 2.5 ml of 0.5M sodium hydroxide, and incubated for 20 minutes at 85°C to the stop the reaction and degrade the RNA. cDNAs were purified using two successive CHROMA SPESf 30 gel filtration spin columns (Clontech).
  • Hybridization reactions compared cDNAs derived from tumor tissue and uninvolved tissue from the same donor. cDNA from all donors except Dn5795 were hybridized to HUMAN GENOME GEMs 1-5. cDNAs from Dn5795 were hybridized to HUMAN GENOME GEM 1.
  • Hybridization reactions contained 9 ⁇ l of sample mixture containing 0.2 ⁇ g each of Cy3 and Cy5 labeled cDNA synthesis products in 5X SSC, 0.2% SDS hybridization buffer. The mixture was heated to 65°C for 5 minutes and was aliquoted onto the microarray surface and covered with an 1.8 cm 2 coverslip. The microarrays were transferred to a waterproof chamber having a cavity just slightly larger than a microscope slide.
  • the chamber was kept at 100% humidity internally by the addition of 140 ⁇ l of 5x SSC in a corner of the chamber.
  • the chamber containing the microarrays was incubated for about 6.5 hours at 60°C.
  • the microarrays were washed for 10 min at 45°C in low stringency wash buffer (lx SSC, 0.1% SDS), three times for 10 minutes each at 45°C in high stringency wash buffer (O.lx SSC), and dried.
  • Reporter-labeled hybridization complexes were detected with a microscope equipped with an Innova 70 mixed gas 10 W laser (Coherent, Santa Clara CA) capable of generating spectral lines at 488 nm for excitation of Cy3 and at 632 nm for excitation of Cy5.
  • the excitation laser light was focused on the microarray using a 20X microscope objective (Nikon, Melville NY).
  • the slide containing the microarray was placed on a computer-controlled X-Y stage on the microscope and raster-scanned past the objective.
  • the 1.8 cm x 1.8 cm microarray used in the present example was scanned with a resolution of 20 micrometers. In two separate scans, the mixed gas multiline laser excited the two fluorophores sequentially.
  • Emitted light was split, based on wavelength, into two photomultiplier tube detectors (PMT R1477; Hamamatsu Photonics Systems, Bridgewater NJ) corresponding to the two fluorophores.
  • PMT R1477 Hamamatsu Photonics Systems, Bridgewater NJ
  • Appropriate filters positioned between the microarray and the photomultiplier tubes were used to filter the signals.
  • the emission maxima of the fluorophores used were 565 nm for Cy3 and 650 nm for Cy5.
  • Each microarray was typically scanned twice, one scan per fluorophore using the appropriate filters at the laser source, although the apparatus was capable of recording the spectra from both fluorophores simultaneously.
  • the sensitivity of the scans was calibrated using the signal intensity generated by a cDNA control species.
  • Samples of the calibrating cDNA were separately labeled with the two fluorophores and identical amounts of each were added to the hybridization mixture.
  • a specific location on the microarray contained a complementary DNA sequence, allowing the intensity of the signal at that location to be correlated with a weight ratio of hybridizing species of 1:100,000.
  • the output of the photomultiplier tube was digitized using a 12-bit RTI-835H analog-to-digital (A/D) conversion board (Analog Devices, Norwood, MA) installed in an IBM-compatible PC computer. 5
  • the digitized data were displayed as an image where the signal intensity was mapped using a linear 20- color transformation to a pseudocolor scale ranging from blue (low signal) to red (high signal).
  • the data was also analyzed quantitatively. Where two different fluorophores were excited and measured simultaneously, the data were first corrected for optical crosstalk (due to overlapping emission spectra) between the fluorophores using each fluorophore 's emission spectrum.
  • Array elements that exhibited a 4-fold or greater change in expression at one or more time points, a signal intensity over 250 units, a signal-to-background ratio of at least 2.5, and an element spot size of at least 40% were identified as differentially expressed using the GEMTOOLS program (Incyte 20 Genomics).
  • the cDNAs that were differentially expressed are shown in Tables 1-4 and are identified by both SEQ ID NO and Incyte ID.
  • Genomics using BLAST analysis and an Incyte template was chosen for each clone.
  • the template was 25 aligned against GenBank database using BLAST analysis to acquire annotation.
  • the nucleotide sequences were translated into amino acid sequences which were aligned against GenPept and other protein databases to acquire annotation and characterization.
  • Percent sequence identity can be determined electronically for two or more amino acid or nucleic acid sequences using the MEGALIGN program, a component of LASERGENE software 30 (DNASTAR). The percent identity between two amino acid sequences is calculated by dividing the length of sequence A, minus the number of gap residues in sequence A, minus the number of gap residues in sequence B, into the sum of the residue matches between sequence A and sequence B, times one hundred. Gaps of low or of no homology between the two amino acid sequences are not included in determining percentage identity. 35 Sequences with conserved protein motifs may be searched using the BLOCKS search program.
  • PROSITE database is a useful source for identifying functional or structural domains that are not detected using motifs due to extreme sequence divergence. Using weight matrices, these domains are calibrated against the SWISSPROT database to obtain a measure of the chance distribution of the matches.
  • the PRINTS database can be searched using the BLIMPS search program to obtain protein family "fingerprints".
  • the PRINTS database complements the PROSITE database by exploiting groups of conserved motifs within sequence alignments to build characteristic signatures of different protein families. For both BLOCKS and PRINTS analyses, the cutoff scores for local similarity were:
  • Pfam is a large collection of multiple sequence alignments and hidden Markov models covering many common protein domains. Version 5.5 of Pfam (Sept 2000) contains alignments and models for 2478 protein families, based on the SWISSPROT 38 and SP-TrEMBL 11 protein sequence databases.
  • hybridization technologies utilize a variety of substrates such as nylon membranes, capillary tubes, etc.
  • Arranging cDNAs on polymer coated slides is described in Example V; sample cDNA preparation, hybridization, and analysis using polymer coated slides is described in examples VI and VII, respectively.
  • the cDNAs are applied to a membrane substrate by one of the following methods.
  • a mixture of cDNAs is fractionated by gel electrophoresis and transferred to a nylon membrane by capillary transfer.
  • the cDNAs are individually ligated to a vector and inserted into bacterial host cells to form a library.
  • the cDNAs are then arranged on a substrate by one of the following methods, hi the first method, bacterial cells containing individual clones are robotically picked and arranged on a nylon membrane.
  • the membrane is placed on LB agar containing selective agent (carbenicillin, kanamycin, ampicillin, or chloramphenicol depending on the vector used) and incubated at 37°C for 16 hours.
  • the membrane is removed from the agar and consecutively placed colony side up in 10% SDS, denaturing solution (1.5 M NaCl, 0.5 M NaOH ), neutralizing solution (1.5 M NaCl, 1 M Tris, pH 8.0), and twice in 2x SSC for 10 minutes each.
  • the membrane is then UV irradiated in a STRATALINKER UV- crosslinker (Stratagene).
  • cDNAs are amplified from bacterial vectors by thirty cycles of PCR using primers complementary to vector sequences flanking the insert. PCR amplification increases a starting concentration of 1-2 ng nucleic acid to a final quantity greater than 5 ⁇ g.
  • Amplified nucleic acids from about 400 bp to about 5000 bp in length are purified using SEPHACRYL-400 beads (APB). Purified nucleic acids are arranged on a nylon membrane manually or using a dot/slot blotting manifold and suction device and are immobilized by denaturation, neutralization, and UV irradiation as described above.
  • Hybridization probes derived from cDNAs of the Sequence Listing are employed for screening cDNAs, mRNAs, or genomic DNA in membrane-based hybridizations. Probes are prepared by diluting the cDNAs to a concentration of 40-50 ng in 45 ⁇ l Tris-EDTA (ethylenediamine tetraacetic acid) (TE) buffer, denaturing by heating to 100°C for five minutes and centrifuging briefly. The denatured cDNA is then added to a REDIPRIME tube (APB), gently mixed until blue color is evenly distributed, and centrifuged briefly. Five microliters of [ 32 P]dCTP is added to the tube, and the contents are incubated at 37°C for 10 minutes.
  • Tris-EDTA ethylenediamine tetraacetic acid
  • the labeling reaction is stopped by adding 5 ⁇ l of 0.2M EDTA, and probe is purified from unincorporated nucleotides using a PROBEQUANT G-50 microcolumn (APB).
  • the purified probe is heated to 100°C for five minutes and then snap cooled for 2 minutes on ice.
  • Membranes are pre-hybridized in hybridization solution containing 1% Sarkosyl and lx high phosphate buffer (0.5 M NaCl, 0.1 M Na 2 HPO 4 , 5 mM EDTA, pH 7) at 55°C for 2 hours.
  • the probe diluted in 15 ml fresh hybridization solution, is then added to the membrane.
  • the membrane is hybridized with the probe at 55°C for 16 hours.
  • the membrane is washed for 15 minutes at 25°C in lmM Tris (pH 8.0), 1% Sarkosyl, and four times for 15 minutes each at 25°C in lmM Tris (pH 8.0).
  • XOMAT-AR film Eastman Kodak, Rochester NY
  • cDNA is synthesized from 1 ug total RNA in a 25 ul reaction with 100 units M-
  • MLV reverse transcriptase (Ambion, Austin TX), 0.5 mM dNTPs (Epicentre, Madison WI), and 40 ng/ml random hexamers (Fisher Scientific, Chicago IL). Reactions are incubated at 25C for 10 minutes, 42C for 50 minutes, and 70C for 15 minutes, diluted to 500 ul, and stored at -30C.
  • cDNA is obtained from Human MTC panels (Clontech Laboratories, Palo Alto CA).
  • PCR primers and probes (5' 6-FAM-labeled, 3' TAMRA) are designed using PRIMER EXPRESS 1.5 software (ABI) and synthesized by Biosearch Technologies (Novato CA) or ABI.
  • QPCR reactions are performed using an PRISM 7700 sequencing system (ABI) in 25 ul total volume with 5 ul cDNA template, lx TAQMAN UNIVERSAL PCR master mix (ABI), 100 nM each PCR primer, 200 nM probe, and lx VIC-labeled beta-2-microglobulin endogenous control (ABI). Reactions are incubated at 50C for 2 minutes, 95C for 10 minutes, followed by 40 cycles of incubation at 95C for 15 seconds and 60C for 1 minute.
  • ABSI PRISM 7700 sequencing system
  • XI Northern Analysis and Transcript Imaging Northern analysis is a laboratory technique used to detect the presence of a transcript of a gene and involves the hybridization of a labeled nucleotide sequence to a membrane on which RNAs from a particular cell type or tissue have been bound (Sambrook, supra, ch. 7 and Ausubel, supra, ch. 4 and 16.)
  • Analogous computer techniques applying BLAST are used to search for identical or related molecules in nucleotide databases such as GenBank or the LIFESEQ database (Incyte Genomics). This analysis is faster than multiple membrane-based hybridizations.
  • the sensitivity of the computer search can be modified to determine whether any particular match is categorized as exact or homologous.
  • the basis of the search is the product score, which is defined as:
  • the product score takes into account both the degree of similarity between two sequences and the length of the sequence match. For example, with a product score of 40, the match will be exact within a 1% to
  • homologous molecules are usually identified by selecting those which show product scores between 15 and 40, although lower scores may identify related molecules.
  • a transcript image is performed using the LIFESEQ GOLD database (Incyte Genomics). This process allows assessment of the relative abundance of the expressed polynucleotides in all of the cDNA libraries and was described in USPN 5,840,484, incorporated herein by reference. All sequences and cDNA libraries in the LIFESEQ database are categorized by system, organ/tissue and cell type.
  • the categories include cardiovascular system, connective tissue, digestive system, embryonic structures, endocrine system, exocrine glands, female and male genitalia, germ cells, hemic/immune system, liver, musculoskeletal system, nervous system, pancreas, respiratory system, sense organs, skin, stomatognathic system, unclassified/mixed, and the urinary tract. Criteria for transcript imaging are selected from category, number of cDNAs per library, library description, disease indication, clinical relevance of sample, and the like.
  • the number of libraries in which the sequence is expressed are counted and shown over the total number of libraries in that category.
  • the number of cDNAs are counted and shown over the total number of cDNAs in that library.
  • all enriched, normalized or subtracted libraries, which have high copy number sequences can be removed prior to processing, and all mixed or pooled tissues, which are considered non-specific in that they contain more than one tissue type or more than one subject's tissue, can be excluded from the analysis. Treated and untreated cell lines and/or fetal tissue data can also be excluded where clinical relevance is emphasized.
  • fetal tissue can be emphasized wherever elucidation of inherited disorders or differentiation of particular adult or embryonic stem cells into tissues or organs such as heart, kidney, nerves or pancreas would be aided by removing clinical samples from the analysis.
  • Transcript imaging can be used to support data from other methodologies and technologies.
  • Radiation hybrid and genetic mapping data available from public resources such as the Stanford Human Genome Center (SHGC), Whitehead Institute for Genome Research (WIGR), and Genethon are used to determine if any of the cDNAs presented in the Sequence Listing have been mapped. Any of the fragments of the cDNA encoding a particular protein that have been mapped result in the assignment of all related regulatory and coding sequences to the same location.
  • the genetic map locations are described as ranges, or intervals, of human chromosomes. The map position of an interval, in cM (which is roughly equivalent to 1 megabase of human DNA), is measured relative to the terminus of the chromosomal p-arm.
  • the pUB6/V5-His vector system (Invitrogen) is used to express GSCC in CHO cells.
  • the vector contains the selectable bsd gene, multiple cloning sites, the promoter/enhancer sequence from the human ubiquitin C gene, a C-terminal V5 epitope for antibody detection with anti-V5 antibodies, and a C-terminal polyhistidine (6xHis) sequence for rapid purification on PROBOND resin (Invitrogen).
  • Transformed cells are selected on media containing blasticidin.
  • Spodoptera frugiperda (Sf9) insect cells are infected with recombinant Autographica californica nuclear polyhedrosis virus (baculovirus).
  • the polyhedrin gene is replaced with the cDNA by homologous recombination and the polyhedrin promoter drives cDNA transcription.
  • the protein is synthesized as a fusion protein with 6xhis which enables purification as described above. Purified protein is used in the following activity and to make antibodies.
  • Purification using polyacrylamide gel electrophoresis or similar techniques is used to isolate protein for immunization of hosts or host cells to produce antibodies using standard protocols.
  • a denatured protein from a reverse phase HPLC separation is obtained in quantities up to 75 mg.
  • This denatured protein is used to immunize mice or rabbits following standard protocols. About 100 ⁇ g is used to immunize a mouse, while up to 1 mg is used to immunize a rabbit.
  • the denatured protein is radioiodinated and incubated With murine B-cell hybridomas to screen for monoclonal antibodies. About 20 mg of protein is sufficient for labeling and screening several thousand clones.
  • the amino acid sequence translated from a cDNA of the invention is analyzed using PROTEAN software (DNASTAR) to select regions of high antigenicity, antigenically- effective epitopes of the protein.
  • oligopeptides about 15 residues in length are synthesized using a 431 peptide synthesizer (ABI) using Fmoc-chemistry and then coupled to keyhole limpet hemocyanin (KLH; Sigma-Aldrich) by reaction with M-maleimidobenzoyl-N-hydroxysuccinimide ester. If necessary, a cysteine may be introduced at the N-te ⁇ ninus of the peptide to permit coupling to KLH.
  • ABSI peptide synthesizer
  • KLH keyhole limpet hemocyanin
  • Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant.
  • the resulting antisera are tested for antipeptide activity by binding the peptide to plastic, blocking with 1% BSA, reacting with rabbit antisera, washing, and reacting with radioiodinated goat anti-rabbit IgG.
  • Hybridomas are prepared and screened using standard techniques. Hybridomas of interest are detected by screening with radioiodinated protein to identify those fusions producing a monoclonal antibody specific for the protein. In a typical protocol, wells of 96 well plates (FAST,
  • Becton-Dickinson, Palo Alto CA) are coated with affinity-purified, specific rabbit-anti-mouse (or suitable anti-species Ig) antibodies at 10 mg/ml.
  • the coated wells are blocked with 1% BSA, washed, and exposed to supematants from hybridomas. After incubation, the wells are exposed to radiolabeled protein at 1 mg/ml. Clones producing antibodies bind a quantity of labeled protein that is detectable above background.
  • Such clones are expanded and subjected to 2 cycles of cloning at 1 cell/3 wells.
  • Cloned hybridomas are injected into pristane-treated mice to produce ascites, and monoclonal antibody is purified from the ascitic fluid by affinity chromatography on protein A (APB).
  • Monoclonal antibodies with affinities of at least 10 8 M " ⁇ preferably 10 9 to 10 10 M "1 or stronger, are made by procedures well known in the art.
  • Naturally occurring or recombinant protein is immunopurified by affinity chromatography using antibodies specific for the protein.
  • An immunoaffinity column is constructed by covalently coupling the antibody to CNBr-activated SEPHAROSE resin (APB). Media containing the protein is passed over the immunoaffinity column, and the column is washed using high ionic strength buffers in the presence of detergent to allow preferential absorbance of the protein. After coupling, the protein is eluted from the column using a buffer of pH 2-3 or a high concentration of urea or thiocyanate ion to disrupt antibody/protein binding, and the protein is collected.
  • APB CNBr-activated SEPHAROSE resin
  • XVI Screening Molecules for Specific Binding with the cDNA or Protein The cDNA or fragments thereof and the protein or portions thereof are labeled with 32 P-dCTP, Cy3-dCTP, Cy5-dCTP (APB), or BIODIPY or FITC (Molecular Probes), respectively.
  • Candidate molecules or compounds previously arranged on a substrate are incubated in the presence of labeled nucleic or amino acid. After incubation under conditions for either a cDNA or a protein, the substrate is washed, and any position on the substrate retaining label, which indicates specific binding or complex formation, is assayed. The binding molecule is identified by its arrayed position on the substrate.
  • an antibody array can be used to study protein-protein interactions and phosphorylation.
  • a variety of protein ligands are immobilized on a membrane using methods well known in the art. The array is incubated in the presence of cell lysate until proteimantibody complexes are formed. Proteins of interest are identified by exposing the membrane to an antibody specific to the protein of interest.
  • a protein of interest is labeled with digoxigenin (DIG) and exposed to the membrane; then the membrane is exposed to anti- DIG antibody which reveals where the protein of interest forms a complex.
  • DIG digoxigenin
  • the identity of the proteins with which the protein of interest interacts is determined by the position of the protein of interest on the membrane.
  • Antibody arrays can also be used for high-throughput screening of recombinant antibodies. Bacteria containing antibody genes are robotically-picked and gridded at high density (up to 18,342 different double-spotted clones) on a filter. Up to 15 antigens at a time are used to screen for clones to identify those that express binding antibody fragments. These antibody arrays can also be used to identify proteins which are differentially expressed in samples (de Wildt, supra) XVIII Two-Hybrid Screen
  • a yeast two-hybrid system MATCHMAKER LexA Two-Hybrid system (Clontech Laboratories), is used to screen for peptides that bind the protein of the invention.
  • a cDNA encoding the protein is inserted into the multiple cloning site of a pLexA vector, ligated, and transformed into E. coli.
  • cDNA, prepared from mRNA is inserted into the multiple cloning site of a pB42AD vector, ligated, and transformed into E. coli to construct a cDNA library.
  • the pLexA plasmid and pB42AD- cDNA library constructs are isolated from E.
  • Transformed yeast cells are plated on synthetic dropout (SD) media lacking histidine (-His), tryptophan (-Trp), and uracil (-Ura), and incubated at 30C until the colonies have grown up and are counted.
  • SD synthetic dropout
  • the colonies are pooled in a minimal volume of lx TE (pH 7.5), replated on SD/-His/-Leu/-Trp/-Ura media supplemented with 2% galactose (Gal), 1% raffinose (Raf), and 80 mg/ml 5-bromo-4-chloro-3-indolyl ⁇ -d-galactopyranoside (X-Gal), and subsequently examined for growth of blue colonies.
  • Interaction between expressed protein and cDNA fusion proteins activates expression of a LEU2 reporter gene in EGY48 and produces colony growth on media lacking leucine (-Leu).
  • Interaction also activates expression of ⁇ -galactosidase from the p8op-lacZ reporter construct that produces blue color in colonies grown on X-Gal.

Abstract

The present invention relates to a combination comprising a plurality of cDNAs or a plurality of proteins which are differentially expressed in a respiratory disorder and which may be used to diagnose, to stage, to treat, or to monitor the treatment of a subject with a respiratory disorder.

