WO2003083074A2 - Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas - Google Patents

Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas Download PDF

Info

Publication number
WO2003083074A2
WO2003083074A2 PCT/US2003/009534 US0309534W WO03083074A2 WO 2003083074 A2 WO2003083074 A2 WO 2003083074A2 US 0309534 W US0309534 W US 0309534W WO 03083074 A2 WO03083074 A2 WO 03083074A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
gene
colon
ofthe
antigen
Prior art date
Application number
PCT/US2003/009534
Other languages
French (fr)
Other versions
WO2003083074A9 (en
Inventor
Karen Mclachlan
Dennis Gately
Original Assignee
Idec Pharmaceuticals 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 Idec Pharmaceuticals Corporation filed Critical Idec Pharmaceuticals Corporation
Priority to AU2003222103A priority Critical patent/AU2003222103A1/en
Priority to US10/509,131 priority patent/US20060089493A1/en
Publication of WO2003083074A2 publication Critical patent/WO2003083074A2/en
Publication of WO2003083074A9 publication Critical patent/WO2003083074A9/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/16Primer sets for multiplex assays

Definitions

  • the present invention relates the identification of gene targets for treatment and diagnosis of neoplastic diseases, such as colon or colorectal cancer, and other cancers wherein the subject genes are upregulated and the use thereof to express the corresponding antigen, and to produce ligands that specifically bind such antigen, e.g. monoclonal antibodies and small molecules.
  • Colorectal cancers are among the most common cancers in men and women in the U.S. and are one ofthe leading causes of death. Other than surgical resection no other systemic or adjuvant therapy is available. Vogelstein and colleagues have described the sequence of genetic events that appear to be associated with the multistep process of colon cancer development in humans (Fearon and Vogelstein, 1990). An understanding ofthe molecular genetics of carcinogenesis, however, has not led to preventative or therapeutic measures. It can be expected that advances in molecular genetics will lead to better risk assessment and early diagnosis but colorectal cancers will remain a deadly disease for a majority of patients due to the lack of an adjuvant therapy.
  • Endogenous gastrins and exogenous gastrins seem to promote the growth of established colon cancers in mice (Singh, et al., 1986; Singh, et al., 1987; et al., 1984; Smith and Solomon, 1988; Singh, et al., 1990; Rehfeld and van Solinge, 1994) and promote carcinogen induced colon cancers in rats (Williamson et al., 1978; Karlin et al., 1985; Lamoste and Willems; 1988). Recent studies of Montag et al (1993) further support a possible co-carcinogenic role of gastrin in the initiation of tumors.
  • the MDR gene products actively transport the toxic substances out ofthe cell before the chemotherapeutic agents can damage the DNA machinery ofthe cell. These toxic substances harm the normal cell populations more than they harm the colon cancer cells for the above reasons.
  • growth promoting factors such as EGF, estradiol, IGF-II
  • this knowledge has not been applied and therefore the treatment outcome for colon cancers remains bleak.
  • Antisense RNA technology has been developed as an approach to inhibiting gene expression, including oncogene expression.
  • An "antisense" RNA molecule is one which contains the complement of, and can therefore hybridize with, protein-encoding RNAs ofthe cell. It is believed that the hybridization of antisense RNA to its cellular RNA complement can prevent expression ofthe cellular RNA, perhaps by limiting its franslatability.
  • a principle application of antisense RNA technology has been in connection with attempts to affect the expression of specific genes. For example, Delauney, et al. have reported the use antisense transcripts to inhibit gene expression in transgenic plants (Delauney, et al., 1988). These authors report the down-regulation of chloramphenicol acetyl transferase activity in tobacco plants transformed with CAT sequences through the application of antisense technology.
  • Antisense technology has also been applied in attempts to inhibit the expression of various oncogenes.
  • Kasid, et al., 1989 report the preparation of recombinant vector construct employing Craf-1 cDNA fragments in an antisense orientation, brought under the control of an adenoviras 2 late promoter.
  • These authors report that the introduction of this recombinant construct into a human squamous carcinoma resulted in a greatly reduced tumorigenic potential relative to cells transfected faith control sense transfectants.
  • Prochownik, et al., 1988 have reported the use of Cmiyc antisense constructs to accelerate differentiation and inhibit G.sub.l progression in Friend Murine Erythroleukemia cells.
  • Khokha, et al., 1989 discloses the use of antisense RNAs to confer oncogenicity on 3T3 cells, through the use of antisense RNA to reduce murine tissue inhibitor or metalloproteinases levels.
  • Antisense methodology takes advantage ofthe fact that nucleic acids tend to pair with "complementary" sequences.
  • complementary it is meant that polynucleotides are those which are capable of base-pairing according to the standard Watson-Crick complementary rales. That is, the larger purines base pair with the smaller pyrimidines to form combinations of guanine paired with cytosine (G:C) and adenine paired with either thymine (A:T) in the case of DNA, or adenine paired with uracil (A:U) in the case of RNA. Inclusion of less common bases such as inosine, 5-methylcytosine, 6-methyladenine, hypoxanthine and others in hybridizing sequences does not interfere with pairing.