Description

DIAGNOSTIC MARKERS FOR LUNG CANCER
TECHNICAL FIELD
The present invention relates to a combination comprising a plurality of cDNAs or a plurality of proteins which are differentially expressed in lung cancer and which may be used to diagnose, to stage, to treat, or to monitor the progression or treatment of respiratory disorders and in particular, lung cancer.
BACKGROUND OF THE INVENTION Bronchogenic carcinoma is a leading cause of cancer death in the United States affecting men (32%) and women (25%) each year. Cigarette smoking is the largest contributory factor in the diagnosis of males (90%) and females (85%) while occupational hazards, such as asbestos, arsenic, nickel and chloromethyl esters account for another small proportion of these cancers (15% in men and 5% in women). Damage to the DNA, activation of cellular oncogenes, and stimulation by growth factors are of primary importance in disease development. In nearly 80% of patients diagnosed with lung cancer, metastasis to the pleura, brain, bone, pericardium, and liver has already occurred. The decision to treat with surgery, radiation therapy, or chemotherapy is made on the basis of tumor histology, response to hormones or growth factors, and sensitivity to inhibitors or drugs. With current treatments, most patients die within one year of diagnosis.
The three most common types of bronchogenic carcinoma are squamous cell, small cell, and adenocarcinoma. These cancers progress through a series of morphologically distinct stages from hyperplasia to invasive carcinoma. Malignant lung cancers are divided into small cell and non-small cell groups. The non small cell carcinoma group accounts for about 70% of all lung cancer cases and includes adenocarcinomas, squamous cell carcinomas, and large cell carcinomas. Adenocarcinomas typically arise in the peripheral airways and often form mucin secreting glands. Squamous cell carcinomas typically arise in proximal airways in apparent response to chronic inflammation and injury of the bronchial epithelium. The small cell carcinomas account for about 20% of lung cancers, typically arise in proximal airways, and inappropriately produce adrenocorticotropin and anti-diuretic hormone.
Lung cancer cells accumulate numerous genetic lesions, many of which are associated with cytologically visible chromosomal aberrations. The high frequency of chromosomal deletions associated with lung cancer may reflect the roles of multiple tumor suppressor loci in the etiology of this disease. Deletion of the short arm of chromosome 3 is found in over 90% of cases and represents one of the earliest genetic lesions leading to lung cancer. Deletions at chromosome arms 9p and 17p are also common. Other frequently observed genetic lesions include overexpression of telomerase, activation of oncogenes such as K-ras and c-myc, and inactivation of tumor suppressor genes such as retinoblastoma, p53 and cyclin-dependent kinase inhibitors such as CDKN2.
Genes differentially regulated in lung cancer have been identified by a variety of methods. Using mRNA differential display technology, Manda et al. (1999, Genomics 51:5-14) identified five genes differentially expressed in lung cancer cell lines compared to normal bronchial epithelial cells. Among the known genes, pulmonary surfactant apoprotein A and alpha 2 macroglobulin were down- regulated, and nm23Hl was upregulated. Petersen et al. (2000, Int J Cancer 86:512-517) used suppression subtractive hybridization to identify 552 clones differentially expressed in lung tumor derived cell lines; 205 of these clones represented known genes. Among the known genes, thrombospondin-1, fϊbronectin, intercellular adhesion molecule 1, and cytokeratins 6 and 18 had been observed previously to be differentially expressed in lung cancers. Wang et al. (2000, Oncogene 19:1519-1528) used a combination of microarray analysis and subtractive hybridization to identify 17 genes differentially overexpresssed in squamous cell carcinoma compared with normal lung epithelium. Keratin isoform 6, connexin 26, plakofillin 1 and cytokeratin 13 were identified among the known genes. Array technologies provide a simple way to explore the expression profile of a large number of related or unrelated genes. When an expression profile is examined, arrays provide a platform for examining which genes are tissue-specific, carrying out housekeeping functions, parts of a signaling cascade, or specifically related to a particular genetic predisposition, condition, disease, or disorder. The potential application of gene expression profiling is particularly relevant to improving diagnosis, prognosis, and treatment of disease.
The present invention satisfies a need in the art by providing a plurality of cDNAs, proteins or antibodies that can be used to diagnose, to stage, to treat, or to monitor the progression or treatment of a subject with a respiratory disorder and in particular, lung cancer.
SUMMARY The present invention provides a combination comprising a plurality of cDNAs wherein the cDNAs are SEQ ID NOs: 1-170 as presented in the Sequence Listing that are differentially expressed in lung cancers and the complements of SEQ ID NOs: 1-170. The invention also provides a combination comprising a plurality of cDNAs wherein the cDNAs are SEQ ID NOs: 1-37 that are differentially expressed at least 16-fold in lung cancers and the complements of SEQ ID NOs: 1-37. The invention further provides a combination comprising a plurality of cDNAs wherein the cDNAs are SEQ ID NOs:38-170 that are differentially expressed at least 4-fold in lung cancers and the complements of SEQ ID NOs: 38- 170. In one aspect, the combination is useful to diagnose a respiratory disorder such as lung cancer. In another aspect, the combination is immobilized on a substrate. The invention also provides a high throughput method to detect differential expression of one or more of the cDNAs of the combination. The method comprises hybridizing the substrate comprising the combination with the nucleic acids of a sample, thereby forming one or more hybridization complexes, detecting the hybridization complexes, and comparing the hybridization complexes with those of a standard, wherein differences in the size and signal intensity of each 5 hybridization complex indicates differential expression of nucleic acids in the sample. In one aspect, the nucleic acids of the sample are amplified prior to hybridization, hi another aspect, the sample is from a subject with lung cancer and differential expression identifies the type of lung cancer or stages the lung cancer.
The invention provides a high throughput method of screening a library or a plurality of
10 molecules or compounds to identify a ligand. The method comprises combining the substrate comprising the combination with a library or a plurality of molecules or compounds under conditions to allow specific binding and detecting specific binding, thereby identifying a ligand. The library or plurality of molecules or compounds are selected from DNA molecules, enhancers, mimetics, peptide nucleic acids, proteins, repressors, regulatory proteins, RNA molecules, and transcription factors.
15 The invention also provides a method for purifying a ligand, the method comprising combining a cDNA of the invention with a sample under conditions which allow specific binding, recovering the bound cDNA, and separating the cDNA from the ligand, thereby obtaining purified ligand.
The invention provides an isolated cDNA comprising a nucleic acid selected from SEQ ID NOs:2, 3, 32, 34, 35, 39, 43, 44, 45, 80, 81, 82, 139, 140, 146, 147, 148, 149, 151, 152, 154, 155, 157,
20 158, 160, 161, 162, and 164 as presented in the Sequence Listing. The invention also provides a vector comprising the cDNA, a host cell comprising the vector, and a method for producing a protein comprising culturing the host cell under conditions for the expression of a protein and recovering the protein from the host cell culture.
The present invention also provides a combination of purified proteins comprising the amino
25 acid sequences of SEQ ID NOs: 171-296 that are differentially expressed in lung cancer. The combination can be used in assays with standards to diagnose lung cancer. The invention also provides a purified protein comprising the amino acid sequence of SEQ ID NO: 174 encoded by a cDNA of the invention. The invention also provides a high-throughput method for using a protein to screen a library or a plurality of molecules or compounds to identify a ligand. The method comprises
30 combining the protein or a portion thereof with the library or plurality of molecules or compounds under conditions to allow specific binding and detecting specific binding, thereby identifying a ligand which specifically binds the protein. The library or plurality of molecules or compounds is selected from agonists, antagonists, antibodies, DNA molecules, small molecule drugs, immunoglobulins, inhibitors, mimetics, peptide nucleic acids, peptides, pharmaceutical agents, proteins, RNA
35 molecules, and ribozymes. In one aspect, an antibody which specifically binds a protein of the invention is selected from a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a recombinant antibody, a humanized antibody, single chain antibodies, a Fab fragment, an F(ab'>2 fragment, an Fv fragment; and an antibody-peptide fusion protein. The invention further provides a method for using a protein to purify a ligand. The method comprises combining the protein or a portion thereof with a sample under conditions to allow specific binding, recovering the bound protein, and separating the protein from the ligand, thereby obtaining purified ligand. The invention still further provides method for using the protein to produce a polyclonal or a monoclonal antibody. The method for producing a polyclonal antibody comprises immunizing a animal with a protein under conditions to elicit an antibody response, isolating animal antibodies, attaching the protein to a substrate, contacting the substrate with isolated antibodies under conditions to allow specific binding to the protein, dissociating the antibodies from the protein, thereby obtaining purified polyclonal antibodies. The method for preparing a monoclonal antibody comprises immunizing a animal with a protein under conditions to elicit an antibody response, isolating antibody producing cells from the animal, fusing the antibody producing cells with immortalized cells in culture to form monoclonal antibody producing hybridoma cells, culturing the hybridoma cells, and isolating a monoclonal antibody from culture. The method comprises immunizing an animal with the protein or an antigenic determinant thereof under conditions to elicit an antibody response, isolating animal antibodies, and screening the isolated antibodies with the protein to identify an antibody which specifically binds the protein. The invention yet still further provides a method for using the protein to purify antibodies which bind specifically to the protein.
The invention provides a purified antibody which specifically binds a protein of the invention. The invention also provides a method of using an antibody to detect the expression of a protein in a sample, the method comprising contacting the antibody with a sample under conditions for the formation of an antibody:protein complex and detecting complex formation wherein the formation of the complex indicates the expression of the protein in the sample. In one aspect, complex formation is compared to standards and is diagnostic of a respiratory disorder such as lung cancer. The invention further provides using an antibody to immunopurify a protein comprising combining the antibody with a sample under conditions to allow formation of an antibody:protein complex, and separating the antibody from the protein, thereby obtaining purified protein. The invention provides a composition comprising a cDNA, a protein, an antibody, or a ligand which has agonistic or antagonistic activity.
DESCRIPTION OF THE SEQUENCE LISTING AND TABLES A portion of the disclosure of this patent document contains material that is subject to copyright protection. The copyright owner has no objection to the facsimile reproduction by anyone of the patent document or the patent disclosure, as it appears in the Patent and Trademark Office patent file or records, but otherwise reserves all copyright rights whatsoever.
The Sequence Listing provides SEQ ID NOs: 1-170, exemplary polynucleotides of the invention. The Sequence Listing also provides SEQ ID NOs: 171-296, exemplary proteins of the invention. Each sequence is identified by a sequence identification number (SEQ ID NO) and by INCYTE ID.
Figures 1 A-4A show the amino acid sequence of SEQ ID NO: 174 and a nucleic acid sequence of its encoding cDNA (SEQ ID NO:43). The alignment was produced using MACDNASIS PRO software (Hitachi Software Engineering, South San Francisco CA).
Table 1 lists the cDNAs whose transcripts are differentially up-regulated by at least 16-fold in lung tumors as determined by microarray analysis. The column descriptions for Tables 1-4 are: Column 1 shows SEQ ID NO: (SEQ ID ); column 2, Incyte ID; column 3, the number of samples showing greater than 16-fold (Tables 1-2) or 4-fold (Tables 3-4) expression levels; column 4, the closest GenBank homolog; column 5, the E-value for the comparison with the GenBank homolog; and column 6, the description of the GenBank homolog. Table 2 lists the cDNAs whose transcripts are differentially down-regulated by at least 16- fold in lung tumors as determined by microarray analysis.
Table 3 lists-the cDNAs whose transcripts are differentially up-regulated by at least 4-fold and less than 16-fold in lung tumors as determined by microarray analysis.
Table 4 lists the cDNAs whose transcripts are differentially down-regulated by at least 4-fold and less than 16-fold in lung tumors as determined by microarray analysis.
Table 5 lists the proteins comprising the amino acid sequences of SEQ ID NOs: 171-296 as shown in the Sequence Listing by SEQ ID NO (columns 1, 3, and 5) and Incyte ID (columns 2, 4, and
6).
DESCRIPTION OF THE INVENTION Definitions
"Antibody" refers to intact immunoglobulin molecule, a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a recombinant antibody, a humanized antibody, single chain antibodies, a Fab fragment, an F(ab')2 fragment, an Fv fragment; and an antibody-peptide fusion protein. "Antigenic determinant" refers to an antigenic or immunogenic epitope, structural feature, or region of an oligopeptide, peptide, or protein which is capable of inducing formation of an antibody which specifically binds the protein. Biological activity is not a prerequisite for immunogenicity.
"Array" refers to an ordered arrangement of at least two cDNAs, proteins, or antibodies on a substrate. At least one of the cDNAs, proteins, or antibodies represents a control or standard, and the other cDNA, protein, or antibody is of diagnostic or therapeutic interest. The arrangement of at least two and up to about 40,000 cDNAs, proteins, or antibodies on the substrate assures that the size and signal intensity of each labeled complex, formed between each cDNA and at least one nucleic acid, each protein and at least one ligand or antibody, or each antibody and at least one protein to which the antibody specifically binds, is individually distinguishable. A "combination" refers to at least two cDNAs, and up to 170 cDNAs selected from SEQ ID
NOs: 1-170 and the complete complements of SEQ ID NOs: 1-170, or at least two proteins, and up to 125 proteins selected from SEQ ID NOs: 171-296.
The "complement" of a cDNA of the Sequence Listing refers to a nucleic acid molecule which is completely complementary over its full length and which will hybridize to a nucleic acid molecule under conditions of high stringency.
"cDNA" refers to an isolated polynucleotide, nucleic acid, or a fragment thereof, that contains from about 400 to about 12,000 nucleotides. It may have originated recombinantly or synthetically, may be double-stranded or single-stranded, represents coding and noncoding 3 ' or 5' sequence, generally lacks introns and may be purified or combined with carbohydrate, lipids, protein or inorganic elements or substances.
The phrase "cDNA encoding a protein" refers to a nucleic acid whose sequence closely aligns with sequences that encode conserved regions, motifs or domains identified by employing analyses well known in the art. These analyses include BLAST (Altschul (1993) J Mol Evol 36:290-300; Altschul et al. (1990) J Mol Biol 215:403-410; and BLAST2 (Altschul (1997) supra) which provide identity within the conserved region. Brenner et al. (1998, Proc Natl Acad Sci 95:6073-6078) who analyzed BLAST for its ability to identify structural homologs by sequence identity found 30% identity is a reliable threshold for sequence alignments of at least 150 residues and 40% is a reasonable threshold for alignments of at least 70 residues (Brenner, page 6076, column 2). A "composition" refers to the polynucleotide and a vector or a labeling moiety; a purified protein and a pharmaceutical carrier or a heterologous, labeling, or purification moiety; an antibody and a labeling moiety or pharmaceutical agent; and the like.
"Differential expression" refers to an increased, upregulated or present, or decreased, downregulated or absent, gene expression as detected by the absence, presence, or at least two-fold change in the amount of transcribed messenger RNA or translated protein in a sample.
"Disorder" refers to conditions, diseases or syndromes of the respiratory system including, but not limited to, lung cancer, particularly adenocarcinomas and squamous cell carcinomas; chronic obstructive pulmonary disease, emphysema, or asthma.
An "expression profile" is a representation of gene expression in a sample. A nucleic acid expression profile is produced using sequencing, hybridization, or amplification technologies and „ ,_ _ __ ^ | mRNAs or cDNAs from a sample. A protein expression profile, although time delayed, mirrors the nucleic acid expression profile and uses two-dimensional polyacrylamide electrophoresis (2D-PAGE, mass spectrophotometry (MS), enzyme-linked immunosorbent assays (ELISAs), radioimmunoassays (RIAs), and fluorescence activated cell sorting (FACS) or arrays and labeling moieties or antibodies to detect expression in a sample. The nucleic acids, proteins, or antibodies may be used in solution or attached to a substrate, and their detection is based on methods and labeling moieties well known in the art.
A "hybridization complex" is formed between a polynucleotide of the invention and a nucleic acid of a sample when the purines of one molecule hydrogen bond with the pyrimidines of the complementary molecule, e.g., 5'-A-G-T-C-3'base pairs with its complete complement, 3 -T-C-A-G- 5'. The degree of complementarity and the use of nucleotide analogs affect the efficiency and stringency of hybridization reactions.
"Identity" as applied to sequences, refers to the quantification (usually percentage) of nucleotide or residue matches between at least two sequences aligned using a standardized algorithm such as Smith-Waterman alignment (Smith and Waterman (1981) J Mol Biol 147:195-197), CLUSTALW (Thompson et al- (1994) Nucleic Acids Res 22:4673-4680), or BLAST2 (Altschul (1997) supra). BLAST2 may be used in a standardized and reproducible way to insert gaps in one of the sequences in order to optimize alignment and to achieve a more meaningful comparison between them. "Similarity" as applied to proteins uses the same algorithms but takes into account conservative substitutions of nucleotides or residues.
"Isolated" or "purified" refers to any molecule or compound that is separated from its natural environment and is from about 60% free to about 90% free from other components with which it is naturally associated.
"Labeling moiety" refers to any reporter molecule whether a visible or radioactive label, stain or dye that can be attached to or incorporated into a cDNA or protein. Visible labels and dyes include but are not limited to anthocyanins, β glucuronidase, BIODIPY, Coomassie blue, Cy3 and Cy5, digoxigenin, FITC, green fluorescent protein, luciferase, spyro red, silver, and the like. Radioactive markers include radioactive forms of hydrogen, iodine, phosphorous, sulfur, and the like. "Ligand" refers to any agent, molecule, or compound which will bind specifically to a complementary site on a cDNA molecule or polynucleotide, or to an epitope or a protein. Such ligands stabilize or modulate the activity of polynucleotides or proteins and may be composed of inorganic or organic substances including nucleic acids, proteins, carbohydrates, fats, and lipids.
"Oligonucleotide" refers a single stranded molecule from about 18 to about 60 nucleotides in length which may be used in hybridization or amplification technologies or in regulation of replication, transcription or translation. Equivalent terms are amplimer, primer, and oligomer. "Portion" refers to any part of a protein used for any purpose which retains at least one biological or antigenic characteristic of a native protein, but especially, to an epitope for the screening of ligands or for the production of antibodies.
"Post-translational modification" of a protein can involve lipidation, glycosylation, phosphorylation, acetylation, racemization, proteolytic cleavage, and the like. These processes may occur synthetically or biochemically. Biochemical modifications will vary by cellular location, cell type, pH, enzymatic milieu, and the like.
"Probe" refers to a cDNA that hybridizes to at least one nucleic acid molecule in a sample. Probes can be labeled for use in hybridization reactions including Southern, northern, in situ, dot blot, array, and like technologies or in screening assays.
"Protein" refers to a polypeptide or peptide comprising an amino acid sequence selected from SEQ ID NOs: 171-296. An "oligopeptide" is an amino acid sequence from about five residues to about 15 residues that is used as part of a fusion protein to produce an antibody.
"Sample" is used in its broadest sense as containing nucleic acids, proteins, antibodies, and the like. A sample may comprise a bodily fluid such as ascites, blood, lymph, semen, spinal fluid, sputum, tears, urine, and the like; the soluble fraction of a cell preparation, or an aliquot of media in which cells were grown; a chromosome, an organelle, or membrane isolated or extracted from a cell; genomic DNA, RNA, or cDNA in solution or bound to a substrate; a cell; a tissue, tissue biopsy or tissue print; buccal cells, skin, a hair or its follicle; and the like. "Specific binding" refers to a special and precise interaction between two molecules which is dependent upon their structure, particularly their molecular side groups. Examples include the intercalation of a regulatory protein into the major groove of a DNA molecule, the hydrogen bonding along the backbone between two single stranded nucleic acids, or the binding between an epitope of a protein and an agonist, antagonist, or antibody. "Substrate" refers to any rigid or semi-rigid support to which cDNAs or proteins are bound and includes membranes, filters, chips, slides, wafers, fibers, magnetic or nonmagnetic beads, gels, capillaries or other tubing, plates, polymers, and microparticles with a variety of surface forms including wells, trenches, pins, channels and pores.
A "transcript image" (TI) is a profile of gene transcription activity in a particular tissue at a particular time. TI provides assessment of the relative abundance of expressed polynucleotides in the cDNA libraries of an EST database as described in USPN 5,840,484, incorporated herein by reference.
"Variant" refers to molecules that are recognized variations of a cDNA or a protein encoded by the cDNA. Splice variants may be determined by BLAST score, wherein the score is at least 100, and most preferably at least 400. Allelic variants have high percent identity to the cDNAs of the invention and may differ by about three bases per hundred bases. "Single nucleotide polymorphism" (SNP) refers to a change in a single base as a result of a substitution, insertion or deletion. The change may be conservative (purine for purine) or non-conservative (purine to pyrimidine) and may or may not result in a change in an encoded amino acid. The Invention
The present invention provides a combination comprising a plurality of cDNAs, wherein the cDNAs are SEQ ID NOs: 1-170 of the Sequence Listing and the complete complements of SEQ ID NOs: 1-170, that are differentially expressed in lung cancers. The invention also provides a combination comprising a plurality of proteins, wherein the proteins are SEQ ID NOs: 171-296 of the Sequence Listing, that are differentially expressed in lung cancers. These compositions when immobilized on an array can be used in a clinically relevant manner with subject tissues to detect changes in expression associated with respiratory disorders, particularly adenocarcinomas, neuroendocrine carcinoids, and squamous cell carcinomas of the lung, chronic obstructive pulmonary disease, emphysema, and asthma. The invention further provides a subcombinations comprising a plurality of cDNAs, wherein the cDNAs are SEQ ID NOs: 1-18 and the complete complements of SEQ ID NOs: 1-18 that are up- regulated by at least 16-fold in lung cancers. The invention still further provides a subcombination comprising a plurality of cDNAs, wherein the cDNAs are SEQ ID NOs: 19-37 and the complete complements of SEQ ID NOs: 19-37 that are down-regulated by at least 16-fold in lung cancers. The invention further provides a subcombination comprising a plurality of cDNAs, wherein the cDNAs are SEQ ID NOs:38-83 and the complete complements of SEQ ID NOs:38-83 that are up-regulated between 4- and 16-fold in lung cancers. The invention still further provides a subcombination comprising a plurality of cDNAs, wherein the cDNAs are SEQ ID NOs: 84-170 and the complete complements of SEQ ID NOs:84-170 that are down-regulated between 4- and 16-fold in lung cancers. The invention yet further provides cDNAs comprising the nucleic acid sequence of SEQ ID
NOs:2, 3, 32, 34, 35, 39, 43, 44, 45, 80, 81, 82, 139, 140, 146, 147, 148, 149, 151, 152, 154, 155, 157, 158, 160, 161, 162, and 164 that are novel cDNAs and are known only by their differential expression in lung cancer. The usefulness of both the combinations and the novel cDNAs rests on their immediate value as diagnostics for respiratory disorders such as lung cancer. The invention still yet further provides a purified protein comprising the amino acid sequence of SEQ ID NO: 174. The novel protein is 285 amino acids in length and identical, from residue M52 to residue N285, to human thymidine kinase (g339719) as determined using BLAST. SEQ ID NO: 174 has one potential cAMP- protein kinase phosphorylation site at residue K242; three potential casein kinase II phosphorylation sites at residues S27, T37, and T219; six potential protein kinase C phosphorylation sites at residues S2, S27, S41, S81, T214, and T219; two potential tyrosine kinase phosphorylation site at residues Y34 and Yl 12; two potential ATP/GTP-binding site motif A (P-loop) at residues S84 and T169; and one thymidine kinase cellular-type signature from residues G227 to Y240. SEQ ID NO: 174 also contains a thymidine kinase domain from residues G70 to Y240 as determined by searching for statistically significant matches in the hidden Markov model (HMM)- based PFAM database of conserved protein family domains. Data from BLIMPS, PROFILESCAN, and MOTIFS provide further evidence that SEQ ID NO: 174 is a member of the thymidine kinase family.
Figures 1A-4A show the amino acid sequence of SEQ ID NO: 174 and a nucleic acid sequence of its encoding cDNA (SEQ ID NO:43). The alignment was produced using MACDNASIS PRO software (Hitachi Software Engineering, South San Francisco CA).
Table 1 shows those cDNAs of the present invention having the nucleic acid sequences of SEQ ID NOs: 1-18 that showed at least 16-fold up-regulation in lung tumors. Columns 1 and 2 show the SEQ ID NO and Incyte ID, respectively. Column 3 shows the number of tumors in which transcripts are up-regulated by at least 16-fold. Columns 4 and 6 show the GenBank identification number and description of the nearest GenBank homolog identified using BLAST2 analysis with default parameters. Column 5 shows the probability scores for the matches between each cDNA and its homolog.
Table 2 shows those cDNAs of the present invention having the nucleic acid sequences of SEQ ID NOs: 19-37 that showed at least 16-fold down-regulation in lung tumors. Columns 1 and 2 show the SEQ ID NO and Incyte ID, respectively. Column 3 shows the number of tumors in which transcripts are down-regulated by at least 16-fold. Columns 4 and 6 show the GenBank identification number and description of the nearest GenBank homolog identified using BLAST2 analysis with default parameters. Column 5 shows the probability scores for the matches between each cDNA and its homolog. Table 3 shows those cDNAs of the present invention having the nucleic acid sequences of
SEQ ID NOs:38-83 that showed between 4- to 16-fold up-regulation in lung tumors. Columns 1 and 2 show the SEQ ID NO and Incyte ID, respectively. Column 3 shows the number of tumors in which transcripts are up-regulated between 4- and 16-fold. Columns 4 and 6 show the GenBank identification number and description of the nearest GenBank homolog identified using BLAST2 analysis with default parameters. Column 5 shows the probability scores for the matches between each cDNA and its homolog.
Table 4 shows those cDNAs of the present invention having the nucleic acid sequences of SEQ ID NOs:84-170 that showed between 4- to 16-fold down-regulation in lung tumors. Table 4 lists the differential expression of clones representing cDNAs that are down-regulated by at least 4-fold and less than 16-fold on microarrays. Columns 1 and 2 show the SEQ ID NO and Incyte ID, respectively. Column 3 shows the number of tumors in which transcripts are down-regulated between 4- and 16-fold. Columns 4 and 6 show the GenBank identification number and description of the nearest GenBank homolog identified using BLAST2 analysis with default parameters. Column 5 shows the probability scores for the matches between each cDNA and its homolog. Table 5 lists the proteins comprising the amino acid sequences of SEQ ID NOs: 171-296 as shown in the Sequence Listing by SEQ ID NO (columns 1, 3, and 5) and Incyte ID (columns 2, 4, and 6).
The cDNAs of the invention define a differential expression pattern against which to compare the expression profile of biopsied, in vitro or in vivo treated lung samples. The proteins may be arranged on an array and used to screen for therapeutics. The combination of nucleotides or antibodies which specifically bind the proteins may be arrayed and contacted with samples from subjects with undiagnosed respiratory disorders. Expression profiles will provide information useful for identifying and distinguishing among different disorders, or stages of those disorders, identifying those sequences of highest diagnostic and potential therapeutic value for each disorder, or evaluating patient treatment. Such nucleotide, protein and antibody combinations may be useful in the elucidation of pathways which are affected in a particular disorder or to identify new, coexpressed, candidate, therapeutic molecules.
Samples for these assays are prepared by methods well known in the art and are derived from mammalian cells or tissues which are in a certain stage of development; have been treated with a known molecule or compound, such as a cytokine, growth factor, drug, or the like; or have been extracted or biopsied from a mammal with a known or unknown condition, disorder, or disease before or after treatment. The sample nucleic acids or protein are contacted with the cDNA or antibody array for the purpose of defining a novel gene profile associated with that developmental stage, treatment, or disorder. cDNAs and Their Uses cDNAs can be prepared by a variety of synthetic or enzymatic methods well known in the art. cDNAs can be synthesized, in whole or in part, using chemical methods well known in the art (Caruthers et al. (1980) Nucleic Acids Symp Ser (7) 215-233). Alternatively, cDNAs can be produced enzymatically or recombinantly, by in vitro or m vivo transcription. Nucleotide analogs can be incorporated into cDNAs by methods well known in the art. The only requirement is that the incorporated analog must base pair with native purines or pyrimidines. For example, 2, 6-diaminopurine can substitute for adenine and form stronger bonds with thymidine than those between adenine and thymidine. A weaker pair is formed when hypoxanthine is substituted for guanine and base pairs with cytosine. Additionally, cDNAs can include nucleotides that have been derivatized chemically or enzymatically. cDNAs can be synthesized on a substrate. Synthesis on the surface of a substrate may be accomplished using a chemical coupling procedure and a piezoelectric printing apparatus as described by Baldeschweiler et al- (PCT publication WO95/251116). Alternatively, the cDNAs can be synthesized on a substrate surface using a self-addressable electronic device that controls when reagents are added as described by Heller et al. (USPN 5,605,662). cDNAs can be synthesized directly on a substrate by sequentially dispensing reagents for their synthesis on the substrate surface or by dispensing preformed DNA fragments to the substrate surface. Typical dispensers include a micropipette delivering solution to the substrate with a robotic system to control the position of the micropipette with respect to the substrate. There can be a multiplicity of dispensers so that reagents can be delivered to the reaction regions efficiently. cDNAs can be immobilized on a substrate by covalent means such as by chemical bonding procedures or UV irradiation. In one method, a cDNA is bound to a glass surface which has been modified to contain epoxide or aldehyde groups. In another method, a cDNA is placed on a polylysine coated surface and UV cross-linked to it as described by Shalon et al. (WO95/35505). In yet another method, a cDNA is actively transported from a solution to a given position on a substrate by electrical means (Heller, supra). cDNAs do not have to be directly bound to the substrate, but rather can be bound to the substrate through a linker group. The linker groups are typically about 6 to 50 atoms long to provide exposure of the attached cDNA. Preferred linker groups include ethylene glycol oligomers, diamines, diacids and the like. Reactive groups on the substrate surface react with a terminal group of the linker to bind the linker to the substrate. The other terminus of the linker is then bound to the cDNA. Alternatively, polynucleotides, plasmids or cells can be arranged on a filter. In the latter case, cells are lysed, proteins and cellular components degraded, and the DNA is coupled to the filter by UV cross-linking.
The cDNAs may be used for a variety of purposes. For example, the combination of the invention may be used on an array. The array, in turn, can be used in high-throughput methods for detecting a related polynucleotide in a sample, screening a plurality of molecules or compounds to identify a ligand, diagnosing a respiratory disorder such as chronic obstructive pulmonary disease or lung cancer, or inhibiting or inactivating a therapeutically relevant gene related to the cDNA.
When the cDNAs of the invention are employed on a microarray, the cDNAs are arranged in an ordered fashion so that each cDNA is present at a specified location. Because the cDNAs are at specified locations on the substrate, the hybridization patterns and intensities, which together create a unique expression profile, can be interpreted in terms of expression levels of particular genes and can be correlated with a particular metabolic process, condition, disorder, disease, stage of disease, or treatment. Hybridization The cDNAs or fragments or complements thereof may be used in various hybridization technologies. The cDNAs may be labeled using a variety of reporter molecules by either PCR , recombinant, or enzymatic techniques. For example, a commercially available vector containing the cDNA is transcribed in the presence of an appropriate polymerase, such as T7 or SP6 polymerase, and at least one labeled nucleotide. Commercial kits are available for labeling and cleanup of such cDNAs. Radioactive (Amersham Biosciences (APB), Piscataway NJ), fluorescent (Qiagen-Operon, Alameda CA), and chemiluminescent labeling (Promega, Madison WI) are well known in the art.
A cDNA may represent the complete coding region of an mRNA or be designed or derived from unique regions of the mRNA or genomic molecule, an intron, a 3' untranslated region, or from a conserved motif. The cDNA is at least 18 contiguous nucleotides in length and is usually single stranded. Such a cDNA may be used under hybridization conditions that allow binding only to an identical sequence, a naturally occurring molecule encoding the same protein, or an allelic variant. Discovery of related human and mammalian sequences may also be accomplished using a pool of degenerate cDNAs and appropriate hybridization conditions. Generally, a cDNA for use in Southern or northern hybridizations may be from about 400 to about 6000 nucleotides long. Such cDNAs have high binding specificity in solution-based or substrate-based hybridizations. An oligonucleotide, a fragment of the cDNA, may be used to detect a polynucleotide in a sample using PCR.
The stringency of hybridization is determined by G+C content of the cDNA, salt concentration, and temperature. In particular, stringency is increased by reducing the concentration of salt or raising the hybridization temperature. In solutions used for some membrane based hybridizations, addition of an organic solvent such as formamide allows the reaction to occur at a lower temperature. Hybridization may be performed with buffers, such as 5x saline sodium citrate (SSC) with 1% sodium dodecyl sulfate (SDS) at 60°C, that permit the formation of a hybridization complex between nucleic acid sequences that contain some mismatches. Subsequent washes are performed with buffers such as 0.2xSSC with 0.1% SDS at either 45°C (medium stringency) or 65°C- 68°C (high stringency). At high stringency, hybridization complexes will remain stable only where the nucleic acids are completely complementary. In some membrane-based hybridizations, preferably 35% or most preferably 50%, formamide may be added to the hybridization solution to reduce the temperature at which hybridization is performed. Background signals may be reduced by the use of detergents such as Sarkosyl or TRITON X-100 (Sigma-Aldrich, St. Louis MO) and a blocking agent such as denatured salmon sperm DNA. Selection of components and conditions for hybridization are well known to those skilled in the art and are reviewed in Ausubel et al. (1997, Short Protocols in Molecular Biology. John Wiley & Sons, New York NY, Units 2.8-2.11, 3.18-3.19 and 4-6-4.9). Dot-blot, slot-blot, low density and high density arrays are prepared and analyzed using methods known in the art. cDNAs from about 18 consecutive nucleotides to about 6000 consecutive nucleotides in length are contemplated by the invention and used in array technologies. The number of cDNAs on a substrate is at least about five and can be up to about 100,000. The array may be used to monitor the expression level of large numbers of genes simultaneously and to identify genetic variants, mutations, and SNPs. Such information may be used to determine gene function; to 5 understand the genetic basis of a disorder; to diagnose a disorder; and to develop and monitor the activities of therapeutic agents being used to control or cure a disorder. (See, e.g., USPN 5,474,796; WO95/11995; WO95/35505; USPN 5,605,662; and USPN 5,958,342.) Screening and Purification Assays
A cDNA may be used to screen a library or a plurality of molecules or compounds for a
L0 ligand which specifically binds the cDNA. Ligands may be DNA molecules, RNA molecules, peptide nucleic acid molecules, peptides, proteins such as transcription factors, promoters, enhancers, repressors, and other proteins that regulate replication, transcription, or translation of the polynucleotide in the biological system. The assay involves combining the cDNA or a fragment thereof with the molecules or compounds under conditions that allow specific binding and detecting 5 the bound cDNA to identify at least one ligand that specifically binds the cDNA.
In one embodiment, the cDNA may be incubated with a library of isolated and purified molecules or compounds and binding activity determined by methods such as a gel-retardation assay (USPN 6,010,849) or a reticulocyte lysate transcriptional assay. In another embodiment, the cDNA may be incubated with nuclear extracts from biopsied and/or cultured cells and tissues. Specific 0 binding between the cDNA and a molecule or compound in the nuclear extract is initially determined by gel shift assay. Protein binding may be confirmed by raising antibodies against the protein and adding the antibodies to the gel-retardation assay where specific binding will cause a supershift in the assay.
In another embodiment, the cDNA may be used to purify a molecule or compound using 5 affinity chromatography methods well known in the art. In one embodiment, the cDNA is chemically reacted with cyanogen bromide groups on a polymeric resin or gel. Then a sample is passed over and reacts with or binds to the cDNA. The molecule or compound which is bound to the cDNA may be released from the cDNA by increasing the salt concentration of the flow-through medium and collected. 0 The cDNA may be used to purify a ligand from a sample. A method for using a cDNA to purify a ligand would involve combining the cDNA or a fragment thereof with a sample under conditions to allow specific binding, recovering the bound cDNA, and using an appropriate agent to separate the cDNA from the purified ligand. Protein Production and Uses 5 The cDNAs or fragment thereof may be used to produce purified proteins using recombinant DNA technologies described herein and taught in Ausubel (supra; Units 16.1-16.62). One of the advantages of producing proteins by these procedures is the ability to obtain highly-enriched sources of the proteins thereby simplifying purification procedures.
The proteins may contain amino acid substitutions, deletions or insertions made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity, and/or the amphipathic nature of the residues involved. Such substitutions may be conservative in nature when the substituted residue has structural or chemical properties similar to the original residue (e.g., replacement of leucine with isoleucine or valine) or they may be nonconservative when the replacement residue is radically different (e.g., a glycine replaced by a tryptophan). Computer programs included in LASERGENE software (DNASTAR, Madison WI) and algorithms included in RasMol software (University of Massachusetts, Amherst MA) may be used to determine reading frame and which and how many amino acid residues in a particular portion of the protein may be substituted, inserted, or deleted without abolishing biological or immunological activity. Expression of Encoded Proteins Expression of a particular cDNA may be accomplished by cloning the cDNA into a vector and transforming this vector into a host cell. The cloning vector used for the construction of cDNA libraries in the LIEESEQ databases (Incyte Genomics, Palo Alto CA) may also be used for expression. Such vectors usually contain a promoter and a polylinker useful for cloning, priming, and transcription. An exemplary vector may also contain the promoter for β-galactosidase, an amino-terminal methionine and the subsequent seven amino acid residues of β-galactosidase. The vector may be transformed into competent E. coli cells. Induction of the isolated bacterial strain with isopropylthiogalactoside (IPTG) using standard methods will produce a fusion protein that contains an N terminal methionine, the first seven residues of β-galactosidase, about 15 residues of linker, and the protein encoded by the cDNA. The cDNA may be shuttled into other vectors known to be useful for expression of protein in specific hosts. Oligonucleotides containing cloning sites and fragments of DNA sufficient to hybridize to stretches at both ends of the cDNA may be chemically synthesized by standard methods. These primers may then be used to amplify the desired fragments by PCR. The fragments may be digested with appropriate restriction enzymes under standard conditions and isolated using gel electrophoresis. Alternatively, similar fragments are produced by digestion of the cDNA with appropriate restriction enzymes and filled in with chemically synthesized oligonucleotides. Fragments of the coding sequence from more than one gene may be ligated together and expressed.
Signal sequences that dictate secretion of soluble proteins are particularly desirable as component parts of a recombinant sequence. For example, a chimeric protein may be expressed that includes one or more additional purification-facilitating domains. Such domains include, but are not limited to, metal-chelating domains that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (I munex, Seattle WA). The inclusion of a cleavable-linker sequence such as ENTEROKESfASEMAX (Invitrogen, San Diego CA) between the protein and the purification domain may also be used to recover the protein.
Suitable host cells may include, but are not limited to, mammalian cells such as Chinese Hamster Ovary (CHO) and human 293 cells, insect cells such as Sf9 cells, plant cells such as Nicotiana tabacum yeast cells such as Saccharomvces cerevisiae. and bacteria such as E. coli. For each of these cell systems, a useful vector may also include an origin of replication and one or two selectable markers to allow selection in bacteria as well as in a transformed eukaryotic host. Vectors for use in eukaryotic host cells may require the addition of 3'poly(A) tail if the cDNA lacks poly(A).
Additionally, the vector may contain promoters or enhancers that increase gene expression. Many promoters are known and used in the art. Most promoters are host specific and exemplary promoters includes SV40 promoters for CHO cells; T7 promoters for bacterial hosts; viral promoters and enhancers for plant cells; and PGH promoters for yeast. Adenoviral vectors with the rous sarcoma virus enhancer or retroviral vectors with long terminal repeat promoters may be used to drive protein expression in mammalian cell lines. Once homogeneous cultures of recombinant cells are obtained, large quantities of secreted soluble protein may be recovered from the conditioned medium and analyzed using chromatographic and electrophoretic methods well known in the art. An alternative method for the production of large amounts of secreted protein involves the transformation of mammalian embryos and the recovery of the recombinant protein from milk produced by transgenic cows, goats, sheep, and the like.
In addition to recombinant production, proteins or portions thereof may be produced manually, using solid-phase techniques (Stewart et al. (1969) Solid-Phase Peptide Synthesis. WH Freeman, San Francisco CA; Merrifield (1963) J Am Chem Soc 5:2149-2154), or using machines such as the 431 A peptide synthesizer (Applied Biosystems (ABI), Foster City CA). Proteins produced by any of the above methods may be used as pharmaceutical compositions to treat disorders associated with null or inadequate expression of the genomic sequence. Protein Identification Several techniques have been developed which permit rapid identification of proteins from complex samples using high performance liquid chromatography and mass spectrometry (MS). Beginning with a sample containing proteins, the major steps involved are: 1) proteins are separated using two-dimensional gel electrophoresis (2-DE), 2) selected proteins are excised from the gel and digested with a protease to produce a set of peptides; and 3) the peptides are subjected to mass spectral analysis to derive peptide ion mass and spectral pattern information. The MS information is used to identify the protein by comparing it with information in a protein database (Shevenko et al.(1996) Proc Natl Acad Sci 93: 14440-14445).
Proteins are separated by 2DE employing isoelectric focusing (IEF) in the first dimension followed by SDS-PAGE in the second dimension. For IEF, an immobilized pH gradient strip is useful to increase reproducibility and resolution of the separation. Alternative techniques may be used to improve resolution of very basic, hydrophobic, or high molecular weight proteins. The separated proteins are detected using a stain or dye such as silver stain, Coomassie blue, or spyro red (Molecular Probes, Eugene OR) that is compatible with MS. Gels may be blotted onto a PVDF membrane for western analysis and optically scanned using a STORM scanner (APB) to produce a computer-readable output which is analyzed by pattern recognition software such as MELANTE (GeneBio, Geneva, Switzerland). The software annotates individual spots by assigning a unique identifier and calculating their respective x,y coordinates, molecular masses, isoelectric points, and signal intensity. Individual spots of interest, such as those representing differentially expressed proteins, are excised and proteolytically digested with a site-specific protease such as trypsin or chymotrypsin, singly or in combination, to generate a set of small peptides, preferably in the range of 1-2 kDa. Prior to digestion, samples may be treated with reducing and alkylating agents, and following digestion, the peptides are then separated by liquid chromatography or capillary electrophoresis and analyzed using MS.
MS converts components of a sample into gaseous ions, separates the ions based on their mass-to-charge ratio, and determines relative abundance. For peptide mass fingerprinting analysis, a MALDI-TOF (Matrix Assisted Laser Desorption/Ionization-Time of Flight), ESI (Electrospray Ionization), and TOF-TOF (Time of Flight/Time of Flight) machines are used to determine a set of highly accurate peptide masses. Using analytical programs, such as TURBOSEQUEST software (Finnigan, San Jose CA), the MS data is compared against a database of theoretical MS data derived from known or predicted proteins. A minimum match of three peptide masses is usually required for reliable protein identification. If additional information is needed for identification, Tandem-MS may be used to derive information about individual peptides. In tandem-MS, a first stage of MS is performed to determine individual peptide masses. Then selected peptide ions are subjected to fragmentation using a technique such as collision induced dissociation to produce an ion series. The resulting fragmentation ions are analyzed in a second round of MS, and their spectral pattern may be used to determine a short stretch of amino acid sequence (Dancik et al. (1999) J Comput Biol 6:327- 342).
Assuming the protein is represented in the database, a combination of peptide mass and fragmentation data, together with the calculated MW and pi of the protein, will usually yield an unambiguous identification. If no match is found, protein sequence can be obtained using direct chemical sequencing procedures well known in the art (cf. Creighton (1984) Proteins. Structures and Molecular Properties. WH Freeman, New York NY). Chemical Synthesis of Peptides