  • Antisense polynucleotides when introduced into a target cell, specifically bind to their target polynucleotide and interfere with transcription, RNA processing, transport, translation and/or stability.
  • Antisense RNA constructs, or DNA encoding such antisense RNAs can be employed to inhibit gene transcription or translation or both within a host cell, either in vitro or in vivo, such as within a host animal, including a human subject.
  • expression vector or construct is meant to include any type of genetic construct containing a nucleic acid coding for a gene product in which part or all ofthe nucleic acid encoding sequence is capable of being transcribed.
  • the transcript can be translated into a protein but it need not be.
  • expression includes both transcription of a gene and translation of mRNA into a gene product. In other embodiments, expression only includes transcription ofthe nucleic acid encoding a gene of interest.
  • the nucleic acid encoding a gene product is under transcriptional control of a promoter.
  • a “promoter” refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a gene.
  • under transcriptional control means that the promoter is in the correct location and orientation in relation to the nucleic acid to control RNA polymerase initiation and expression ofthe gene.
  • promoter is used to refer to a group of transcriptional control modules that are clustered around the initiation site for RNA polymerase II.
  • Much ofthe thinking about how promoters are organized derives from analyses of several viral promoters, including those for the HSV thymidine kinase (tk) and SV40 early transcription units. These studies, augmented by more recent work, have shown that promoters are composed of discrete functional modules, each consisting of approximately 7-20 base pairs of DNA, and containing one or more recognition sites for transcriptional activator or repressor proteins. At least one module in each promoter functions to position the start site for RNA synthesis.
  • TATA box in some promoters lacking a TATA box, such as the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the SV40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation.
  • promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 base pairs upstream ofthe start site, although a number of promoters have recently been shown to contain functional elements downstream ofthe start site as well.
  • the spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the tk promoter, the spacing between promoter elements can be increased to 50 base pairs apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription.
  • a promoter is selected based on its capability to direct gene expression in the targeted cell.
  • the nucleic acid coding region can be positioned adjacent to and under the control of a promoter that is capable of being expressed in a human cell.
  • a promoter might include either a human or viral promoter.
  • the human cytomegalovirus (CMV) immediate early gene promoter, the S V40 early promoter and the Rous sarcoma virus long terminal repeat can be used to obtain high-level expression ofthe gene of interest.
  • CMV cytomegalovirus
  • the use of other viral or mammalian cellular or bacterial phage promoters which are well known in the art to achieve expression of a gene of interest is contemplated as well, provided that the levels of expression are sufficient for a given purpose.
  • a promoter with well-known properties, the level and pattern of expression ofthe gene product following transfection can be optimized. Further, selection of a promoter that is regulated in response to specific physiologic signals can permit inducible expression ofthe gene product.
  • Representative elements/promoters useful in accordance with the present invention include but are not limited to those listed below.
  • Enhancers were originally detected as genetic elements that increased transcription from a promoter located at a distant position on the same molecule of DNA. This ability to act over a large distance had little precedent in classic studies of prokaryotic transcriptional regulation. Subsequent work showed that regions of DNA with enhancer activity are organized much like promoters. That is, they are composed of many individual elements, each of which binds to one or more transcriptional proteins.
  • enhancers The basic distinction between enhancers and promoters is operational.
  • An enhancer region as a whole must be able to stimulate transcription at a distance; this need not be true of a promoter region or its component elements.
  • a promoter includes one or more elements that direct initiation of RNA synthesis at a particular site and in a particular orientation, whereas enhancers lack these specificities. Promoters and enhancers are often overlapping and contiguous, often seeming to have a very similar modular organization.
  • Viral promoters cellular promoters/enhancers and inducible promoters/enhancers that could be used in combination with the nucleic acid encoding a gene of interest in an expression construct.
  • enhancers include Immunoglobulin Heavy Chain; Immunoglobulin Light Chain; T-Cell Receptor; HLA DQ a and DQ b b-Interferon; Interleukin-2; Interleukin-2 Receptor: Gibbon Ape Leukemia Virus; MHC Class II 5 or HLA- DRa; b-Actin; Muscle Creatine Kinase; Prealbumin (Transthyretin); Elastase I; Metallothionein; Collagenase, Albumin Gene; ⁇ -Fetoprotein; ⁇ -Globin; ⁇ -Globin; c-fos: c- HA-ras; Insulin Neural Cell Adhesion Molecule (NCAM); al-Antitrypsin; H2B
  • Inducers such as phorbol ester (TFA) heavy metals; glucocorticoids; poly (rl)X; poly(rc); Ela; H 2 O 2 ; IL 1 ; Interferon, Newcastle Disease Vims; A23187; IL-6; Serum; SV40 Large T Antigen; FMA; thyroid Hormone; could be used. Additionally, any promoter/enhancer combination (as per the Eukaryotic Promoter Data Base EPDB) could also be used to drive expression ofthe gene. Eukaryotic cells can support cytoplasmic transcription from certain bacterial promoters if the appropriate bacterial polymerase is provided, either as part ofthe delivery complex or as an additional genetic expression construct.