Proteins or portions thereof may be produced not only by recombinant methods, but also by using chemical methods well known in the art. Solid phase peptide synthesis may be carried out in a batchwise or continuous flow process which sequentially adds α-amino- and side chain-protected amino acid residues to an insoluble polymeric support via a linker group. A linker group such as methylamine-derivatized polyethylene glycol is attached to poly(styrene-co-divinylbenzene) to form the support resin. The amino acid residues are N-α-protected by acid labile Boc (t-butyloxycarbonyl) or base-labile Fmoc (9-fluorenylmethoxycarbonyl). The carboxyl group of the protected amino acid is coupled to the amine of the linker group to anchor the residue to the solid phase support resin. Trifluoroacetic acid or piperidine are used to remove the protecting group in the case of Boc or Fmoc, respectively. Each additional amino acid is added to the anchored residue using a coupling agent or pre-activated amino acid derivative, and the resin is washed. The full length peptide is synthesized by sequential deprotection, coupling of derivitized amino acids, and washing with dichloromethane and/or N, N-dimethylformamide. The peptide is cleaved between the peptide carboxy terminus and the linker group to yield a peptide acid or amide. (Novabiochem 1997/98 Catalog and Peptide Synthesis Handbook, San Diego CA pp. S1-S20). Automated synthesis may also be carried out on machines such as the 431 A peptide synthesizer (ABI). A protein or portion thereof may be purified by preparative high performance liquid chromatography and its composition confirmed by amino acid analysis or by sequencing (Creighton (1984) Proteins. Structures and Molecular Properties, WH Freeman, New York NY). Screening. Purification, and Immunoassays
A protein or a portion thereof encoded by the cDNA may be used to screen a library or a plurality of molecules or compounds for a ligand with specific binding affinity or to purify a molecule or compound from a sample. The protein or portion thereof employed in such screening may be free in solution, affixed to an abiotic or biotic substrate, or located intracellularly. For example, viable or fixed prokaryotic host cells that are stably transformed with recombinant nucleic acids that have expressed and positioned a protein on their cell surface can be used in screening assays. The cells are screened against a library or a plurality of ligands and the specificity of binding or formation of complexes between the expressed protein and the ligand may be measured. The ligands can be agonists, antagonists, antibodies, DNA molecules, enhancers, small drug molecules, immunoglobulins, inhibitors, mimetics, peptide nucleic acid molecules, peptides, pharmaceutical agents, proteins, and regulatory proteins, repressors, RNA molecules, ribozymes, and transcription factors, or any other test molecule or compound that specifically binds the protein. An exemplary assay involves combining the mammalian protein or a portion thereof with the molecules or compounds under conditions that allow specific binding and detecting the bound protein to identify at least one ligand that specifically binds the protein.
This invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of binding the protein specifically compete with a test compound capable of binding to the protein or oligopeptide or fragment thereof. One method for high throughput screening using very small assay volumes and very small amounts of test compound is described in USPN 5,876,946. Molecules or compounds identified by screening may be used in a model system to evaluate their toxicity, diagnostic, or therapeutic potential. The protein may be used to purify a ligand from a sample. A method for using a protein to purify a ligand would involve combining the protein or a portion thereof with a sample under conditions to allow specific binding, recovering the bound protein, and using an appropriate chaotropic agent to separate the protein from the purified ligand.
Immunoassay procedures may be used to quantify expression of the protein in cell cultures, in subjects with a particular disorder or in model animal systems under various conditions. Increased or decreased production of proteins as monitored by immunoassay may contribute to knowledge of the cellular activities associated with developmental pathways, engineered conditions or diseases, or treatment efficacy. The quantity of a given protein in a given tissue may be determined by performing immunoassays on freeze-thawed detergent extracts of biological samples and comparing the slope of the binding curves to binding curves generated by purified protein. Antibody Production and Use
A protein encoded by a cDNA of the invention may be used to produce specific antibodies. Antibodies may be produced using an protein, polypeptide, peptide or oligopeptide with inherent antigenicity. Methods for producing antibodies include: 1) injecting an animal, usually goats, rabbits, or mice, with the protein, or an epitope or oligopeptide thereof, to induce an immune response; 2) engineering hybridomas to produce monoclonal antibodies; 3) inducing in vivo production in the lymphocyte population; or 4) screening libraries of recombinant immunoglobulins. Recombinant immunoglobulins may be produced as taught in USPN 4,816,567.
Antibodies produced using the proteins of the invention are useful for the diagnosis of prepathologic disorders as well as the diagnosis of chronic or acute diseases characterized by abnormalities in the expression, amount, or distribution of the protein. A variety of protocols for competitive binding or immunoradiometric assays using either polyclonal or monoclonal antibodies specific for proteins are well known in the art. Immunoassays typically involve the formation of complexes between a protein and its specific binding molecule or compound and the measurement of complex formation. Immunoassays may employ a two-site, monoclonal-based assay that utilizes monoclonal antibodies reactive to two noninterfering epitopes on a specific protein or a competitive binding assay (Pound (1998) Immunochemical Protocols. Humana Press, Totowa NJ). Assays Using Antibodies
Antibodies directed against epitopes on a protein encoded by a cDNA of the invention may be used in assays to quantify the amount of protein found in a particular human cell. Such assays include methods utilizing the antibody and a label to detect expression level under normal or disease conditions. The antibodies may be used with or without modification, and labeled by joining them, either covalently or noncovalently, with a labeling moiety.
Protocols for detecting and measuring protein expression using either polyclonal or monoclonal antibodies are well known in the art. Examples include ELISA, RIA, FACS, and arrays. Such immunoassays typically involve the formation of complexes between the protein and its specific antibody and the measurement of such complexes. Antibody Arrays
In an alternative to yeast two hybrid system analysis of proteins, an antibody array can be used to study protein-protein interactions and phosphorylation. A variety of protein ligands are immobilized on a membrane using methods well known in the art. The array is incubated in the presence of cell lysate until protenr.antibody complexes are formed. Proteins of interest are identified by exposing the membrane to an antibody specific to the protein of interest. In the alternative, a protein of interest is labeled with digoxigenin (DIG) and exposed to the membrane; then the membrane is exposed to anti-DIG antibody which reveals where the protein of interest forms a complex. The identity of the proteins with which the protein of interest interacts is determined by the position of the protein of interest on the membrane.
Antibody arrays can also be used for high-throughput screening of recombinant antibodies. Bacteria containing antibody genes are robotically-picked and gridded at high density (up to 18,342 different double-spotted clones) on a filter. Up to 15 antigens at a time are used to screen for clones to identify those that express binding antibody fragments. These antibody arrays can also be used to identify proteins which are differentially expressed in samples (de Wildt et al. (2000) Nature Biotechnol 18:989-94). Labeling of Molecules for Assay A wide variety of reporter molecules and conjugation techniques are known by those skilled in the art and can be used with cDNAs, polynucleotides, proteins, peptides or antibodies in screening, purification, and diagnostic assays. Synthesis of labeled molecules may be achieved using commercial kits for incorporation of a labeled nucleotide such as 32P-dCTP, Cy3-dCTP or Cy5-dCTP or amino acid such as 35S-methionine. Polynucleotides, cDNAs, proteins, or antibodies may be directly labeled with a reporter molecule by chemical conjugation to amines, thiols and other groups present in the molecules using reagents such as BIOD-PY or FITC (Molecular Probes, Eugene OR).
The proteins and antibodies may be labeled for purposes of assay by joining them, either covalently or noncovalently, with a reporter molecule that provides for a detectable signal. A wide variety of labels and conjugation techniques are known and have been reported in the scientific and patent literature including, but not limited to USPNs 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241. DIAGNOSTICS
The cDNAs, proteins, or antibodies of the invention may be used to detect and quantify differential gene or protein expression; absence, presence, or excess expression of mRNAs or proteins; or to monitor mRNA or protein levels during therapeutic intervention. Disorders associated with altered expression of the nucleic or amino acid include chronic obstructive pulmonary disease, emphysema, asthma, and in particular, lung cancer. These cDNAs, proteins or antibodies can also be utilized as markers of treatment efficacy against the disorders noted above and other disorders, conditions, and diseases over a period ranging from several days to months. Assays for the presence or transcripts or protein may be hybridization or amplification technologies, ELISAs, RIAs, FACs or any other method used to compare gene or protein expression in a biological sample from a patient to standard samples in order to detect altered expression. Qualitative or quantitative methods for such comparisons are well known in the art.
For example, the cDNA may be labeled by standard methods and added to a biological sample from a patient under conditions for hybridization complex formation. After an incubation period, the sample is washed and the amount of label (or signal) associated with hybridization complexes is quantified and compared with a standard value. If the amount of label in the patient sample is significantly altered in comparison to the standard value, then the presence of the associated condition, disease or disorder is indicated. In order to provide a basis for the diagnosis of a condition, disease or disorder associated with gene expression, a normal or standard expression profile is established. This may be accomplished by combining a biological sample taken from normal subjects, either animal or human, with a probe under conditions for hybridization or amplification. Standard hybridization may be quantified by comparing the values obtained using normal subjects with values from an experiment in which a known amount of a purified target sequence is used. Standard values obtained in this manner may be compared with values obtained from samples from patients who are symptomatic for a particular condition, disease, or disorder. Deviation from standard values toward those associated with a particular condition is used to diagnose that condition.
Such assays may also be used to evaluate the efficacy of a particular therapeutic treatment regimen in animal studies and in clinical trials or to monitor the treatment of an individual patient. Once the presence of a condition is established and a treatment protocol is initiated, diagnostic assays may be repeated on a regular basis to determine if the level of expression in the patient begins to approximate that which is observed in a normal subject. The results obtained from successive assays may be used to show the efficacy of treatment over a period ranging from several days to months. Gene Expression Profiles
A gene expression profile comprises a plurality of cDNAs and a plurality of detectable hybridization complexes, wherein each complex is formed by hybridization of one or more probes to one or more complementary nucleic acids in a sample. The cDNAs of the invention are used as elements on an array to analyze gene expression profiles. In one embodiment, the array is used to monitor the progression of disease. Researchers can assess and catalog the differences in gene expression between healthy and diseased tissues or cells. By analyzing changes in patterns of gene expression, disease can be diagnosed at earlier stages before the patient is symptomatic. The invention can be used to formulate a prognosis and to design a treatment regimen. The invention can also be used to monitor the efficacy of treatment. For treatments with known side effects, the array is employed to improve the treatment regimen. A dosage is established that causes a change in genetic expression patterns indicative of successful treatment. Expression patterns associated with the onset of undesirable side effects are avoided. This approach may be more sensitive and rapid than waiting for the patient to show inadequate improvement, or to manifest side effects, before altering the course of treatment. Experimentally, expression profiles can also be evaluated by methods including, but not limited to, differential display by spatial immobilization, gel electrophoresis, genome mismatch scanning, representational discriminate analysis, clustering, and transcript imaging, and protein or antibody arrays. Expression profiles produced by these methods may be used alone or in combination. The correspondence between mRNA and protein expression has been discussed by Zweiger (2001, Transducing the Genome. McGraw-Hill, San Francisco, CA) and Glavas et al. (2001, T cell activation upregulates cyclic nucleotide phosphodiesterases 8A1 and 7A3, Proc Natl Acad Sci 98:6319-6342) among others.
In another embodiment, animal models which mimic a human disease can be used to characterize expression profiles associated with a particular condition, disorder or disease or its treatment. Novel treatment regimens may be tested in these animal models using arrays to establish and then follow expression profiles over time. In addition, arrays can be used with cell cultures or tissues removed from animal models to rapidly screen large numbers of candidate drug molecules, looking for ones that produce an expression profile similar to those of known therapeutic drugs, with the expectation that molecules with the same expression profile will likely have similar therapeutic effects. Thus, the invention provides the means to rapidly determine the molecular mode of action of a drug. THERAPEUTICS
The cDNAs and fragments thereof can be used in gene therapy. cDNAs can be delivered ex vivo to target cells, such as cells of bone marrow. Once stable integration and transcription and or translation are confirmed, the bone marrow may be reintroduced into the subject. Expression of the protein encoded by the cDNA may correct a disorder associated with mutation of a normal sequence, reduction or loss of an endogenous target protein, or overexpression of an endogenous or mutant protein. Alternatively, cDNAs may be delivered in vivo using vectors such as retrovirus, adenovirus, adeno-associated virus, herpes simplex virus, and bacterial plasmids. Non-viral methods of gene delivery include cationic liposomes, polylysine conjugates, artificial viral envelopes, and direct injection of DNA (Anderson (1998) Nature 392:25-30; Dachs et al. (1997) Oncol Res 9:313-325; Chu et al- (1998) J Mol Med 76(3-4): 184-192; Weiss et al- (1999) Cell Mol Life Sci 55(3):334-358; Agrawal (1996) Antisense Therapeutics. Humana Press, Totowa NJ; and August et al. (1997) Gene Therapy (Advances in Pharmacology, Vol. 40). Academic Press, San Diego CA). In addition, expression of a particular protein can be regulated through the specific binding of a fragment of a cDNA to a genomic sequence or an mRNA which encodes the protein or directs its transcription or translation. The cDNA can be modified or derivatized to any RNA-like or DNA-like material including peptide nucleic acids, branched nucleic acids, and the like. These sequences can be produced biologically by transforming an appropriate host cell with a vector containing the sequence of interest.
Molecules which regulate the activity of the cDNA or expressed protein are useful as therapeutics for respiratory disorders including chronic obstructive pulmonary disease, emphysema, asthma and in particular, lung cancer. Such molecules include agonists which increase the expression or activity of the polynucleotide or encoded protein, respectively; or antagonists which decrease expression or activity of the polynucleotide or encoded protein, respectively. In one aspect, an antibody which specifically binds the protein may be used directly as an antagonist or indirectly as a delivery mechanism for bringing a pharmaceutical agent to cells or tissues which express the protein. Additionally, any of the proteins, or their ligands, or complementary nucleic acid sequences may be administered as pharmaceutical compositions or in combination with other appropriate therapeutic agents. Selection of the appropriate agents for use in combination therapy may be made by one of ordinary skill in the art, according to conventional pharmaceutical principles. The combination of therapeutic agents may act synergistically to affect the treatment or prevention of the conditions and disorders associated with an immune response. Using this approach, one may be able to achieve therapeutic efficacy with lower dosages of each agent, thus reducing the potential for adverse side effects. Further, the therapeutic agents may be combined with pharmaceutically- acceptable carriers including excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Further details on techniques for formulation and administration used by doctors and pharmacists may be found in the latest edition of Remington's Pharmaceutical Sciences (Mack Publishing, Easton PA). Model Systems
Animal models may be used as bioassays where they exhibit a phenotypic response similar to that of humans and where exposure conditions are relevant to human exposures. Mammals are the most common models, and most infectious agent, cancer, drug, and toxicity studies are performed on rodents such as rats or mice because of low cost, availability, lifespan, reproductive potential, and abundant reference literature. Inbred and outbred rodent strains provide a convenient model for investigation of the physiological consequences of underexpression or overexpression of genes of interest and for the development of methods for diagnosis and treatment of diseases. A mammal inbred to overexpress a particular gene (for example, secreted in milk) may also serve as a convenient source of the protein expressed by that gene. Transgenic Animal Models
Transgenic rodents that overexpress or underexpress a gene of interest may be inbred and used to model human diseases or to test therapeutic or toxic agents. (See, e.g., USPN 5,175,383 and USPN 5,767,337.) In some cases, the introduced gene may be activated at a specific time in a specific tissue type during fetal or postnatal development. Expression of the transgene is monitored by analysis of phenotype, of tissue-specific mRNA expression, or of serum and tissue protein levels in transgenic animals before, during, and after challenge with experimental drug therapies. Embryonic Stem Cells
Embryonic (ES) stem cells isolated from rodent embryos retain the potential to form embryonic tissues. When ES cells such as the mouse 129/SvJ cell line are placed in a blastocyst from the C57BL/6 mouse strain, they resume normal development and contribute to tissues of the live-born animal. ES cells are preferred for use in the creation of experimental knockout and knockin animals. The method for this process is well known in the art and the steps are: the cDNA is introduced into a vector, the vector is transformed into ES cells, transformed cells are identified and microinjected into mouse cell blastocysts, blastocysts are surgically transferred to pseudopregnant dams. The resulting chimeric progeny are genotyped and bred to produce heterozygous or homozygous strains. Knockout Analysis
In gene knockout analysis, a region of a gene is enzymatically modified to include a non- natural intervening sequence such as the neomycin phosphotransferase gene (neo; Capecchi (1989) Science 244:1288-1292). The modified gene is transformed into cultured ES cells and integrates into the endogenous genome by homologous recombination. The inserted sequence disrupts transcription and translation of the endogenous gene. Knockin Analysis
ES cells can be used to create knockin humanized animals or transgenic animal models of human diseases. With knockin technology, a region of a human gene is injected into animal ES cells, and the human sequence integrates into the animal cell genome. Transgenic progeny or inbred lines are studied and treated with potential pharmaceutical agents to obtain infoπnation on the progression and treatment of the analogous human condition. I
As described herein, the uses of the cDNAs, provided in the Sequence Listing of this application, and their encoded proteins are exemplary of known techniques and are not intended to reflect any limitation on their use in any technique that would be known to the person of average skill in the art. Furthermore, the cDNAs provided in this application may be used in molecular biology techniques that have not yet been developed, provided the new techniques rely on properties of nucleotide sequences that are currently known to the person of ordinary skill in the art, e.g., the triplet genetic code, specific base pair interactions, and the like. Likewise, reference to a method may include combining more than one method for obtaining, assembling or expressing cDNAs that will be known to those skilled in the art. It is also to be understood that this invention is not limited to the particular methodology, protocols, and reagents described, as these may vary. It is also understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention which will be limited only by the appended claims. The examples below are provided to illustrate the subject invention and are not included for the purpose of limiting the invention.
The disclosures of all patents, applications, and publications mentioned above and below, including U.S. Ser. No. 60/386,005, are hereby expressly incorporated by reference.
EXAMPLES I Construction of cDNA Libraries
RNA was purchased from Clontech Laboratories (Palo Alto CA) or isolated from various tissues. Some tissues were homogenized and lysed in guanidinium isothiocyanate, while others were homogenized and lysed in phenol or in a suitable mixture of denaturants, such as TRIZOL reagent (Invi-rogen). The resulting lysates were centrifuged over CsCl cushions or extracted with chloroform. RNA was precipitated with either isopropanol or ethanol and sodium acetate, or by other routine methods.
Phenol extraction and precipitation of RNA were repeated as necessary to increase RNA purity. In most cases, RNA was treated with DNAse. For most libraries, poly(A) RNA was isolated using oligo d(T)-coupled paramagnetic particles (Promega), OLIGOTEX latex particles (Qiagen, Valencia CA), or an OLIGOTEX mRNA purification kit (Qiagen). Alternatively, poly(A) RNA was isolated directly from tissue lysates using other kits, including the POLY(A)PURE mRNA purification kit (Ambion, Austin TX). hi some cases, Stratagene (La Jolla CA) was provided with RNA and constructed the cDNA libraries. Otherwise, cDNA was synthesized and cDNA libraries were constructed with the UNIZAP vector system (Stratagene) or SUPERSCRIPT plasmid system (Invitrogen) using the recommended procedures or similar methods known in the art. (See Ausubel, supra. Units 5.1 through 6.6.) Reverse transcription was initiated using oligo d(T) or random primers. Synthetic oligonucleotide adapters were ligated to double stranded cDNA, and the cDNA was digested with the appropriate restriction enzyme or enzymes. For most libraries, the cDNA was size-selected (300-1000 bp) using SEPHACRYL S1000, SEPHAROSE CL2B, or SEPHAROSE CL4B column chromatography (APB) or preparative agarose gel electrophoresis. cDNAs were ligated into compatible restriction enzyme sites of the polylinker of the pBLUESCRIPT phagemid (Stratagene), pSPORTl plasmid (Invitrogen), or pINCY plasmid (Incyte Genomics). Recombinant plasmids were transformed into XLl-BLUE, XLl-BLUEMRF, or SOLR competent E. coli cells (Stratagene) or DH5α, DH10B, or ELECTROMAX DH10B competent E. coli cells (Invitrogen). hi some cases, libraries were superinfected with a 5x excess of the helper phage, M13K07, according to the method of Vieira et al. (1987, Methods Enzymol 153:3-11) and normalized or subtracted using a methodology adapted from Soares (1994, Proc Natl Acad Sci 91:9228-9232), Swaroop et al. (1991, Nucleic Acids Res 19: 1954), and Bonaldo et al. (1996, Genome Research 6:791-806). The modified Soares normalization procedure was utilized to reduce the repetitive cloning of highly expressed, high abundance cDNAs while maintaining the overall sequence complexity of the library. Modification included significantly longer hybridization times which allowed for increased gene discovery rates by biasing the normalized libraries toward those infrequently expressed, low-abundance cDNAs which are poorly represented in a standard transcript image (Soares, supra).
II Isolation and Sequencing of cDNA Clones
Plasmids were recovered from host cells by in vivo excision using the UNIZAP vector system (Stratagene) or by cell lysis. Plasmids were purified using one of the following: the Magic or WIZARD M-MPREPS DNA purification system (Promega); the AGTC MIN-PREP purification kit (Edge BioSystems, Gaithersburg MD); the QIAWELL 8, QIAWELL 8 Plus, or QIAWELL 8 Ultra plasmid purification systems, or the REAL PREP 96 plasmid purification kit (Qiagen). Following precipitation, plasmids were resuspended in 0.1 ml of distilled water and stored, with or without lyophilization, at 4°C.
Alternatively, plasmid DNA was amplified from host cell lysates using direct link PCR in a high-throughput format (Rao (1994) Anal Biochem 216: 1-14). Host cell lysis and thermal cycling steps were carried out in a single reaction mixture. Samples were processed and stored in 384-well plates, and the concentration of amplified plasmid DNA was quantified fluorometrically using PICOGREEN dye (Molecular Probes) and a FLUOROSKAN II fluorescence scanner (Labsystems Oy, Helsinki, Finland). cDNA sequencing reactions were processed using standard methods or high-throughput instrumentation such as the CATALYST 800 thermal cycler (ABI) or the DNA ENGINE thermal cycler (MJ Research, Watertown MA) in conjunction with the HYDRA microdispenser (Robbins Scientific, Sunnyvale CA) or the MICROLAB 2200 system (Hamilton, Reno NV). cDNA sequencing reactions were prepared using reagents provided by APB or supplied in sequencing kits such as the PRISM BIGDYE cycle sequencing kit (ABI). Electrophoretic separation of cDNA sequencing reactions and detection of labeled cDNAs were carried out using the MEGABACE 1000 DNA sequencing system (APB); the PRISM 373 or 377 sequencing systems (ABI) in conjunction with standard protocols and base calling software; or other sequence analysis systems known in the art. Reading frames within the cDNA sequences were identified using standard methods and software (reviewed in Ausubel, supra, unit 1.1). Ill Extension of cDNA Sequences
Nucleic acid sequences were extended using the cDNA clones and oligonucleotide primers. One primer was synthesized to initiate 5' extension of the known fragment, and the other, to initiate 3' extension of the known fragment. The initial primers were designed using OLIGO primer analysis software (Molecular Biology Insights, Cascade CO), or another appropriate program, to be about 22 to 30 nucleotides in length, to have a GC content of about 50% or more, and to anneal to the target sequence at temperatures of about 68°C to about 72°C. Any stretch of nucleotides which would result in hairpin structures and primer-primer dimerizations was avoided.
Selected human cDNA libraries were used to extend the sequence. If more than one extension was necessary or desired, additional or nested sets of primers were designed. Preferred libraries are ones that have been size-selected to include larger cDNAs. Also, random primed libraries are preferred because they will contain more sequences with the 5' and upstream regions of genes. A randomly primed library is particularly useful if an oligo d(T) library does not yield a full-length cDNA. High fidelity amplification was obtained by PCR using methods well known in the art. PCR was performed in 96-well plates using the DNA ENGINE thermal cycler (MJ Research). The reaction mix contained DNA template, 200 nmol of each primer, reaction buffer containing Mg2+, (NH4)2S04, and β-mercaptoethanol, Taq DNA polymerase (APB), ELONGASE enzyme (Invitrogen), and Pfu DNA polymerase (Stratagene), with the following parameters for primer pair PCI A and PCI B (Incyte Genomics): Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 60°C, 1 min; Step 4: 68°C, 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68°C, 5 min; Step 7: storage at 4°C. In the alternative, the parameters for primer pair T7 and SK+ (Stratagene) were as follows: Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 57°C, 1 min; Step 4: 68°C, 2 min; Step 5: Steps 2, 3, and 4 repeated 20 times; Step 6: 68°C, 5 min; Step 7: storage at 4°C. 5 The concentration of DNA in each well was determined by dispensing 100 μl PICOGREEN reagent (0.25% reagent in lx TE, v/v; Molecular Probes) and 0.5 μl of undiluted PCR product into each well of an opaque fluorimeter plate (Corning Costar, Acton MA) and allowing the DNA to bind to the reagent. The plate was scanned in a FLUOROSKAN II (Labsystems Oy) to measure the fluorescence of the sample and to quantify the concentration of DNA. A 5 μl to 10 μl aliquot of the
10 reaction mixture was analyzed by electrophoresis on a 1% agarose mini-gel to determine which reactions were successful in extending the sequence.
The extended nucleic acids were desalted and concentrated, transferred to 384-well plates, digested with CviJI cholera virus endonuclease (Molecular Biology Research, Madison WI), and sonicated or sheared prior to religation into pUC18 vector (APB). For shotgun sequencing, the
15 digested nucleic acids were separated on low concentration (0.6 to 0.8%) agarose gels, fragments were excised, and agar digested with AGARACE enzyme (Promega). Extended clones were religated using T4 DNA ligase (New England Biolabs, Beverly MA) into pUC18 vector (APB), treated with Pfu DNA polymerase (Stratagene) to fill in restriction site overhangs, and transformed into competent E. coli cells. Transformed cells were selected on antibiotic-containing media, and
20 individual colonies were picked and cultured overnight at 37°C in 384-well plates in LB/2x carbenicillin liquid media.
The cells were lysed and DNA was amplified by PCR using Taq DNA polymerase (APB) and Pfu DNA polymerase (Stratagene) with the following parameters: Step 1: 94°C, 3 min; Step 2: 94°C, 15 sec; Step 3: 60°C, 1 min; Step 4: 72°C, 2 min; Step 5: steps 2, 3, and 4 repeated 29 times; Step 6:
25 72°C, 5 min; Step 7: storage at 4°C. DNA was quantified using PICOGREEN reagent (Molecular Probes) as described above. Samples with low DNA recoveries were reamplified using the same conditions described above. Samples were diluted with 20% dimethylsulfoxide (DMSO; 1:2, v/v), and sequenced using DYENAMIC energy transfer sequencing primers and the DYENAMIC DIRECT cycle sequencing kit (APB) or the PRISM BIGDYE teπninator cycle sequencing kit (ABI).
30 IV Assembly and Analysis of Sequences
The nucleic acid sequences of the cDNAs presented in the Sequence Listing may contain occasional sequencing errors and unidentified nucleotides (N) that reflect state-of-the-art technology at the time the cDNA was sequenced. Occasional sequencing errors and Ns may be resolved and SNPs verified either by resequencing the cDNA or using algorithms to compare multiple sequences;
35 these techniques are well known to those skilled in the art who wish to practice the invention. The sequences may be analyzed using a variety of algorithms described in Ausubel (supra, unit 1.1) and in Meyers (1995; Molecular Biology and Biotechnology, Wiley VCH, New York NY, pp. 856-853).
Component nucleotide sequences from chromatograms were subjected to PHRED analysis (Phil Green, University of Washington, Seattle WA) and assigned a quality score. The sequences 5 having at least a required quality score were subject to various pre-processing algorithms to eliminate low quality 3' ends, vector and linker sequences, polyA tails, Alu repeats, mitochondrial and ribosomal sequences, bacterial contamination sequences, and sequences smaller than 50 base pairs. Sequences were screened using the BLOCK 2 program (Incyte Genomics), a motif analysis program based on sequence information contained in the SWISS-PROT and PROSITE databases (Bairoch et
10 al. (1997) Nucleic Acids Res 25:217-221; Attwood et al. (1997) J Chem Inf Comput Sci 37:417-424). Processed sequences were subjected to assembly procedures in which the sequences were assigned to bins, one sequence per bin. Sequences in each bin were assembled to produce consensus sequences, referred to as "templates". Subsequent new sequences were added to existing bins using BLAST (Altschul (supra); Altschul (1993, supra); Karlin et al. (1988) Proc Natl Acad Sci 85:841-
15 845), BLASTn (vers.1.4, WashU), and CROSSMATCH software (Green, supra). Candidate pairs were identified as all BLAST hits having a quality score greater than or equal to 150. Alignments of at least 82% local identity were accepted into the bin. The component sequences from each bin were assembled using PHRAP (Green, supra). Bins with several overlapping component sequences were assembled using DEEP PHRAP (Green, supra).
20 Bins were compared against each other, and those having local similarity of at least 82% were combined and reassembled. Reassembled bins having templates of insufficient overlap (less than 95% local identity) were re-split. Assembled templates were also subjected to analysis by STITCHER/EXON MAPPER algorithms which analyzed the probabilities of the presence of splice variants, alternatively spliced exons, splice junctions, differential expression of alternative spliced
25 genes across tissue types, disease states, and the like. These resulting bins were subjected to several rounds of the above assembly procedures to generate the template sequences found in the LIFESEQ GOLD database (Incyte Genomics).
The assembled templates were annotated using the following procedure. Template sequences were analyzed using BLASTn (vers. 2.0, NCBI) versus GBpri (GenBank vers. 116). "Hits" were
30 defined as an exact match having from 95% local identity over 200 base pairs through 100% local identity over 100 base pairs, or a homolog match having an E-value equal to or greater than lxlO"8. (The "E-value" quantifies the statistical probability that a match between two sequences occurred by chance). The hits were subjected to frameshift FASTx versus GENPEPT (GenBank version 109). In this analysis, a homolog match was defined as having an E-value of lxlO"8. The assembly method
35 used above was described in USSN 09/276,534, filed March 25, 1999, and the LIFESEQ GOLD user manual (Incyte Genomics).
Following assembly, template sequences were subjected to motif, BLAST, Hidden Markov Model (HMM; Pearson and Lipman (1988) Proc Natl Acad Sci 85:2444-2448; Smith and Waterman (1981) J Mol Biol 147:195-197), and functional analyses, and categorized in protein hierarchies using methods described in USSN 08/812,290, filed March 6, 1997; USSN 08/947,845, filed October 9, 1997; USPN 5,953,727; and USSN 09/034,807, filed March 4, 1998. Template sequences may be further queried against public databases such as the GenBank rodent, mammalian, vertebrate, eukaryote, prokaryote, and human EST databases.
V Selection of Sequences, Microarray Preparation and Use In most cases, Incyte cDNAs represent template sequences derived from the LIFESEQ
GOLD assembled human sequence database (Incyte Genomics). Where more than one clone was available for a particular template, the 5 -most clone in the template was used on the microarray. The HUMAN GENOME GEM series 1-5 microarray s (Incyte Genomics) contain 45,320 array elements which represent 22,632 annotated clusters and 22,688 unannotated clusters. Tables 1-4 shows the GenBank annotations (where available) for SEQ ID NOs: 1-170 of this invention as produced by BLAST analysis.
To construct microarrays, cDNAs were amplified from bacterial cells using primers complementary to vector sequences flanking the cDNA insert. Thirty cycles of PCR increased the initial quantity of cDNAs from 1-2 ng to a final quantity greater than 5 μg. Amplified cDNAs were then purified using SEPHACRYL-400 columns (APB). Purified cDNAs were immobilized on polymer-coated glass slides. Glass microscope slides (Corning, Corning NY) were cleaned by ultrasound in 0.1% SDS and acetone, with extensive distilled water washes between and after treatments. Glass slides were etched in 4% hydrofluoric acid (VWR Scientific Products, West Chester PA), washed thoroughly in distilled water, and coated with 0.05% aminopropyl silane (Sigma-Aldrich) in 95% ethanol. Coated slides were cured in a 110°C oven. cDNAs were applied to the coated glass substrate using a procedure described in USPN 5,807,522. One microliter of the cDNA at an average concentration of 100 ng/μl was loaded into the open capillary printing element by a high-speed robotic apparatus which then deposited about 5 nl of cDNA per slide.
Microarrays were UV-crosslinked using a STRATALINKER UV-crosslinker (Stratagene), and then washed at room temperature once in 0.2% SDS and three times in distilled water. Nonspecific binding sites were blocked by incubation of microarrays in 0.2% casein in phosphate buffered saline (Tropix, Bedford MA) for 30 minutes at 60°C followed by washes in 0.2% SDS and distilled water as before.
VI Patient Donor Information and Sample Preparation Donor (Dn) Information Dn7160
Moderately differentiated, squamous cell carcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 67 year-old male donor (Roy Castle International Centre for Lung Cancer Research (RCIC), Liverpool, UK). The tumor sample showed 90% overt tumor cells within the tumor. Dn7162
Poorly differentiated, large cell endocrine, stage HB, and grossly uninvolved lung tissue were removed from a 73 year-old male donor (RCIC). The tumor sample showed 60% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue. Dn 7164
Pulmonary carcinoid, stage IA, and grossly uninvolved lung tissue were removed from a 79 year-old male donor (RCIC). The tumor sample showed 100% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue. Dn 7165 Poorly differentiated, adenocarcinoma, stage IA, and grossly uninvolved lung tissue were removed from a 70 year-old female donor (RCIC). The tumor sample showed 20% overt tumor cells within the tumor. Dn 5792
Poorly differentiated, squamous cell carcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 68 year-old female donor (RCIC). The tumor sample showed 50% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 5793
Moderately differentiated, squamous cell carcinoma, stage DA, and grossly uninvolved lung tissue were removed from a 73 year-old male donor (RCIC). The tumor sample showed 60% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 7168
Poorly differentiated, adenocarcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 75 year-old male donor (RCIC). The tumor sample showed 90% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue. Dn 5795
Moderately differentiated, adenocarcinoma, stage IA, and grossly uninvolved lung tissue were removed from a 71 year-old female donor (RCIC). The tumor sample showed 75% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 5796 Moderately differentiated, squamous cell carcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 66 year-old male donor (RCIC). The tumor sample showed 50% overt tumor cells within the tumor, and less than 10% overt tumor cells in the uninvolved tissue. Dn 7173
Moderately differentiated, squamous cell carcinoma, stage IDB, and grossly uninvolved lung tissue were removed from a 70 year-old male donor (RCIC). The tumor sample showed 70% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 5797
Moderately differentiated, squamous cell carcinoma, stage HB, and grossly uninvolved lung tissue were removed from a 73 year-old male donor (RCIC). The tumor sample showed 80% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 7175
Moderately differentiated, adenocarcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 67 year-old male donor (RCIC). The tumor sample showed 50% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue. Dn 7176
Poorly differentiated, adenosquamous, stage IB, and grossly uninvolved lung tissue were removed from a 72 year-old male donor (RCIC). The tumor sample showed 60% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 7178 Moderately differentiated, squamous cell carcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 68 year-old female donor (RCIC). The tumor sample showed 70% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue. Dn 7179
Poorly differentiated, adenocarcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 62 year-old male donor (RCIC). The tumor sample showed 90% overt tumor cells within the tumor, and less than 20% overt tumor cells in the uninvolved tissue. Dn 7186
Atypical carcinoid, stage IA, and grossly uninvolved lung tissue were removed from a 61 year-old male donor (RCIC). The tumor sample showed 90% overt tumor cells within the tumor. Dn 5799
Moderately differentiated, adenocarcinoma, stage IIB, and grossly uninvolved lung tissue were removed from a 66 year-old female donor (RCIC). The tumor sample showed 80% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue. Dn 7188 Poorly differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 54 year-old male donor (RCIC). The tumor sample showed 90% overt tumor cells within the tumor, and less than 10% overt tumor cells in the uninvolved tissue.
Dn 7189
Poorly differentiated, adenocarcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 78 year-old male donor (RCIC). The tumor sample showed 70% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 7190
Moderately differentiated, squamous cell carcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 50 year-old female donor (RCIC). The tumor sample showed 70% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue. Dn 7191
Poorly differentiated, squamous cell carcinoma, stage DB, and grossly uninvolved lung tissue were removed from a 43 year-old male donor (RCIC). The tumor sample showed 70% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 7192
Large cell endocrine, stage IB, and grossly uninvolved lung tissue were removed from a 54 year-old female donor (RCIC). The tumor sample showed 70% overt tumor cells within the tumor. Dn 7194
Moderately differentiated, squamous cell carcinoma, stage DB, and grossly uninvolved lung tissue were removed from a 60 year-old female donor (RCIC). The tumor sample showed 50% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 5800
Moderately differentiated, squamous cell carcinoma, stage DB, and grossly uninvolved lung tissue were removed from a 75 year-old female donor (RCIC). The tumor sample showed 70% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 7196
Well differentiated, squamous cell carcinoma, stage IB, and grossly uninvolved lung tissue were removed from a 71 year-old male donor (RCIC). The tumor sample showed 80% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 7197
Poorly differentiated, adenocarcinoma, stage IA, and grossly uninvolved lung tissue were removed from a 53 year-old male donor (RCIC). The tumor sample showed 70% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. Dn 7962 Moderately differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 71 year-old male donor (RCIC). The tumor sample showed 85% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with partial remission of the tumor prior to surgery. Dn 7963 Poorly differentiated, adenocarcinoma, stage DIA, and grossly uninvolved lung tissue were removed from a 71 year-old male donor (RCIC). The tumor sample showed 60% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with progressive disease in the tumor prior to surgery. Dn 7964 Moderately differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 50 year-old male donor (RCIC). The tumor sample showed 70% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with partial remission of the tumor prior to surgery. Dn 7965 Moderately differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 54 year-old female donor (RCIC). The tumor sample showed 60% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with no change in the tumor prior to surgery. Dn 7966 Moderately differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 39 year-old female donor (RCIC). The tumor sample showed 10% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with complete remission of the tumor prior to surgery. Dn 7967 Moderately differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 57 year-old female donor (RCIC). The tumor sample showed 60% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with no change in the tumor prior to surgery. Dn 7968 Moderately differentiated, squamous cell carcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 58 year-old male donor (RCIC). The tumor sample showed 10% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with no change in the tumor prior to surgery. Dn 7969 Poorly differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 62 year-old male donor (RCIC). The tumor sample showed 60% overt tumor cells within the tumor, and less than 5% overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with no change in the tumor prior to surgery. Dn 7970 Poorly differentiated, adenocarcinoma, stage DIA, and grossly uninvolved lung tissue were removed from a 53 year-old female donor (RCIC). The tumor sample showed no overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with partial remission of the tumor prior to surgery. Dn 7971 Poorly differentiated, large cell carcinoma, stage DIA, and grossly uninvolved lung tissue were removed from a 54 year-old male donor (RCIC). The tumor sample showed 90% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with no change in the tumor prior to surgery. Dn 7972 Moderately differentiated, squamous cell carcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 62 year-old male donor (RCIC). The tumor sample showed 10% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with complete remission of the tumor prior to surgery. Dn 7973 Poorly differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 54 year-old male donor (RCIC). The tumor sample showed 60% overt tumor cells within the tumor, and no apparent overt tumor cells in the uninvolved tissue. The patient had undergone 3 cycles of chemotherapy with no change in the tumor prior to surgery. Dn 7975 Poorly differentiated, adenocarcinoma, stage IDA, and grossly uninvolved lung tissue were removed from a 62 year-old male donor (RCIC). The tumor sample showed 10% overt tumor cells within the tumor. The patient had undergone 3 cycles of chemotherapy with partial remission of the tumor prior to surgery. Sample Preparation and Utilization Donor samples Dn 7160, Dn 7165, Dn 7186, and Dn 7192, were matched against a pool of normal/uninvolved tissues from Dn 7173, 7175, 7176, 7178, 7188, 7189, 7190, 7191, 7196, and 7197. Donor sample Dn 7975 was matched against a pool of normal/uninvolved tissues fromDn 7964, 7965, 7966, 7967, and 7968. All other patient samples were matched against normal/uninvolved tissue from the same patient. Each of the lung tissue samples was lysed in 1 ml of TRIZOL reagent (Invitrogen). The lysate was vortexed thoroughly, incubated at room temperature for 2-3 minutes, and extracted with 0.5 ml chloroform. The extract was mixed, incubated at room temperature for 5 minutes, and centrifuged at 15,000 rpm for 15 minutes at 4°C. The aqueous layer was collected, and an equal volume of isopropanol was added. Samples were mixed, incubated at room temperature for 10 minutes, and centrifuged at 15,000 rpm for 20 minutes at 4°C. The supernatant was removed, and the RNA pellet was washed with 1 ml of 70% ethanol, centrifuged at 15,000 rpm at 4°C, and resuspended in RNAse-free water. The concentration of the total RNA was determined by measuring the optical density at 260 nm.
Poly(A) RNA was prepared using an OLIGOTEX mRNA kit (Qiagen) with the following modifications: OLIGOTEX beads were washed in tubes instead of on spin columns, resuspended in elution buffer, and then loaded onto spin columns to recover mRNA. To obtain maximum yield, the mRNA was eluted twice.
Each poly(A) RNA sample was reverse transcribed using a cDNA synthesis system (Invitrogen) with Not I-T7-VN primers (5' GCATTAGCGGCCGCGAAATTAATACGACTCACTATAGGGAGAT TTTTTTTTTTTTTTTTVN 3*) and 100 units MMLV RNAseH (-) reverse-transcriptase (Progmega) in the first strand reaction. The resulting cDNA was purified on a CHROMASPDSf TE-200 column (Clontech) and lyophilized until dry. The cDNA was amplified 200-400 fold using an AMPLISCRIBE IVT kit (Epicentre Technologies, Madison WI) in a procedure modified from USPN 5,716,785 and USPN 5,891,636. The amplified RNA was purified on a CHROMASPIN DEPC-200 column (Clontech).
Amplified RNA was labeled using MMLV reverse-transcriptase, random primer (9mer), lx first strand buffer, 0.03 units/μl RNAse inhibitor, 500 μM dATP, 500 μM dGTP, 500 μM dTTP, 40 μM dCTP, and 40 μM either dCTP-Cy3 or dCTP-Cy5 (APB). The reverse transcription reaction was performed in a 25 ml volume containing 200 ng poly(A) RNA using the GEMBRIGHT kit (Incyte Genomics). Specific control poly(A) RNAs (YCFR06, YCFR45, YCFR67, YCFR85, YCFR43, YCFR22, YCFR23, YCFR25, YCFR44, YCFR26) were synthesized by in vitro transcription from non-coding yeast genomic DNA (W. Lei, unpublished). As quantitative controls, control RNAs (YCFR06, YCFR45, YCFR67, and YCFR85) at 0.002ng, 0.02ng, 0.2 ng, and 2ng were diluted into reverse transcription reaction at ratios of 1:100,000, 1:10,000, 1:1000, 1:100 (w/w) to sample mRNA, respectively. To sample differential expression patterns, control mRNAs (YCFR43, YCFR22, YCFR23, YCFR25, YCFR44, YCFR26) were diluted into reverse transcription reaction at ratios of 1:3, 3:1, 1:10, 10: 1, 1:25, 25: 1 (w/w) to sample mRNA. Reactions were incubated at 37°C for 2 hr, treated with 2.5 ml of 0.5M sodium hydroxide, and incubated for 20 minutes at 85°C to the stop the reaction and degrade the RNA. cDNAs were purified using two successive CHROMA SPESf 30 gel filtration spin columns (Clontech). Cy3- and Cy5-labeled reaction samples were combined as described below and ethanol precipitated using 1 ml of glycogen (1 mg/ml), 60 ml sodium acetate, and 300 ml of 100% ethanol. The cDNAs were then dried to completion using a SPEED VAC system (Savant Instruments, Holbrook NY) and resuspended in 14 μl 5X SSC/0.2% SDS. VII Hybridization and Detection