  • TFA phorbol ester
  • the expression construct can comprise a virus or engineered construct derived from a viral genome.
  • viruses to enter cells via receptor-mediated endocytosis and to integrate into host cell genome and express viral genes stably and efficiently have made them attractive candidates for the transfer of foreign genes into mammalian cells (Ridgeway, 1988; Nicolas and Rubenstein, 1988; Baichwal et al., 1986: Temin, 1986).
  • the first viruses used as gene vectors were DNA viruses including the papo viruses (simian virus 40, bovine papilloma virus, and polyoma) (Ridgeway, 1988; Baichwal et al., 1986) and adenovirases (Ridgeway, 1988; Baichwal et al., 1986). These have a relatively low capacity for foreign DNA sequences and have a restricted host spectrum. Furthermore, their oncogenic potential and cytopathic effects in permissive cells raise safety concerns. They can accommodate only up to 8 kB of foreign genetic material but can be readily introduced in a variety of cell lines and laboratory animals (Nicolas and Rubenstein, 1988; Temin, 1986).
  • a polyadenylation signal is typically inserted to effect proper polyadenylation ofthe gene transcript.
  • Any suitable polyadenylation sequence can be used.
  • An expression cassette can also include a terminator sequence. These elements enhance message levels and minimize read through from the cassette into other sequences.
  • a coding sequence under the control of a promoter, or operatively linking a sequence to a promoter, one positions the 5' end ofthe transcription initiation site ofthe transcriptional reading frame ofthe protein between about land about 50 nucleotides "downstream" of (i.e., 3' of) the chosen promoter.
  • an appropriate polyadenylation site e.g., 5'- AATAAA-3' (SEQ ID NO:66)
  • the poly A addition site is placed about 30 to 2000 nucleotides "downstream" of the termination site ofthe protein at a position prior to transcription termination.
  • ligands that bind antigens expressed by certain cancers, such as colon or colorectal cancers.
  • Representative ligands include monoclonal antibodies.
  • ligands for example, monoclonal antibodies that specifically bind novel antigens that are expressed by certain cancer tissues including colon cancer
  • a ligand e.g., monoclonal antibody that specifically binds to an antigen expressed by certain colon cancers
  • a detectable label e.g., a radiolabel or fluorophore.
  • colon cancer that comprise DNA primers or probes specific for novel gene targets expressed by colon cancers, and a detectable label, e.g. radiolabel or fluorophore.
  • Figure 1 summarizes expression data for the CICO1, CICO2 and CICO3, which were identified based on overexpression in colon cancer as described in Example 1.
  • Figures 2-5 depict gene expression profiles determined using the Gene Logic datasuite as described in Example 2. The values along the y-axis represent expression intensities in Gene Logic units. Each blue circle represents an individual patient sample. The bar graph on the left ofthe figure depicts the percentage of each tissue type found to express the gene fragment.
  • colon tumor, tumor % above 50 refers to tumor samples for which at least 50% of each sample comprises malignant tissue, as determined by a pathologist. This sample set is a subset of colon tumors, which comprises all colon tumor samples contained within the Gene Logic database.
  • Figure 2 depicts the gene expression profile of Candidate 1, which was determined using the Gene Logic datasuite for GENBANK Accession No. W91975 as described in Example 2.
  • Candidate 1 is overexpressed in colon tumor tissue.
  • Figure 3 depicts the gene expression profile of Candidate 2, which was determined using the Gene Logic datasuite for GENBANK Accession No. Al 694242 as described in Example 2.
  • Candidate 2 is overexpressed in colon tumor tissue.
  • Figure 4 contains the gene expression profile of Candidate 3, which was determined using the Gene Logic datasuite for GENBANK Accession No. AI680111 as described in Example 2.
  • Candidate 3 is overexpressed in colon tumor tissue.
  • Figure 5 depicts the gene expression profile of Candidate 4, which was determined using the Gene Logic datasuite for GENBANK Accession No. AA813827 as described in Example 2.
  • Candidate 4 is overexpressed in colon tumor tissue.
  • Figures 6 A and 6B show PCR data of Candidate 3 expression (Figure 6 A) and GAPDH expression (Figure 6B) in normal human tissues.
  • Candidate 3 was screened against Human Multiple Tissue cDNA panels I & II (Clontech #K1420-1 & # K1421-1 ) according to the manufacturer's instructions. GAPDH was not tested against the prostate sample.
  • the positive confrol for Candidate 3 was IMAGE 2324560, obtained from the American Tissue Type Collection (Manassas, Virginia).
  • the cDNA samples present in each lane are as follows: lane 1, heart; lane 2, brain; lane 3, placenta; lane 4, lung; lane 5, liver; lane 6, skeletal muscle; lane 7, kidney; lane 8, pancreas; lane 9, spleen; lane 10, thymus; lane 11, prostate; lane 12, testis; lane 13, ovary; lane 14, small intestine; lane 15, colon; lane 16, peripheral blood leukocytes; lane 17, positive control; lane 18, negative control.
  • Figures 7A and 7B show PCR data of Candidate 3 expression (Figure 7A) and GAPDH expression (Figure 7B) in colon tumor samples.
  • the cDNA samples present in each lane are as follows: lane 1, grade 3 adenocarcinoma; lane 2, grade 2 adenocarcinoma; lane 3, grade 1 adenocarcinoma; lane 4, grade 2 adenocarcinoma; lane 5, colorectal cancer cell line HCT116; lane 6, positive control (IMAGE clone); lane 7, negative confrol.
  • the results shown in this figure indicate that candidate 3 is expressed in at least 3 of 4 colon tumor samples in addition to colorectal tumor cell line HCT116.
  • Figure 8 depicts E-Northern expression data for Loc 56926, which is overexpressed in colon cancer, as described in Example 4.
  • Figures 9A and 9B are PCR panels showing expression of Loc56926 (Figure 9A) and GAPDH ( Figure 9B) in malignant colon samples.
  • the cDNA samples present in each lane are as follows: lane M, marker; lane 1, no template control; lane 2 colon cancer 8T; lane 3, colon cancer DT; lane 4, colon cancer FT; lane 5, colon cancer GT; lane 6, colon cancer HT; lane 7, colon cancer IT; lane 8, colon cancer QT; lane 9, prostate cancer OT; lane 10, colon cancer RT; lane 11, colon cancer cell line HCT116; lane 12, positive control EST.
  • the results from this figure demonstrate that Loc56926 expression is present in cDNA from three of eight tested colon cancer samples.
  • Figures 10A and 10B are PCR panels showing expression of Loc56926 (Figure 10 A) and GAPDH (Figure 10B) in normal human tissues. Hybridization was performed using Human Multiple Tissue cDNA panel I (Clontech #K1420-1) according to the manufacturer's instructions.
  • the cDNA samples present in each lane are as follows: lane M, marker; lane 1, no template control; lane 2, colon tumor 8T; lane 3, colon tumor HT; lane 4, colon tumor RT; lane 5, colon cancer cell line HCT116; lane 6, normal colon; lane 7, normal brain; lane 8, normal heart; lane 9, kidney; lane 10, normal liver; lane 11, normal lung; lane 12, skeletal muscle; lane 13, normal pancreas; lane 14, normal placenta lane 15; EST control.
  • Loc56926 is present in colon tumors with light expression in the normal pancreas (note the increase in GAPDH in the pancreas lane compared to the colon tumor lanes) and not expressed at detectable levels the other tested normal human tissues.
  • Figures 11 A and 1 IB are PCR panels showing expression of Loc56926 (Figure 11 A) and GAPDH (Figure 1 IB) in human tissues. Hybridization was performed using Human Multiple Tissue cDNA panel II (Clontech # K1421-1) according to the manufacturer's instructions.
  • the cDNA samples present in each lane are as follows: lane M, marker; lane 1, no template control; lane 2, colon tumor 8T; lane 3, colon tumor HT; lane 4, colon tumor RT; lane 5, colon cancer cell line HCT116; lane 6, normal colon; lane 7, normal peripheral blood leukocytes; lane 8, small intestine; lane 9, normal ovary; lane 10, normal prostate; lane 11, normal spleen; lane 12, normal testis; lane 13, normal thymus; lane 14, EST confrol.
  • Figures 12A and 12B are PCR panels showing expression of Loc56926 (Figure 12A) and GAPDH ( Figure 12B) in normal brain tissue samples. Hybridization was performed using Normal Neural System cDNA panel (Biochain, C8234503, C8234504, C8234505).
  • the cDNA samples present in each lane are as follows: lane M, marker; lane 1, no template confrol; lane 2, cerebellum; lane 3, cerebral cortex; lane 4, medulla oblongata; lane 5, pons; lane 6, frontal lobe; lane 7, occipital lobe; lane 8, parietal lobe; lane 9, temporal lobe; lane 10, placental neural system; lane 11, EST control.
  • Figure 13 depicts E-Northern expression data for the AW779536 gene, which is overexpressed in colon cancer, as described in Example 4.
  • Figure 14 depicts E-Northern expression data for the AL531683 gene, which is overexpressed in colon cancer, as described in Example 4.
  • Figure 15 depicts E-Northern expression data for the AI202201 gene, which is overexpressed in colon cancer, as described in Example 4.
  • Figure 16 depicts E-Northern expression data for the AL389942 gene, which is overexpressed in colon cancer, as described in Example 4.
  • Figure 17 depicts E-Northern expression results for the Ly6G6Dgene, also described in Example 5.
  • Figure 18 depicts E-Northem expression results for FLJ32334, also described in Example 6.
  • the present invention relates to the identification of genes which are to be specifically expressed and upregulated in certain cancers, including colon or colorectal tumors. This was determined using the Gene Logic (Gaithersburg, Maryland) datasuite or Celera (Rockville, Maryland) database and by screening malignant colon tumor tissues as described in detail herein.
  • the present invention involves the discovery that certain genes, the nucleic acid sequences and predicted coding sequences of which are identified herein are specifically expressed in certain malignant tissues including colon or colorectal tumor tissues.
  • the disclosed therapies involve the synthesis of oligonucleotides having sequences in the antisense orientation relative to the genes identified by the present inventors which are specifically expressed by malignant tissues, including colon or colorectal tumors.
  • Suitable therapeutic antisense oligonucleotides typically vary in length from two to several hundred nucleotides in length, more typically about 50-70 nucleotides in length.
  • These antisense oligonucleotides can be administered as naked DNAs or in protected forms, e.g., encapsulated in liposomes.
  • liposomal or other protected forms may enhance in vivo stability and delivery to target sites, i.e., colon tumor cells.
  • the subject novel genes can be used to design novel ribozymes that target the cleavage ofthe corresponding mRNAs in colon and other tumor cells.
  • these ribozymes can be administered in free (naked) form or by the use of delivery systems that enhance stability and/or targeting, e.g., liposomes.
  • Ribozymal and antisense therapies used to target genes that are selectively expressed by cancer cells are well known in the art.
  • the present invention embraces the administration of use of DNAs that hybridize to the novel gene targets identified herein, attached to therapeutic effector moieties, for example radiolabels, including metallic and halogen isotopes (e.g., 90 yttrium, I31 iodine), cytotoxins, cytotoxic enzymes, in order to selectively target and kill cells that express these genes, . e. , colon tumor cells.
  • therapeutic effector moieties for example radiolabels, including metallic and halogen isotopes (e.g., 90 yttrium, I31 iodine), cytotoxins, cytotoxic enzymes, in order to selectively target and kill cells that express these genes, . e. , colon tumor cells.
  • the present invention encompasses non-nucleic acid based therapies, for example antigens encoded by the nucleic acids disclosed herein. It is anticipated that these antigens can be used as therapeutic or prophylactic anti-tumor vaccines.
  • antigens ofthe present invention can be admimstered with adjuvants that induce a cytotoxic T lymphocyte response.
  • Representative adjuvants include those disclosed in U.S. Patent Nos. 5,709,860, 5,695,770, and 5,585,103, which promote CTL responses against prostate and papillomavirus related human colon cancer. The disclosures of U.S. Patent Nos. 5,709,860, 5,695,770, and 5,585,103 are incorporated by reference in their entirety.
  • the disclosed antigens can be administered in combination with an adjuvant to elicit a humoral immune response against such antigens, thereby delaying or preventing the development of cancers (e.g., a colon cancer) associated with the overexpression ofthe antigens.
  • an adjuvant to elicit a humoral immune response against such antigens, thereby delaying or preventing the development of cancers (e.g., a colon cancer) associated with the overexpression ofthe antigens.
  • Embodiments ofthe invention comprise administration of one or more novel colon cancer antigens, for example in combination with an adjuvant.
  • a representative adjuvant is PRO VAX®, which comprises a microfluidized adjuvant containing Squalene, TWEEN® and PLURONIC®, in an amount sufficient to be therapeutically or prophylactically effective. See U.S. Patent Nos. 5,709,860, 5,695,770, and 5,585,103.
  • a typical dosage of formulated antigen ranges from about 50 to about 20,000 mg/kg body weight, or from about 100 to about 5000 mg/kg body weight.
  • the subject tumor-associated antigens can be administered with other adjuvants, e.g., ISCOM®, DETOXTM, SAF®, Freund's adjuvant, Alum, Saponin, among others.
  • the present invention provides methods for preparing monoclonal antibodies against the antigens encoded by the DNA sequences disclosed in the examples which are expressed specifically by certain malignant tissues including colon or colorectal tumor tissues.
  • Monoclonal antibodies are produced by conventional methods and include human monoclonal antibodies, humanized monoclonal antibodies, chimeric monoclonal antibodies, single chain antibodies, including scFv's and antigen-binding antibody fragments such as Fabs, 2 Fabs, and Fab' fragments.
  • monoclonal antibodies and fragments thereof for example by pepsin or papain-mediated cleavage, are well known in the art.
  • an appropriate (non-homologous) host is immunized with the subject colon cancer antigens, immune cells are isolated from the host and used to prepare hybridomas.
  • Monoclonal antibodies that specifically bind to either of such antigens are identified by routine screening techniques.
  • Useful monoclonal antibodies typically bind the target antigens with high affinity, e.g., possess a binding affinity (Kd) on the order of lO "6 to lO "10 M.
  • Monoclonal antibodies and fragments ofthe invention are useful for anti-tumor immunotherapy.
  • therapeutic effector moieties e.g., radiolabels, cytotoxins, therapeutic enzymes, agents that induce apoptosis
  • cytotoxicity i.e., killing of human colon tumor cells.