Competitive hybridization reactions compared cDNAs derived from tumor tissue and uninvolved tissue from the same donor. cDNA from all donors except Dn5795 were hybridized to HUMAN GENOME GEMs 1-5. cDNAs from Dn5795 were hybridized to HUMAN GENOME GEM 1. Hybridization reactions contained 9 μl of sample mixture containing 0.2 μg each of Cy3 and Cy5 labeled cDNA synthesis products in 5X SSC, 0.2% SDS hybridization buffer. The mixture was heated to 65°C for 5 minutes and was aliquoted onto the microarray surface and covered with an 1.8 cm2 coverslip. The microarrays were transferred to a waterproof chamber having a cavity just slightly larger than a microscope slide. The chamber was kept at 100% humidity internally by the addition of 140 μl of 5x SSC in a corner of the chamber. The chamber containing the microarrays was incubated for about 6.5 hours at 60°C. The microarrays were washed for 10 min at 45°C in low stringency wash buffer (lx SSC, 0.1% SDS), three times for 10 minutes each at 45°C in high stringency wash buffer (O.lx SSC), and dried.
Reporter-labeled hybridization complexes were detected with a microscope equipped with an Innova 70 mixed gas 10 W laser (Coherent, Santa Clara CA) capable of generating spectral lines at 488 nm for excitation of Cy3 and at 632 nm for excitation of Cy5. The excitation laser light was focused on the microarray using a 20X microscope objective (Nikon, Melville NY). The slide containing the microarray was placed on a computer-controlled X-Y stage on the microscope and raster-scanned past the objective. The 1.8 cm x 1.8 cm microarray used in the present example was scanned with a resolution of 20 micrometers. In two separate scans, the mixed gas multiline laser excited the two fluorophores sequentially.
Emitted light was split, based on wavelength, into two photomultiplier tube detectors (PMT R1477; Hamamatsu Photonics Systems, Bridgewater NJ) corresponding to the two fluorophores. Appropriate filters positioned between the microarray and the photomultiplier tubes were used to filter the signals. The emission maxima of the fluorophores used were 565 nm for Cy3 and 650 nm for Cy5. Each microarray was typically scanned twice, one scan per fluorophore using the appropriate filters at the laser source, although the apparatus was capable of recording the spectra from both fluorophores simultaneously.
The sensitivity of the scans was calibrated using the signal intensity generated by a cDNA control species. Samples of the calibrating cDNA were separately labeled with the two fluorophores and identical amounts of each were added to the hybridization mixture. A specific location on the microarray contained a complementary DNA sequence, allowing the intensity of the signal at that location to be correlated with a weight ratio of hybridizing species of 1:100,000.
The output of the photomultiplier tube was digitized using a 12-bit RTI-835H analog-to-digital (A/D) conversion board (Analog Devices, Norwood, MA) installed in an IBM-compatible PC computer. 5 The digitized data were displayed as an image where the signal intensity was mapped using a linear 20- color transformation to a pseudocolor scale ranging from blue (low signal) to red (high signal). The data was also analyzed quantitatively. Where two different fluorophores were excited and measured simultaneously, the data were first corrected for optical crosstalk (due to overlapping emission spectra) between the fluorophores using each fluorophore 's emission spectrum.
10 A grid was superimposed over the fluorescence signal image such that the signal from each spot was centered in each element of the grid. The fluorescence signal within each element was then integrated to obtain a numerical value corresponding to the average intensity of the signal. The software used for signal analysis was the GEMTOOLS gene expression analysis program (Incyte Genomics). Significance was defined as signal to background ratio exceeding 2x and area hybridization exceeding
15 40%.
VIII Data Analysis and Results
Array elements that exhibited a 4-fold or greater change in expression at one or more time points, a signal intensity over 250 units, a signal-to-background ratio of at least 2.5, and an element spot size of at least 40% were identified as differentially expressed using the GEMTOOLS program (Incyte 20 Genomics). The cDNAs that were differentially expressed are shown in Tables 1-4 and are identified by both SEQ ID NO and Incyte ID.
IX Further Characterization of Differentially Expressed cDNAs and Proteins Clones were aligned against the LIFESEQ GOLD 5.1 database (Jan2002 release; Incyte
Genomics) using BLAST analysis and an Incyte template was chosen for each clone. The template was 25 aligned against GenBank database using BLAST analysis to acquire annotation. The nucleotide sequences were translated into amino acid sequences which were aligned against GenPept and other protein databases to acquire annotation and characterization.
Percent sequence identity can be determined electronically for two or more amino acid or nucleic acid sequences using the MEGALIGN program, a component of LASERGENE software 30 (DNASTAR). The percent identity between two amino acid sequences is calculated by dividing the length of sequence A, minus the number of gap residues in sequence A, minus the number of gap residues in sequence B, into the sum of the residue matches between sequence A and sequence B, times one hundred. Gaps of low or of no homology between the two amino acid sequences are not included in determining percentage identity. 35 Sequences with conserved protein motifs may be searched using the BLOCKS search program. This program analyses sequence information contained in the SWISSPROT and PROSITE databases and is useful for determining the classification of uncharacterized proteins translated from genomic or cDNA sequences (Bairoch, supra; Attwood, supra). PROSITE database is a useful source for identifying functional or structural domains that are not detected using motifs due to extreme sequence divergence. Using weight matrices, these domains are calibrated against the SWISSPROT database to obtain a measure of the chance distribution of the matches.
The PRINTS database can be searched using the BLIMPS search program to obtain protein family "fingerprints". The PRINTS database complements the PROSITE database by exploiting groups of conserved motifs within sequence alignments to build characteristic signatures of different protein families. For both BLOCKS and PRINTS analyses, the cutoff scores for local similarity were:
>1300=strong, 1000-1300=suggestive; for global similarity were: p<exp-3; and for strength (degree of correlation) were: >1300=strong, 1000-1300=weak. Pfam is a large collection of multiple sequence alignments and hidden Markov models covering many common protein domains. Version 5.5 of Pfam (Sept 2000) contains alignments and models for 2478 protein families, based on the SWISSPROT 38 and SP-TrEMBL 11 protein sequence databases.
X Other Hybridization Technologies and Analyses
Other hybridization technologies utilize a variety of substrates such as nylon membranes, capillary tubes, etc. Arranging cDNAs on polymer coated slides is described in Example V; sample cDNA preparation, hybridization, and analysis using polymer coated slides is described in examples VI and VII, respectively.
The cDNAs are applied to a membrane substrate by one of the following methods. A mixture of cDNAs is fractionated by gel electrophoresis and transferred to a nylon membrane by capillary transfer. Alternatively, the cDNAs are individually ligated to a vector and inserted into bacterial host cells to form a library. The cDNAs are then arranged on a substrate by one of the following methods, hi the first method, bacterial cells containing individual clones are robotically picked and arranged on a nylon membrane. The membrane is placed on LB agar containing selective agent (carbenicillin, kanamycin, ampicillin, or chloramphenicol depending on the vector used) and incubated at 37°C for 16 hours. The membrane is removed from the agar and consecutively placed colony side up in 10% SDS, denaturing solution (1.5 M NaCl, 0.5 M NaOH ), neutralizing solution (1.5 M NaCl, 1 M Tris, pH 8.0), and twice in 2x SSC for 10 minutes each. The membrane is then UV irradiated in a STRATALINKER UV- crosslinker (Stratagene). In the second method, cDNAs are amplified from bacterial vectors by thirty cycles of PCR using primers complementary to vector sequences flanking the insert. PCR amplification increases a starting concentration of 1-2 ng nucleic acid to a final quantity greater than 5 μg. Amplified nucleic acids from about 400 bp to about 5000 bp in length are purified using SEPHACRYL-400 beads (APB). Purified nucleic acids are arranged on a nylon membrane manually or using a dot/slot blotting manifold and suction device and are immobilized by denaturation, neutralization, and UV irradiation as described above.
Hybridization probes derived from cDNAs of the Sequence Listing are employed for screening cDNAs, mRNAs, or genomic DNA in membrane-based hybridizations. Probes are prepared by diluting the cDNAs to a concentration of 40-50 ng in 45 μl Tris-EDTA (ethylenediamine tetraacetic acid) (TE) buffer, denaturing by heating to 100°C for five minutes and centrifuging briefly. The denatured cDNA is then added to a REDIPRIME tube (APB), gently mixed until blue color is evenly distributed, and centrifuged briefly. Five microliters of [32P]dCTP is added to the tube, and the contents are incubated at 37°C for 10 minutes. The labeling reaction is stopped by adding 5 μl of 0.2M EDTA, and probe is purified from unincorporated nucleotides using a PROBEQUANT G-50 microcolumn (APB). The purified probe is heated to 100°C for five minutes and then snap cooled for 2 minutes on ice.
Membranes are pre-hybridized in hybridization solution containing 1% Sarkosyl and lx high phosphate buffer (0.5 M NaCl, 0.1 M Na2HPO4, 5 mM EDTA, pH 7) at 55°C for 2 hours. The probe, diluted in 15 ml fresh hybridization solution, is then added to the membrane. The membrane is hybridized with the probe at 55°C for 16 hours. Following hybridization, the membrane is washed for 15 minutes at 25°C in lmM Tris (pH 8.0), 1% Sarkosyl, and four times for 15 minutes each at 25°C in lmM Tris (pH 8.0). To detect hybridization complexes, XOMAT-AR film (Eastman Kodak, Rochester NY) is exposed to the membrane overnight at -70°C, developed, and examined. OPCR Analysis For QPCR, cDNA is synthesized from 1 ug total RNA in a 25 ul reaction with 100 units M-
MLV reverse transcriptase (Ambion, Austin TX), 0.5 mM dNTPs (Epicentre, Madison WI), and 40 ng/ml random hexamers (Fisher Scientific, Chicago IL). Reactions are incubated at 25C for 10 minutes, 42C for 50 minutes, and 70C for 15 minutes, diluted to 500 ul, and stored at -30C. Alternatively, cDNA is obtained from Human MTC panels (Clontech Laboratories, Palo Alto CA). PCR primers and probes (5' 6-FAM-labeled, 3' TAMRA) are designed using PRIMER EXPRESS 1.5 software (ABI) and synthesized by Biosearch Technologies (Novato CA) or ABI.
QPCR reactions are performed using an PRISM 7700 sequencing system (ABI) in 25 ul total volume with 5 ul cDNA template, lx TAQMAN UNIVERSAL PCR master mix (ABI), 100 nM each PCR primer, 200 nM probe, and lx VIC-labeled beta-2-microglobulin endogenous control (ABI). Reactions are incubated at 50C for 2 minutes, 95C for 10 minutes, followed by 40 cycles of incubation at 95C for 15 seconds and 60C for 1 minute. Emissions are measured every 7 seconds, and results are analyzed using SEQUENCE DETECTOR 1.7 software (ABI) and fold differences, relative concentration of mRNA as compared to standards, are calculated using the comparative Cτ method (ABI User Bulletin #2). XI Northern Analysis and Transcript Imaging Northern analysis is a laboratory technique used to detect the presence of a transcript of a gene and involves the hybridization of a labeled nucleotide sequence to a membrane on which RNAs from a particular cell type or tissue have been bound (Sambrook, supra, ch. 7 and Ausubel, supra, ch. 4 and 16.)
Analogous computer techniques applying BLAST are used to search for identical or related molecules in nucleotide databases such as GenBank or the LIFESEQ database (Incyte Genomics). This analysis is faster than multiple membrane-based hybridizations. In addition, the sensitivity of the computer search can be modified to determine whether any particular match is categorized as exact or homologous. The basis of the search is the product score, which is defined as:
% sequence identity x % maximum BLAST score 100
The product score takes into account both the degree of similarity between two sequences and the length of the sequence match. For example, with a product score of 40, the match will be exact within a 1% to
2% error, and, with a product score of 70, the match will be exact. Homologous molecules are usually identified by selecting those which show product scores between 15 and 40, although lower scores may identify related molecules.
The results of northern analysis are reported as a list of libraries in which the transcript occurs. Abundance and percent abundance are also reported. Abundance directly reflects the number of times a particular transcript is represented in a cDNA library, and percent abundance is abundance divided by the total number of sequences examined in the cDNA library. Transcript Imaging
A transcript image is performed using the LIFESEQ GOLD database (Incyte Genomics). This process allows assessment of the relative abundance of the expressed polynucleotides in all of the cDNA libraries and was described in USPN 5,840,484, incorporated herein by reference. All sequences and cDNA libraries in the LIFESEQ database are categorized by system, organ/tissue and cell type. The categories include cardiovascular system, connective tissue, digestive system, embryonic structures, endocrine system, exocrine glands, female and male genitalia, germ cells, hemic/immune system, liver, musculoskeletal system, nervous system, pancreas, respiratory system, sense organs, skin, stomatognathic system, unclassified/mixed, and the urinary tract. Criteria for transcript imaging are selected from category, number of cDNAs per library, library description, disease indication, clinical relevance of sample, and the like.
For each category, the number of libraries in which the sequence is expressed are counted and shown over the total number of libraries in that category. For each library, the number of cDNAs are counted and shown over the total number of cDNAs in that library. In some transcript images, all enriched, normalized or subtracted libraries, which have high copy number sequences can be removed prior to processing, and all mixed or pooled tissues, which are considered non-specific in that they contain more than one tissue type or more than one subject's tissue, can be excluded from the analysis. Treated and untreated cell lines and/or fetal tissue data can also be excluded where clinical relevance is emphasized. Conversely, fetal tissue can be emphasized wherever elucidation of inherited disorders or differentiation of particular adult or embryonic stem cells into tissues or organs such as heart, kidney, nerves or pancreas would be aided by removing clinical samples from the analysis. Transcript imaging can be used to support data from other methodologies and technologies.
XII Chromosome Mapping
Radiation hybrid and genetic mapping data available from public resources such as the Stanford Human Genome Center (SHGC), Whitehead Institute for Genome Research (WIGR), and Genethon are used to determine if any of the cDNAs presented in the Sequence Listing have been mapped. Any of the fragments of the cDNA encoding a particular protein that have been mapped result in the assignment of all related regulatory and coding sequences to the same location. The genetic map locations are described as ranges, or intervals, of human chromosomes. The map position of an interval, in cM (which is roughly equivalent to 1 megabase of human DNA), is measured relative to the terminus of the chromosomal p-arm.
XIII Protein Expression and Purification
Expression and purification of the protein are achieved using either a mammalian cell expression system or an insect cell expression system. The pUB6/V5-His vector system (Invitrogen) is used to express GSCC in CHO cells. The vector contains the selectable bsd gene, multiple cloning sites, the promoter/enhancer sequence from the human ubiquitin C gene, a C-terminal V5 epitope for antibody detection with anti-V5 antibodies, and a C-terminal polyhistidine (6xHis) sequence for rapid purification on PROBOND resin (Invitrogen). Transformed cells are selected on media containing blasticidin.
Spodoptera frugiperda (Sf9) insect cells are infected with recombinant Autographica californica nuclear polyhedrosis virus (baculovirus). The polyhedrin gene is replaced with the cDNA by homologous recombination and the polyhedrin promoter drives cDNA transcription. The protein is synthesized as a fusion protein with 6xhis which enables purification as described above. Purified protein is used in the following activity and to make antibodies.
XIV Production of Specific Antibodies
Purification using polyacrylamide gel electrophoresis or similar techniques is used to isolate protein for immunization of hosts or host cells to produce antibodies using standard protocols.
A denatured protein from a reverse phase HPLC separation is obtained in quantities up to 75 mg. This denatured protein is used to immunize mice or rabbits following standard protocols. About 100 μg is used to immunize a mouse, while up to 1 mg is used to immunize a rabbit. The denatured protein is radioiodinated and incubated With murine B-cell hybridomas to screen for monoclonal antibodies. About 20 mg of protein is sufficient for labeling and screening several thousand clones. In another approach, the amino acid sequence translated from a cDNA of the invention is analyzed using PROTEAN software (DNASTAR) to select regions of high antigenicity, antigenically- effective epitopes of the protein. The optimal sequences for immunization are usually at the C-terminus, the N-terminus, and those intervening hydrophilic regions of the protein that are likely to be exposed to the external environment when the protein is in its natural conformation. Typically, oligopeptides about 15 residues in length are synthesized using a 431 peptide synthesizer (ABI) using Fmoc-chemistry and then coupled to keyhole limpet hemocyanin (KLH; Sigma-Aldrich) by reaction with M-maleimidobenzoyl-N-hydroxysuccinimide ester. If necessary, a cysteine may be introduced at the N-teπninus of the peptide to permit coupling to KLH. Rabbits are immunized with the oligopeptide-KLH complex in complete Freund's adjuvant. The resulting antisera are tested for antipeptide activity by binding the peptide to plastic, blocking with 1% BSA, reacting with rabbit antisera, washing, and reacting with radioiodinated goat anti-rabbit IgG.
Hybridomas are prepared and screened using standard techniques. Hybridomas of interest are detected by screening with radioiodinated protein to identify those fusions producing a monoclonal antibody specific for the protein. In a typical protocol, wells of 96 well plates (FAST,
Becton-Dickinson, Palo Alto CA) are coated with affinity-purified, specific rabbit-anti-mouse (or suitable anti-species Ig) antibodies at 10 mg/ml. The coated wells are blocked with 1% BSA, washed, and exposed to supematants from hybridomas. After incubation, the wells are exposed to radiolabeled protein at 1 mg/ml. Clones producing antibodies bind a quantity of labeled protein that is detectable above background.
Such clones are expanded and subjected to 2 cycles of cloning at 1 cell/3 wells. Cloned hybridomas are injected into pristane-treated mice to produce ascites, and monoclonal antibody is purified from the ascitic fluid by affinity chromatography on protein A (APB). Monoclonal antibodies with affinities of at least 108 M"\ preferably 109 to 1010 M"1 or stronger, are made by procedures well known in the art.
XV Purification of Naturally Occurring Protein Using Specific Antibodies
Naturally occurring or recombinant protein is immunopurified by affinity chromatography using antibodies specific for the protein. An immunoaffinity column is constructed by covalently coupling the antibody to CNBr-activated SEPHAROSE resin (APB). Media containing the protein is passed over the immunoaffinity column, and the column is washed using high ionic strength buffers in the presence of detergent to allow preferential absorbance of the protein. After coupling, the protein is eluted from the column using a buffer of pH 2-3 or a high concentration of urea or thiocyanate ion to disrupt antibody/protein binding, and the protein is collected.
XVI Screening Molecules for Specific Binding with the cDNA or Protein The cDNA or fragments thereof and the protein or portions thereof are labeled with 32P-dCTP, Cy3-dCTP, Cy5-dCTP (APB), or BIODIPY or FITC (Molecular Probes), respectively. Candidate molecules or compounds previously arranged on a substrate are incubated in the presence of labeled nucleic or amino acid. After incubation under conditions for either a cDNA or a protein, the substrate is washed, and any position on the substrate retaining label, which indicates specific binding or complex formation, is assayed. The binding molecule is identified by its arrayed position on the substrate. Data obtained using different concentrations of the nucleic acid or protein are used to calculate affinity between the labeled nucleic acid or protein and the bound molecule. High throughput screening using very small assay volumes and very small amounts of test compound is fully described in USPN 5,876,946. XVII Antibody Arrays Proteimprotein interactions
In an alternative to yeast two hybrid system analysis of proteins, an antibody array can be used to study protein-protein interactions and phosphorylation. A variety of protein ligands are immobilized on a membrane using methods well known in the art. The array is incubated in the presence of cell lysate until proteimantibody complexes are formed. Proteins of interest are identified by exposing the membrane to an antibody specific to the protein of interest. In the alternative, a protein of interest is labeled with digoxigenin (DIG) and exposed to the membrane; then the membrane is exposed to anti- DIG antibody which reveals where the protein of interest forms a complex. The identity of the proteins with which the protein of interest interacts is determined by the position of the protein of interest on the membrane.
Proteomic Profiles
Antibody arrays can also be used for high-throughput screening of recombinant antibodies. Bacteria containing antibody genes are robotically-picked and gridded at high density (up to 18,342 different double-spotted clones) on a filter. Up to 15 antigens at a time are used to screen for clones to identify those that express binding antibody fragments. These antibody arrays can also be used to identify proteins which are differentially expressed in samples (de Wildt, supra) XVIII Two-Hybrid Screen
A yeast two-hybrid system, MATCHMAKER LexA Two-Hybrid system (Clontech Laboratories), is used to screen for peptides that bind the protein of the invention. A cDNA encoding the protein is inserted into the multiple cloning site of a pLexA vector, ligated, and transformed into E. coli. cDNA, prepared from mRNA, is inserted into the multiple cloning site of a pB42AD vector, ligated, and transformed into E. coli to construct a cDNA library. The pLexA plasmid and pB42AD- cDNA library constructs are isolated from E. coli and used in a 2: 1 ratio to co-transform competent yeast EGY48[p8op-lacZ] cells using a polyethylene glycol/lithium acetate protocol. Transformed yeast cells are plated on synthetic dropout (SD) media lacking histidine (-His), tryptophan (-Trp), and uracil (-Ura), and incubated at 30C until the colonies have grown up and are counted. The colonies are pooled in a minimal volume of lx TE (pH 7.5), replated on SD/-His/-Leu/-Trp/-Ura media supplemented with 2% galactose (Gal), 1% raffinose (Raf), and 80 mg/ml 5-bromo-4-chloro-3-indolyl β-d-galactopyranoside (X-Gal), and subsequently examined for growth of blue colonies. Interaction between expressed protein and cDNA fusion proteins activates expression of a LEU2 reporter gene in EGY48 and produces colony growth on media lacking leucine (-Leu). Interaction also activates expression of β-galactosidase from the p8op-lacZ reporter construct that produces blue color in colonies grown on X-Gal.
Positive interactions between expressed protein and cDNA fusion proteins are verified by isolating individual positive colonies and growing them in SD/-Trp/-Ura liquid medium for 1 to 2 days at 30C. A sample of the culture is plated on SD/-Trp/-Ura media and incubated at 30C until colonies appear. The sample is replica-plated on SD/-Trp/-Ura and SD/-His/-Trp/-Ura plates. Colonies that grow on SD containing histidine but not on media lacking histidine have lost the pLexA plasmid. Histidine- requiring colonies are grown on SD/Gal/Raf/X-Gal/-Trp/-Ura, and white colonies are isolated and propagated. The pB42AD-cDNA plasmid, which contains a cDNA encoding a protein that physically interacts with the protein, is isolated from the yeast cells and characterized.
All patents and publications mentioned in the specification are incorporated herein by reference. Various modifications and variations of the described method and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those skilled in the field of molecular biology or related fields are intended to be within the scope of the following claims.
TABLE 1
Figure imgf000047_0001
TABLE 2
Figure imgf000048_0001
TABLE 3
Figure imgf000049_0001
TABLE 3
Figure imgf000050_0001
TABLE 4
©
Figure imgf000051_0001
TABLE 4
Figure imgf000052_0001
TABLE 4
Figure imgf000053_0001
TABLE 4
Figure imgf000054_0001
TABLE 5
Figure imgf000055_0001

Claims

What is claimed is:
1. A combination comprising a plurality of cDNAs wherem the cDNAs are SEQ ID NOs: 1-170 that are differentially expressed in respiratory disorders and the complete complements of SEQ ID NOs: 1-170.
2. The combination of claim 1, wherein the cDNAs are SEQ ID NOs: 1-37 that are differentially expressed at least 16-fold in respiratory disorders and the complements of SEQ ID NOs: 1-37.
3. The combination of claim 1, wherein the respiratory disorder is lung cancer, chronic obstructive pulmonary disease, emphysema, or asthma.
4. The combination of claim 1, wherem the cDNAs are immobilized on a substrate.
5. A method for detecting differential expression of at least one cDNA in a sample containing nucleic acids, the method comprising: a) hybridizing the substrate of claim 4 with nucleic acids of the sample, thereby forming at least one hybridization complex; b) comparing complex formation with that of a standard, wherein differences between sample and standard complex formation indicate differential expression of the cDNA in the sample.
6. The method of claim 5, wherein the nucleic acids of the sample are amplified prior to hybridization.
7. The method of claim 5, wherein the sample is from a subject with a respiratory disorder.
8. A method of screening a plurality of molecules or compounds to identify a ligand which specifically binds a cDNA, the method comprising: a) combining the substrate of claim 4 with the plurality of molecules or compounds under conditions to allow specific binding; and b) detecting specific binding between at least one cDNA and at least one molecule or compound, thereby identifying a ligand that specifically binds to the cDNA.
9. The method of claim 8 wherein the plurality of molecules or compounds are selected from DNA molecules, enhancers, mimetics, peptide nucleic acid molecules, peptides, regulatory proteins, repressors, RNA molecules, and transcription factors.
10. An isolated cDNA comprising a nucleic acid sequence selected from SEQ ID NOs:2, 3, 32, 34, 35, 39, 43, 44, 45, 80, 81, 82, 139, 140, 146, 147, 148, 149, 151, 152, 154, 155, 157, 158, 160, 161, 162, and 164 or the complete complement of SEQ ID NOs:2, 3, 32, 34, 35, 39, 43, 44, 45, 80, 81, 82, 139, 140, 146, 147, 148, 149, 151, 152, 154, 155, 157, 158, 160, 161, 162, and 164.
11. A vector containing a cDNA of claim 10.
12. A host cell containing the vector of claim 11.
13. A method for producing a protein, the method comprising the steps of: a) culturing the host cell of claim 12 under conditions for expression of protein; and b) recovering the protein so produced from the host cell culture.
14. A combination comprising a plurality of proteins wherein the proteins are SEQ ID NOs: 171-296 that are differentially expressed in respiratory disorders.
15. The combination of claim 14, wherein the proteins are attached to a substrate.
16. A method for using a combination of proteins to diagnose a respiratory disorder comprising: a) performing an assay to quantify the expression of the proteins of the combination of claim 14 in a sample; b) comparing the expression of the proteins of the combination to standards, thereby diagnosing a respiratory disorder.
17. The method of claim 16 wherein the respiratory disorder is lung cancer, chronic obstructive pulmonary disease, emphysema, or asthma.
18. A method for using a protein to screen a plurality of molecules or compounds to identify at least one ligand which specifically binds the protein, the method comprising: a) combining the substrate of claim 15 with the plurality of molecules or compounds under conditions to allow specific binding; and b) detecting specific binding between the protein and a molecule or compound, thereby identifying a ligand which specifically binds the protein.
19. The method of claim 18 wherein the plurality of molecules or compounds is selected from agonists, antagonists, antibodies, DNA molecules, small molecule drugs, immunoglobulins, inhibitors, mimetics, peptide nucleic acids, peptides, pharmaceutical agents, proteins, RNA molecules, and ribozymes.
20. A purified protein comprising an amino acid sequence of SEQ ID NO: 174.
21. An isolated antibody which specifically binds to the protein of claim 20.
22. The antibody of claim 21, wherein the antibody is selected from a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a recombinant antibody, a humanized antibody, a single chain antibody, a Fab fragment, an F(ab')2 fragment, an Fv fragment; and an antibody-peptide fusion protein.
23. A small drug molecule which specifically binds to a protein of claim 20.
24. A method for using a protein to diagnose a respiratory disorder comprising: a) performing an assay to quantify the expression of the protein of claim 20 in a sample; b) comparing the expression of the protein to standards, thereby diagnosing a respiratory disorder.
25. A method of using a protein to prepare and purify a polyclonal antibody comprising: a) immunizing a animal with a protein of claim 20 under conditions to elicit an antibody response; b) isolating animal antibodies; c) attaching the protein to a substrate; d) contacting the substrate with isolated antibodies under conditions to allow specific binding to the protein; e) dissociating the antibodies from the protein, thereby obtaining purified polyclonal antibodies.
26. A polyclonal antibody produced by the method of claim 25.
27. A method of using a protein to prepare a monoclonal antibody comprising: a) immunizing a animal with a protein of claim 20 under conditions to elicit an antibody response; b) isolating antibody-producing cells from the animal; c) fusing the antibody-producing cells with immortalized cells in culture to form monoclonal antibody producing hybridoma cells; d) culturing the hybridoma cells; and e) isolating monoclonal antibodies from culture.
28. A monoclonal antibody produced by the method of claim 27.
29. A method for using an antibody to detect expression of a protein in a sample, the method comprising: a) combining the antibody of claim 21 with a sample under conditions that allow the formation of antibody:protein complexes; and b) detecting complex formation, wherein complex formation indicates expression of the protein in the sample.
30. The method of claim 29 wherein the antibody is immobilized on a substrate.
31. The method of claim 29 wherein complex formation is compared with standards and is diagnostic of a respiratory disorder.
32. A composition comprising the antibody of claim 21 and a labeling moiety.
33. A composition comprising the antibody of claim 21 and a pharmaceutical agent.
34. An antagonist which specifically binds the protein of claim 20.
35. An agonist which specifically binds the protein of claim 20.
36. The combination of claim 1, wherein the cDNAs are SEQ ID NOs:38-107 that are differentially expressed at least 4-fold in respiratory disorders and the complements of SEQ ID NOs:38- 107.
PCT/US2003/017409 2002-06-04 2003-06-02 Diagnostics markers for lung cancer WO2003101283A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003240495A AU2003240495A1 (en) 2002-06-04 2003-06-02 Diagnostics markers for lung cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US38600502P 2002-06-04 2002-06-04
US60/386,005 2002-06-04

Publications (2)

Publication Number Publication Date
WO2003101283A2 true WO2003101283A2 (en) 2003-12-11
WO2003101283A3 WO2003101283A3 (en) 2004-12-09

Family

ID=29712224

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/017409 WO2003101283A2 (en) 2002-06-04 2003-06-02 Diagnostics markers for lung cancer

Country Status (2)

Country Link
AU (1) AU2003240495A1 (en)
WO (1) WO2003101283A2 (en)

Cited By (93)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005046720A2 (en) * 2003-11-12 2005-05-26 Children's Hospital Medical Center Diagnosis, prognosis and treatment of pulmonary diseases using foxa2
WO2009052390A1 (en) * 2007-10-19 2009-04-23 Abbott Laboratories Glycosylated mammalian ngal and use thereof
US7626015B2 (en) 2006-06-09 2009-12-01 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801L
US7723052B2 (en) 2006-05-11 2010-05-25 Quark Pharmaceuticals, Inc. Screening systems utilizing RTP801
US7741299B2 (en) 2004-08-16 2010-06-22 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
EP2260858A2 (en) 2003-11-06 2010-12-15 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
WO2011000054A1 (en) 2009-07-03 2011-01-06 Avipep Pty Ltd Immuno-conjugates and methods for producing them
US7872119B2 (en) 2007-02-26 2011-01-18 Quark Pharmaceuticals, Inc. Inhibitors of RTP801 and their use in disease treatment
EP2286844A2 (en) 2004-06-01 2011-02-23 Genentech, Inc. Antibody-drug conjugates and methods
WO2011027132A1 (en) 2009-09-03 2011-03-10 Cancer Research Technology Limited Clec14a inhibitors
WO2011031870A1 (en) 2009-09-09 2011-03-17 Centrose, Llc Extracellular targeted drug conjugates
WO2011056983A1 (en) 2009-11-05 2011-05-12 Genentech, Inc. Zirconium-radiolabeled, cysteine engineered antibody conjugates
EP2333112A2 (en) 2004-02-20 2011-06-15 Veridex, LLC Breast cancer prognostics
WO2011075786A1 (en) 2009-12-23 2011-06-30 Avipep Pty Ltd Immuno-conjugates and methods for producing them 2
US8029986B2 (en) 2004-10-21 2011-10-04 Helmholtz Zentrum Muenchen Deutsches Forschungszentrum fuer Gesundheit und Umwelt(GmbH) KASPP (LRRK2) gene, its production and use for the detection and treatment of neurodegenerative disorders
WO2011130598A1 (en) 2010-04-15 2011-10-20 Spirogen Limited Pyrrolobenzodiazepines and conjugates thereof
US8067570B2 (en) 2006-01-20 2011-11-29 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
WO2011156328A1 (en) 2010-06-08 2011-12-15 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2012074757A1 (en) 2010-11-17 2012-06-07 Genentech, Inc. Alaninyl maytansinol antibody conjugates
US8227194B2 (en) * 2005-04-08 2012-07-24 University Of Maryland, Baltimore Monoclonal antibodies with binding specificity for response gene to complement 32 (RGC-32)
WO2012123295A1 (en) * 2011-03-11 2012-09-20 Roche Diagnostics Gmbh Fen1 as marker for chronic obstructive pulmonary disease (copd)
WO2012142662A1 (en) 2011-04-21 2012-10-26 Garvan Institute Of Medical Research Modified variable domain molecules and methods for producing and using them b
WO2012155019A1 (en) 2011-05-12 2012-11-15 Genentech, Inc. Multiple reaction monitoring lc-ms/ms method to detect therapeutic antibodies in animal samples using framework signature pepides
WO2013130093A1 (en) 2012-03-02 2013-09-06 Genentech, Inc. Biomarkers for treatment with anti-tubulin chemotherapeutic compounds
WO2014057074A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014140862A2 (en) 2013-03-13 2014-09-18 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
US8846036B2 (en) 2007-10-19 2014-09-30 Abbott Laboratories Antibodies that bind to mammalian NGAL and uses thereof
WO2014159981A2 (en) 2013-03-13 2014-10-02 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
US8911732B2 (en) 2010-12-20 2014-12-16 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
CN104297490A (en) * 2007-09-11 2015-01-21 癌症预防和治疗有限公司 Identification of proteins in human serum indicative of pathologies of human lung tissues
WO2015023355A1 (en) 2013-08-12 2015-02-19 Genentech, Inc. 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
WO2015095212A1 (en) 2013-12-16 2015-06-25 Genentech, Inc. 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
WO2015095223A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015095227A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
US9212228B2 (en) 2005-11-24 2015-12-15 Ganymed Pharmaceuticals Ag Monoclonal antibodies against claudin-18 for treatment of cancer
US9228011B2 (en) 2011-01-17 2016-01-05 Lykera Biomed Sa Antibodies against the S100P protein for the treatment and diagnosis of cancer
WO2016037644A1 (en) 2014-09-10 2016-03-17 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2016040856A2 (en) 2014-09-12 2016-03-17 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2016040825A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anthracycline disulfide intermediates, antibody-drug conjugates and methods
WO2016090050A1 (en) 2014-12-03 2016-06-09 Genentech, Inc. Quaternary amine compounds and antibody-drug conjugates thereof
EP3088004A1 (en) 2004-09-23 2016-11-02 Genentech, Inc. Cysteine engineered antibodies and conjugates
US9512232B2 (en) 2012-05-09 2016-12-06 Ganymed Pharmaceuticals Ag Antibodies against Claudin 18.2 useful in cancer diagnosis
WO2017059289A1 (en) 2015-10-02 2017-04-06 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
US9617336B2 (en) 2012-02-01 2017-04-11 Compugen Ltd C10RF32 antibodies, and uses thereof for treatment of cancer
WO2017064675A1 (en) 2015-10-16 2017-04-20 Genentech, Inc. Hindered disulfide drug conjugates
WO2017068511A1 (en) 2015-10-20 2017-04-27 Genentech, Inc. Calicheamicin-antibody-drug conjugates and methods of use
WO2017165734A1 (en) 2016-03-25 2017-09-28 Genentech, Inc. Multiplexed total antibody and antibody-conjugated drug quantification assay
US9775785B2 (en) 2004-05-18 2017-10-03 Ganymed Pharmaceuticals Ag Antibody to genetic products differentially expressed in tumors and the use thereof
EP3235820A1 (en) 2014-09-17 2017-10-25 Genentech, Inc. Pyrrolobenzodiazepines and antibody disulfide conjugates thereof
WO2017201449A1 (en) 2016-05-20 2017-11-23 Genentech, Inc. Protac antibody conjugates and methods of use
WO2017205741A1 (en) 2016-05-27 2017-11-30 Genentech, Inc. Bioanalytical method for the characterization of site-specific antibody-drug conjugates
WO2017214024A1 (en) 2016-06-06 2017-12-14 Genentech, Inc. Silvestrol antibody-drug conjugates and methods of use
WO2018031662A1 (en) 2016-08-11 2018-02-15 Genentech, Inc. Pyrrolobenzodiazepine prodrugs and antibody conjugates thereof
US9919056B2 (en) 2012-10-12 2018-03-20 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US9931415B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US9931414B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
WO2018065501A1 (en) 2016-10-05 2018-04-12 F. Hoffmann-La Roche Ag Methods for preparing antibody drug conjugates
US9950078B2 (en) 2013-10-11 2018-04-24 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US9956299B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepine—antibody conjugates
CN108181457A (en) * 2009-03-12 2018-06-19 癌症预防和治疗有限公司 Identification, assessment, prevention and the method and kit for the treatment of lung disease
US10010624B2 (en) 2013-10-11 2018-07-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10029018B2 (en) 2013-10-11 2018-07-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10098934B2 (en) 2007-09-04 2018-10-16 Compugen Ltd Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
WO2019060398A1 (en) 2017-09-20 2019-03-28 Ph Pharma Co., Ltd. Thailanstatin analogs
US10392393B2 (en) 2016-01-26 2019-08-27 Medimmune Limited Pyrrolobenzodiazepines
US10414824B2 (en) 2002-11-22 2019-09-17 Ganymed Pharmaceuticals Ag Genetic products differentially expressed in tumors and the use thereof
US10420777B2 (en) 2014-09-12 2019-09-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10543279B2 (en) 2016-04-29 2020-01-28 Medimmune Limited Pyrrolobenzodiazepine conjugates and their use for the treatment of cancer
US10544223B2 (en) 2017-04-20 2020-01-28 Adc Therapeutics Sa Combination therapy with an anti-axl antibody-drug conjugate
WO2020049286A1 (en) 2018-09-03 2020-03-12 Femtogenix Limited Polycyclic amides as cytotoxic agents
WO2020086858A1 (en) 2018-10-24 2020-04-30 Genentech, Inc. Conjugated chemical inducers of degradation and methods of use
WO2020123275A1 (en) 2018-12-10 2020-06-18 Genentech, Inc. Photocrosslinking peptides for site specific conjugation to fc-containing proteins
US10695433B2 (en) 2012-10-12 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10695439B2 (en) 2016-02-10 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine conjugates
WO2020157491A1 (en) 2019-01-29 2020-08-06 Femtogenix Limited G-a crosslinking cytotoxic agents
US10736903B2 (en) 2012-10-12 2020-08-11 Medimmune Limited Pyrrolobenzodiazepine-anti-PSMA antibody conjugates
US10751346B2 (en) 2012-10-12 2020-08-25 Medimmune Limited Pyrrolobenzodiazepine—anti-PSMA antibody conjugates
US10780096B2 (en) 2014-11-25 2020-09-22 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US10799595B2 (en) 2016-10-14 2020-10-13 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11059893B2 (en) 2015-04-15 2021-07-13 Bergenbio Asa Humanized anti-AXL antibodies
US11135303B2 (en) 2011-10-14 2021-10-05 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US11160872B2 (en) 2017-02-08 2021-11-02 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
WO2022023735A1 (en) 2020-07-28 2022-02-03 Femtogenix Limited Cytotoxic agents
US11318211B2 (en) 2017-06-14 2022-05-03 Adc Therapeutics Sa Dosage regimes for the administration of an anti-CD19 ADC
US11352324B2 (en) 2018-03-01 2022-06-07 Medimmune Limited Methods
US11370801B2 (en) 2017-04-18 2022-06-28 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11517626B2 (en) 2016-02-10 2022-12-06 Medimmune Limited Pyrrolobenzodiazepine antibody conjugates
US11524969B2 (en) 2018-04-12 2022-12-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof as antitumour agents
US11612665B2 (en) 2017-02-08 2023-03-28 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11649250B2 (en) 2017-08-18 2023-05-16 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11702473B2 (en) 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates
US11769596B2 (en) 2017-04-04 2023-09-26 Lung Cancer Proteomics Llc Plasma based protein profiling for early stage lung cancer diagnosis