  • Antibodies and/or antibody fragments are administered to a subject in labeled or unlabeled form, alone or in combination with other therapeutics, such as chemotherapeutics such as progestin, EGFR, TAXOL®, and the like.
  • the administered composition can include a pharmaceutically acceptable carrier, and optionally adjuvants, stabilizers, etc., used in antibody compositions for therapeutic use.
  • the present invention also provides diagnostic methods for detection ofthe colon or colorectal tumor-specific genes disclosed herein. Diagnostic methods include detecting the expression of one or more of these genes at the DNA level or at the protein level. Patients who test positive for the disclosed tumor-specific genes diagnosed are identified as having or being at increased risk of developing colon cancer. Additionally, the levels of antigen expression can be useful in determining patient status, i.e., how far the disease has advanced. For example, the expression or expression level of a tumor-specific gene can indicate a particular stage of tumor progression.
  • gene expression is detected by known DNA detection methods, including but not limited to Northern blot hybridization, strand displacement amplification (SDA), catalytic hybridization amplification (CHA), PCR amplification (for example, using primers corresponding to the novel genes disclosed herein), and other known DNA detection methods.
  • SDA strand displacement amplification
  • CH catalytic hybridization amplification
  • PCR amplification for example, using primers corresponding to the novel genes disclosed herein
  • DNA detection methods for example, the presence or absence of cancer associated with the genes disclosed herein can be determined based on whether PCR products are obtained, and the level of expression. Expression levels can also be monitored to determine the prognosis of a colon cancer patient as the levels of expression ofthe PCR product likely increase as the disease progresses. Suitable controls and quantification is are performed for diagnostic methods as known in the art.
  • the status of a subject to be tested for colon cancer, or other cancer associated by overexpression of a gene disclosed herein can be evaluated by testing biological fluids, such as blood, urine, colon tissue, with an antibody or antibodies or fragment that specifically binds to the novel colon tumor antigens disclosed herein.
  • biological fluids such as blood, urine, colon tissue
  • an antibody or antibodies or fragment that specifically binds to the novel colon tumor antigens disclosed herein are well known and include ELISA, competitive binding assays, and the like.
  • Representative assays use an antibody or antibody fragment that specifically binds the target antigen directly or indirectly bound to a label that provides for detection, for example, a radiolabel, an enzyme, or a fluorophore.
  • the present invention provides novel genes and corresponding antigens that correlate to human colon cancer.
  • the present invention also embraces variants thereof.
  • variants is intended sequences that are at least 75% identical thereto, for example at least 85% identical, or at least 90% identical when these DNA sequences are aligned to the subject DNAs or a fragment thereof having a size of at least 50 nucleotides.
  • Representative variants include allelic variants.
  • the present invention also provides primers for amplification of nucleic acids encoding the subject novel genes or a portion thereof, which are present is a biological sample, for example, an mRNA library obtained from a desired cell source, including human colon cell or tissue samples.
  • primers are about 12 to 50 nucleotides in length and are constmcted such that they provide for amplification ofthe entire or most ofthe target gene.
  • the present invention further provides antigens encoded by the disclosed DNAs or fragments thereof that bind to or elicit antibodies specific to the full-length antigens. Typically, such fragments are at least 10 amino acids in length, more typically at least 25 amino acids in length.
  • the colon or colorectal tumor-specific genes ofthe invention are expressed in a majority of colon tumor samples tested. Some of these genes are also upregulated in other cancers. Thus, the present invention further contemplates identification of other cancers wherein the expression ofthe disclosed genes or variants thereof correlate to a cancer or an increased likelihood of cancer, for example breast, pancreas, lung or colon cancers. Also provided are compositions and methods to detect and treat such cancers.
  • isolated refers to any human protein that is not in its normal cellular millieu. This includes by way of example compositions comprising recombinant protein, pharmaceutical compositions comprising purified protein, diagnostic compositions comprising purified protein, and isolated protein compositions comprising protein.
  • an isolated protein comprises a substantially pure protein, in that it is substantially free of other proteins, for example, at least 90% pure, that comprises the amino acid sequence disclosed herein or natural homologues or mutants having essentially the same sequence. A naturally occurring mutant might be found, for instance, in tumor cells expressing a gene encoding a mutated protein sequence.
  • Native human protein refers to a protein that comprises the amino acid sequence of the protein expressed in its endogenous environment, i.e., a human colon or colorectal tumor tissue.
  • “Native non-human primate protein” refers to a protein that is a non-human primate homologue ofthe protein having the amino acid sequence discussed in the examples. Given the phylogenetic closeness of humans to other primates, it is anticipated that human and non- human proteins expressed by the genes disclosed in the examples have non-human primate counterparts that possess amino acid sequences that are highly similar, such as 95% sequence identity or higher.
  • isolated human or non-human primate nucleic acid molecule or sequence refers to a nucleic acid molecule that encodes human protein which is not in its normal human cellular millieu, e.g., is not comprised in the human or non-human primate chromosomal DNA.
  • a promoter or a DNA encoding a detectable marker or effector moiety Representative nucleic acid sequence encoding human proteins are disclosed herein. Also included are natural homologues or mutants having substantially the same sequence. Naturally occurring homologies that are degenerate would encode the same protein as discussed herein in the examples, but would include nucleotide differences that do not change the corresponding amino acid sequence. Naturally occurring mutants might be found in tumor cells, wherein such nucleotide differences result in a mutant protein. Naturally occurring homologues containing conservative substitutions are also encompassed.
  • Variant of human or non-human primate protein refers to a protein possessing an amino acid sequence that possess at least 90% sequence identity, such as at least 91% sequence identity, or at least 92% sequence identity, or at least 93% sequence identity, or at least 94% sequence identity, or at least 95% sequence identity, or at least 96% sequence identity, or at least 97% sequence identity, or at least 98% sequence identity, and including at least 99% sequence identity, to the corresponding native human or non-human primate protein wherein sequence identity is as defined herein.
  • a variant possesses at least one biological property in common with the human or non-human protein.
  • Variant of human or non-human primate nucleic acid molecule or sequence refers to a nucleic acid sequence that possesses at least 90% sequence identity, such as at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96%, or at least 97%, or at least 98% sequence identity, and including at least 99% sequence identity, to the corresponding native human or non-human primate nucleic acid sequence, wherein "sequence identity" is as defined herein.
  • “Fragment of human or non-human primate nucleic acid molecule or sequence” refers to a nucleic acid sequence corresponding to a portion ofthe native human nucleic acid sequence discussed herein in the examples or a primate native non-human homolog molecule, wherein said portion is at least about 50 nucleotides in length, or 100, for example, at least 200 or 300 nucleotides in length.
  • Antigenic fragments of colon or colorectal refer to polypeptides corresponding to a fragmentt o ⁇ f colon antigen encoded by any ofthe genes disclosed herein or a variant or homologue thereof that when used itself or attached to an immunogenic carrier that elicits antibodies that specifically bind the protein.
  • antigenic fragments are at least 20 amino acids in length.
  • Sequence identity or percent identity is intended to mean the percentage ofthe same residues shared between two sequences, referenced to the human DNA or amino acid sequences disclosed herein, when the two sequences are aligned using the Clustal method [Higgins et al, Cabios 8:189-191 (1992)] of multiple sequence alignment in the Lasergene biocomputing software (DNASTAR, INC. of Madison, Wisconsin). In this method, multiple alignments are carried out in a progressive manner, in which larger and larger alignment groups are assembled using similarity scores calculated from a series of pairwise alignments.
  • Optimal sequence alignments are obtained by finding the maximum alignment score, which is the average of all scores between the separate residues in the alignment, determined from a residue weight table representing the probability of a given amino acid change occurring in two related proteins over a given evolutionary interval. Penalties for opening and lengthening gaps in the alignment contribute to the score.
  • the residue weight table used for the alignment program is PAM25O [Dayhoffet al., in Atlas of Protein Sequence and Stracture, Dayhoff, Ed., NDRF, Washington, Vol. 5, suppl. 3, p. 345, (1978)].
  • Percent conservation is calculated from the above alignment by adding the percentage of identical residues to the percentage of positions at which the two residues represent a conservative substitution (defined as having a log odds value of greater than or equal to 0.3 in the PAM250 residue weight table).
  • Conservation is referenced to a human gene ofthe invention when determining percent conservation with a non-human gene and when determining percent conservation.
  • Conservative amino acid changes satisfying this requirement include: R-K; E-D, Y-F, L-M; V-I, Q-H.
  • polypeptide fragments ofthe disclosed proteins can comprise at least 8 amino acid residues, such as at least 25 or at least 50 amino acid residues of human or non-human primate gene according to the invention or an analogue thereof.
  • Polypeptide fragments can also comprise at least 75, 100, 125, 150, 175, 200, 225, 250, or 275 residues ofthe polypeptide encoded by gene the subject genes which are specifically expressed by certain human colon or colorectal as well as some other tumor tissues.
  • a protein fragment can also comprise a majority ofthe native protein colon or colorectal protein, i.e. at least about 100 contiguous residues ofthe native colon or colorectal protein antigen.
  • the invention also encompasses biologically active mutants of protein colon or colorectal proteins according to the invention, which comprise an amino acid sequence that is at least 80%, for example, 90% or 95-99% similar to the subject tumor-associated proteins.
  • Guidance in determining which amino acid residues can be substituted, inserted, or deleted without abolishing biological or immunological activity can be found using computer programs well known in the art, such as DNASTAR software.
  • Protein variants can include conoservative amino acid changes, i.e., substitutions of similarly charged or uncharged amino acids. A conservative amino acid change involves substitution of one of a family of amino acids which are related in their side chains.
  • Naturally occurring amino acids are generally divided into four families: acidic (aspartate, glutamate), basic (lysine, arginine, histidine), non-polar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), and uncharged polar (glycine, asparagine, glutamine, cystine, serine, threonine, tyrosine) amino acids. Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids.
  • mutants are a group of polypeptides in which neutral amino acids, such as serines, are substituted for cysteine residues which do not participate in disulfide bonds. These mutants may be stable over a broader temperature range than native secreted proteins. See Mark et al, U.S. Patent 4,959,314.
  • Human or non-human primate protein variants include glycosylated forms, aggregative conjugates with other molecules, and covalent conjugates with unrelated chemical moieties. Also, protein variants also include allelic variants, species variants, and muteins. Truncations or deletions of regions which do not affect the differential expression ofthe protein gene are also variants. Covalent variants can be prepared by linking functionalities to groups which are found in the amino acid chain or at the N- or C-terminal residue, as is known in the art.
  • polypeptides ofthe present invention can include one or more amino acid substitutions, deletions or additions, either from natural mutations or human manipulation.
  • the invention further includes variations ofthe protein subject colon or colorectal which show comparable expression patterns or which include antigenic regions.
  • Protein mutants include deletions, insertions, inversions, repeats, and type substitutions.
  • Guidance concerning which amino acid changes are likely to be phenotypically silent can be found in Bowie, J.U., et al., "Deciphering the Message in Protein Sequences: Tolerance to Amino Acid Substitutions," Science 247:1306-1310 (1990).
  • charged amino acids can be substituted with another charged amino acid, or with neutral or negatively charged amino acids.
  • the latter results in proteins with reduced positive charge to improve the characteristics ofthe disclosed protein.
  • the prevention of aggregation is highly desirable. Aggregation of proteins not only results in a loss of activity but can also be problematic when preparing pharmaceutical formulations, because they can be immunogenic. (Pinckard et al., Clin. Exp. Immunol. 2:331-340 (1967); Robbins et al., Diabetes 36:838-845 (1987); Cleland et al., Crit. Rev. Therapeutic Drug Carrier Systems 10:307-377 (1993)).
  • Amino acids in the polypeptides ofthe present invention that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine- scanning mutagenesis (Cunningham and Wells, Science 244: 1081-1085 (1989)). The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity such as binding to a natural or synthetic binding partner. Sites that are critical for ligand-receptor binding can also be determined by structural analysis such as crystallization, nuclear magnetic resonance or photoaffinity labeling (Smith et al., JMol. Biol. 224:899-904 (1992) and de Vos et al. Science 255: 306-312 (1992)).
  • amino acid substitutions often do not significantly affect the folding or activity ofthe protein.
  • a skilled artisan could determine an appropriate number and nature of amino acid substitutions based on factors as described above. Generally speaking, the number of substitutions for any given polypeptide are fewer than 50, 40, 30, 25, 20, 15, 10, 5 or 3 residues.
  • Fusion proteins comprising proteins or polypeptide fragments ofthe subject colon or colorectal proteins can also be constructed. Fusion proteins are useful for generating antibodies against amino acid sequences and for use in various assay systems. For example, fusion proteins can be used to identify proteins which interact with a protein ofthe invention or which interfere with its biological function. Physical methods, such as protein affinity chromatography, or library-based assays for protein-protein interactions, such as the yeast two-hybrid or phage display systems, can also be used for this purpose. The foregoing can also be adapted as a screening technique.
  • Fusion proteins comprising a signal sequence and or a transmembrane domain of a protein according to the invention or a fragment thereof can be used to target other protein domains to cellular locations in which the domains are not normally found, such as bound to a cellular membrane or secreted extracellularly.
  • a fusion protein comprises two protein segments fused together by means of a peptide bond.
  • Amino acid sequences for use in fusion proteins ofthe invention can utilize any ofthe amino acid sequences or encoded by the nucleotide sequences disclosed herein, or can be prepared from biologically active variants or fragment of said protein sequence, such as those described above.
  • the first protein segment can consist of a full-length protein or a variant or fragment thereof. These fragments can range in size from about 8 amino acids up to the full length ofthe protein.
  • the second protein segment can be a full-length protein or a polypeptide fragment.
  • Proteins commonly used in fusion protein construction include ⁇ -galactosidase, ⁇ - glucuronidase, green fluorescent protein (GFP), autofluorescent proteins, including blue fluorescent protein (BFP), glutathione-S-transferase (GST), luciferase, horseradish peroxidase (HRP), and chloramphenicol acetyltransferase (CAT).
  • epitope tags can be used in fusion protein constructions, including histidine (His) tags, FLAG tags, influenza hemagglutinin (HA) tags, Myc tags, VSV-G tags, and thioredoxin (Trx) tags.
  • Other fusion constructions can include maltose binding protein (MBP), S-tag, Lex a DNA binding domain (DBD) fusions, GAL4 DNA binding domain fusions, and herpes simplex virus (HSV) BP16 protein fusions.
  • fusions can be made, for example, by covalently linking two protein segments or by standard procedures in