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2836855C (en) 2011-06-30 2020-07-14 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
US9567340B2 (en) 2012-12-21 2017-02-14 Medimmune Limited Unsymmetrical pyrrolobenzodiazepines-dimers for use in the treatment of proliferative and autoimmune diseases
ES2658888T5 (en) 2012-12-21 2021-10-19 Medimmune Ltd Pyrrolobenzodiazepines and their conjugates

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DATABASE GENBANK [Online] HATTORI M. ET AL: 'Homo sapiens genomic DNA, chromosome 11q, clone:CTD-3161I21, complete sequence', XP002903255 Retrieved from NCBI Database accession no. (AP003166) *

Cited By (139)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10414824B2 (en) 2002-11-22 2019-09-17 Ganymed Pharmaceuticals Ag Genetic products differentially expressed in tumors and the use thereof
EP3858387A1 (en) 2003-11-06 2021-08-04 Seagen Inc. Monomethylvaline compounds capable of conjugation to ligands
EP2478912A1 (en) 2003-11-06 2012-07-25 Seattle Genetics, Inc. Auristatin conjugates with anti-HER2 or anti-CD22 antibodies and their use in therapy
EP3120861A1 (en) 2003-11-06 2017-01-25 Seattle Genetics, Inc. Intermediate for conjugate preparation comprising auristatin derivatives and a linker
EP2486933A1 (en) 2003-11-06 2012-08-15 Seattle Genetics, Inc. Monomethylvaline compounds conjugated with antibodies
EP2489364A1 (en) 2003-11-06 2012-08-22 Seattle Genetics, Inc. Monomethylvaline compounds onjugated to antibodies
EP3434275A1 (en) 2003-11-06 2019-01-30 Seattle Genetics, Inc. Assay for cancer cells based on the use of auristatin conjugates with antibodies
EP2260858A2 (en) 2003-11-06 2010-12-15 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
WO2005046720A2 (en) * 2003-11-12 2005-05-26 Children's Hospital Medical Center Diagnosis, prognosis and treatment of pulmonary diseases using foxa2
WO2005046720A3 (en) * 2003-11-12 2005-10-06 Childrens Hosp Medical Center Diagnosis, prognosis and treatment of pulmonary diseases using foxa2
EP2333112A2 (en) 2004-02-20 2011-06-15 Veridex, LLC Breast cancer prognostics
US9775785B2 (en) 2004-05-18 2017-10-03 Ganymed Pharmaceuticals Ag Antibody to genetic products differentially expressed in tumors and the use thereof
EP2286844A2 (en) 2004-06-01 2011-02-23 Genentech, Inc. Antibody-drug conjugates and methods
US8642571B2 (en) 2004-08-06 2014-02-04 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
US8168607B2 (en) 2004-08-06 2012-05-01 Quark Pharmaceuticals Inc. Methods of treating eye diseases in diabetic patients
EP2319925A2 (en) 2004-08-16 2011-05-11 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
US7741299B2 (en) 2004-08-16 2010-06-22 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
EP3088004A1 (en) 2004-09-23 2016-11-02 Genentech, Inc. Cysteine engineered antibodies and conjugates
US8409809B2 (en) 2004-10-21 2013-04-02 Helmholtz Zentrum München Deutsches Forschungszentrum Für Gesundheit Und Umwelt (Gmbh) KASPP (LRRK2) gene, its production and use for the detection and treatment of neurodegenerative disorders
EP2316927A3 (en) * 2004-10-21 2011-10-19 Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) KASPP (LRRK2) gene, its production and use for the detection and treatment of neurodegenerative disorders
US8029986B2 (en) 2004-10-21 2011-10-04 Helmholtz Zentrum Muenchen Deutsches Forschungszentrum fuer Gesundheit und Umwelt(GmbH) KASPP (LRRK2) gene, its production and use for the detection and treatment of neurodegenerative disorders
US8227194B2 (en) * 2005-04-08 2012-07-24 University Of Maryland, Baltimore Monoclonal antibodies with binding specificity for response gene to complement 32 (RGC-32)
US9499609B2 (en) 2005-11-24 2016-11-22 Ganymed Pharmaceuticals Ag Monoclonal antibodies against claudin-18 for treatment of cancer
US10174104B2 (en) 2005-11-24 2019-01-08 Ganymed Pharmaceuticals Gmbh Monoclonal antibodies against claudin-18 for treatment of cancer
US9212228B2 (en) 2005-11-24 2015-12-15 Ganymed Pharmaceuticals Ag Monoclonal antibodies against claudin-18 for treatment of cancer
US10017564B2 (en) 2005-11-24 2018-07-10 Ganymed Pharmaceuticals Gmbh Monoclonal antibodies against claudin-18 for treatment of cancer
US10738108B2 (en) 2005-11-24 2020-08-11 Astellas Pharma Inc. Monoclonal antibodies against claudin-18 for treatment of cancer
US11739139B2 (en) 2005-11-24 2023-08-29 Astellas Pharma Inc. Monoclonal antibodies against Claudin-18 for treatment of cancer
US9751934B2 (en) 2005-11-24 2017-09-05 Ganymed Pharmaceuticals Ag Monoclonal antibodies against claudin-18 for treatment of cancer
EP2402443A2 (en) 2006-01-20 2012-01-04 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of rtp801
US8067570B2 (en) 2006-01-20 2011-11-29 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
US9056903B2 (en) 2006-01-20 2015-06-16 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801
US7723052B2 (en) 2006-05-11 2010-05-25 Quark Pharmaceuticals, Inc. Screening systems utilizing RTP801
US8344104B2 (en) 2006-05-11 2013-01-01 Quark Pharmaceuticals, Inc. Screening systems utilizing RTP801
US8034575B2 (en) 2006-05-11 2011-10-11 Quark Pharmaceuticals, Inc. Screening systems utilizing RTP801
US8017764B2 (en) 2006-06-09 2011-09-13 Quark Pharmaceuticals Inc. Therapeutic uses of inhibitors of RTP801L
US7626015B2 (en) 2006-06-09 2009-12-01 Quark Pharmaceuticals, Inc. Therapeutic uses of inhibitors of RTP801L
US7872119B2 (en) 2007-02-26 2011-01-18 Quark Pharmaceuticals, Inc. Inhibitors of RTP801 and their use in disease treatment
US10098934B2 (en) 2007-09-04 2018-10-16 Compugen Ltd Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
CN104297490A (en) * 2007-09-11 2015-01-21 癌症预防和治疗有限公司 Identification of proteins in human serum indicative of pathologies of human lung tissues
WO2009052390A1 (en) * 2007-10-19 2009-04-23 Abbott Laboratories Glycosylated mammalian ngal and use thereof
US8846036B2 (en) 2007-10-19 2014-09-30 Abbott Laboratories Antibodies that bind to mammalian NGAL and uses thereof
CN108181457A (en) * 2009-03-12 2018-06-19 癌症预防和治疗有限公司 Identification, assessment, prevention and the method and kit for the treatment of lung disease
US11474104B2 (en) 2009-03-12 2022-10-18 Cancer Prevention And Cure, Ltd. Methods of identification, assessment, prevention and therapy of lung diseases and kits thereof including gender-based disease identification, assessment, prevention and therapy
WO2011000054A1 (en) 2009-07-03 2011-01-06 Avipep Pty Ltd Immuno-conjugates and methods for producing them
JP2013503842A (en) * 2009-09-03 2013-02-04 キャンサー リサーチ テクノロジー リミテッド CLEC14A inhibitor
WO2011027132A1 (en) 2009-09-03 2011-03-10 Cancer Research Technology Limited Clec14a inhibitors
US9255148B2 (en) 2009-09-03 2016-02-09 Cancer Research Technology Limited CLEC14A inhibitors
WO2011031870A1 (en) 2009-09-09 2011-03-17 Centrose, Llc Extracellular targeted drug conjugates
WO2011056983A1 (en) 2009-11-05 2011-05-12 Genentech, Inc. Zirconium-radiolabeled, cysteine engineered antibody conjugates
WO2011075786A1 (en) 2009-12-23 2011-06-30 Avipep Pty Ltd Immuno-conjugates and methods for producing them 2
WO2011130598A1 (en) 2010-04-15 2011-10-20 Spirogen Limited Pyrrolobenzodiazepines and conjugates thereof
WO2011156328A1 (en) 2010-06-08 2011-12-15 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2012074757A1 (en) 2010-11-17 2012-06-07 Genentech, Inc. Alaninyl maytansinol antibody conjugates
US9719996B2 (en) 2010-12-20 2017-08-01 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
US10022452B2 (en) 2010-12-20 2018-07-17 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
US8911732B2 (en) 2010-12-20 2014-12-16 Genentech, Inc. Anti-mesothelin antibodies and immunoconjugates
US9868784B2 (en) 2011-01-17 2018-01-16 Lykera Biomed Sa Antibodies against the S100P protein for the treatment and diagnosis of cancer
US9228011B2 (en) 2011-01-17 2016-01-05 Lykera Biomed Sa Antibodies against the S100P protein for the treatment and diagnosis of cancer
WO2012123295A1 (en) * 2011-03-11 2012-09-20 Roche Diagnostics Gmbh Fen1 as marker for chronic obstructive pulmonary disease (copd)
WO2012142662A1 (en) 2011-04-21 2012-10-26 Garvan Institute Of Medical Research Modified variable domain molecules and methods for producing and using them b
EP3103810A2 (en) 2011-04-21 2016-12-14 Garvan Institute of Medical Research Modified variable domain molecules and methods for producing and using them
WO2012155019A1 (en) 2011-05-12 2012-11-15 Genentech, Inc. Multiple reaction monitoring lc-ms/ms method to detect therapeutic antibodies in animal samples using framework signature pepides
US11135303B2 (en) 2011-10-14 2021-10-05 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9617336B2 (en) 2012-02-01 2017-04-11 Compugen Ltd C10RF32 antibodies, and uses thereof for treatment of cancer
WO2013130093A1 (en) 2012-03-02 2013-09-06 Genentech, Inc. Biomarkers for treatment with anti-tubulin chemotherapeutic compounds
US10053512B2 (en) 2012-05-09 2018-08-21 Ganymed Pharmaceuticals Ag Antibodies against claudin 18.2 useful in cancer diagnosis
US9512232B2 (en) 2012-05-09 2016-12-06 Ganymed Pharmaceuticals Ag Antibodies against Claudin 18.2 useful in cancer diagnosis
US9889207B2 (en) 2012-10-12 2018-02-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10736903B2 (en) 2012-10-12 2020-08-11 Medimmune Limited Pyrrolobenzodiazepine-anti-PSMA antibody conjugates
EP2839860A1 (en) 2012-10-12 2015-02-25 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
US10695433B2 (en) 2012-10-12 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11690918B2 (en) 2012-10-12 2023-07-04 Medimmune Limited Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US11701430B2 (en) 2012-10-12 2023-07-18 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10646584B2 (en) 2012-10-12 2020-05-12 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US11771775B2 (en) 2012-10-12 2023-10-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11779650B2 (en) 2012-10-12 2023-10-10 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10994023B2 (en) 2012-10-12 2021-05-04 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9919056B2 (en) 2012-10-12 2018-03-20 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US9931415B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US9931414B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10799596B2 (en) 2012-10-12 2020-10-13 Adc Therapeutics S.A. Pyrrolobenzodiazepine-antibody conjugates
WO2014057074A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
US10722594B2 (en) 2012-10-12 2020-07-28 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US10335497B2 (en) 2012-10-12 2019-07-02 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10751346B2 (en) 2012-10-12 2020-08-25 Medimmune Limited Pyrrolobenzodiazepine—anti-PSMA antibody conjugates
US10780181B2 (en) 2012-10-12 2020-09-22 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
WO2014159981A2 (en) 2013-03-13 2014-10-02 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014140862A2 (en) 2013-03-13 2014-09-18 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2015023355A1 (en) 2013-08-12 2015-02-19 Genentech, Inc. 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
US9950078B2 (en) 2013-10-11 2018-04-24 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10029018B2 (en) 2013-10-11 2018-07-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10010624B2 (en) 2013-10-11 2018-07-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US9956299B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepine—antibody conjugates
WO2015095223A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2015095212A1 (en) 2013-12-16 2015-06-25 Genentech, Inc. 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
WO2015095227A2 (en) 2013-12-16 2015-06-25 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
WO2016037644A1 (en) 2014-09-10 2016-03-17 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10188746B2 (en) 2014-09-10 2019-01-29 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10420777B2 (en) 2014-09-12 2019-09-24 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2016040856A2 (en) 2014-09-12 2016-03-17 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2016040825A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anthracycline disulfide intermediates, antibody-drug conjugates and methods
EP3235820A1 (en) 2014-09-17 2017-10-25 Genentech, Inc. Pyrrolobenzodiazepines and antibody disulfide conjugates thereof
US10780096B2 (en) 2014-11-25 2020-09-22 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
WO2016090050A1 (en) 2014-12-03 2016-06-09 Genentech, Inc. Quaternary amine compounds and antibody-drug conjugates thereof
US11702473B2 (en) 2015-04-15 2023-07-18 Medimmune Limited Site-specific antibody-drug conjugates
US11059893B2 (en) 2015-04-15 2021-07-13 Bergenbio Asa Humanized anti-AXL antibodies
WO2017059289A1 (en) 2015-10-02 2017-04-06 Genentech, Inc. Pyrrolobenzodiazepine antibody drug conjugates and methods of use
WO2017064675A1 (en) 2015-10-16 2017-04-20 Genentech, Inc. Hindered disulfide drug conjugates
WO2017068511A1 (en) 2015-10-20 2017-04-27 Genentech, Inc. Calicheamicin-antibody-drug conjugates and methods of use
US10392393B2 (en) 2016-01-26 2019-08-27 Medimmune Limited Pyrrolobenzodiazepines
US10695439B2 (en) 2016-02-10 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11517626B2 (en) 2016-02-10 2022-12-06 Medimmune Limited Pyrrolobenzodiazepine antibody conjugates
EP4273551A2 (en) 2016-03-25 2023-11-08 F. Hoffmann-La Roche AG Multiplexed total antibody and antibody-conjugated drug quantification assay
WO2017165734A1 (en) 2016-03-25 2017-09-28 Genentech, Inc. Multiplexed total antibody and antibody-conjugated drug quantification assay
US10543279B2 (en) 2016-04-29 2020-01-28 Medimmune Limited Pyrrolobenzodiazepine conjugates and their use for the treatment of cancer
WO2017201449A1 (en) 2016-05-20 2017-11-23 Genentech, Inc. Protac antibody conjugates and methods of use
WO2017205741A1 (en) 2016-05-27 2017-11-30 Genentech, Inc. Bioanalytical method for the characterization of site-specific antibody-drug conjugates
WO2017214024A1 (en) 2016-06-06 2017-12-14 Genentech, Inc. Silvestrol antibody-drug conjugates and methods of use
WO2018031662A1 (en) 2016-08-11 2018-02-15 Genentech, Inc. Pyrrolobenzodiazepine prodrugs and antibody conjugates thereof
WO2018065501A1 (en) 2016-10-05 2018-04-12 F. Hoffmann-La Roche Ag Methods for preparing antibody drug conjugates
US10799595B2 (en) 2016-10-14 2020-10-13 Medimmune Limited Pyrrolobenzodiazepine conjugates
US11160872B2 (en) 2017-02-08 2021-11-02 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
US11612665B2 (en) 2017-02-08 2023-03-28 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11813335B2 (en) 2017-02-08 2023-11-14 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11769596B2 (en) 2017-04-04 2023-09-26 Lung Cancer Proteomics Llc Plasma based protein profiling for early stage lung cancer diagnosis
US11370801B2 (en) 2017-04-18 2022-06-28 Medimmune Limited Pyrrolobenzodiazepine conjugates
US10544223B2 (en) 2017-04-20 2020-01-28 Adc Therapeutics Sa Combination therapy with an anti-axl antibody-drug conjugate
US11318211B2 (en) 2017-06-14 2022-05-03 Adc Therapeutics Sa Dosage regimes for the administration of an anti-CD19 ADC
US11649250B2 (en) 2017-08-18 2023-05-16 Medimmune Limited Pyrrolobenzodiazepine conjugates
WO2019060398A1 (en) 2017-09-20 2019-03-28 Ph Pharma Co., Ltd. Thailanstatin analogs
US11352324B2 (en) 2018-03-01 2022-06-07 Medimmune Limited Methods
US11524969B2 (en) 2018-04-12 2022-12-13 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof as antitumour agents
WO2020049286A1 (en) 2018-09-03 2020-03-12 Femtogenix Limited Polycyclic amides as cytotoxic agents
WO2020086858A1 (en) 2018-10-24 2020-04-30 Genentech, Inc. Conjugated chemical inducers of degradation and methods of use
WO2020123275A1 (en) 2018-12-10 2020-06-18 Genentech, Inc. Photocrosslinking peptides for site specific conjugation to fc-containing proteins
WO2020157491A1 (en) 2019-01-29 2020-08-06 Femtogenix Limited G-a crosslinking cytotoxic agents
WO2022023735A1 (en) 2020-07-28 2022-02-03 Femtogenix Limited Cytotoxic agents

Also Published As

Publication number Publication date
AU2003240495A8 (en) 2003-12-19
WO2003101283A3 (en) 2004-12-09
AU2003240495A1 (en) 2003-12-19

Similar Documents

Publication Publication Date Title
WO2003101283A2 (en) Diagnostics markers for lung cancer
US6727066B2 (en) Genes expressed in treated human C3A liver cell cultures
US20020156263A1 (en) Genes expressed in breast cancer
US20020137081A1 (en) Genes differentially expressed in vascular tissue activation
US20030190640A1 (en) Genes expressed in prostate cancer
US6673549B1 (en) Genes expressed in C3A liver cell cultures treated with steroids
US20020187472A1 (en) Steap-related protein
US20030134283A1 (en) Genes regulated in dendritic cell differentiation
US20030165924A1 (en) Genes expressed in foam cell differentiation
US20030065157A1 (en) Genes expressed in lung cancer
US20030194721A1 (en) Genes expressed in treated foam cells
US20030165864A1 (en) Genes regulated by DNA methylation in tumor cells
US20030166903A1 (en) Genes associated with vascular disease
US20050130171A1 (en) Genes expressed in Alzheimer&#39;s disease
US20030186333A1 (en) Down syndrome critical region 1-like protein
US20060275314A1 (en) Transmembrane protein differentially expressed in cancer
US20030119009A1 (en) Genes regulated by MYCN activation
US6544742B1 (en) Detection of genes regulated by EGF in breast cancer
US20030165854A1 (en) Marker genes responding to treatment with toxins
US20030211515A1 (en) Novel compounds
US20030124580A1 (en) Lung surfactant molecules
US20030175754A1 (en) RVP-1 variant differentially expressed in crohns disease
US6692923B2 (en) Tapasin-like protein
US20030104418A1 (en) Diagnostic markers for breast cancer
US20190309285A1 (en) Genes expressed in mental illness and mood disorders

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP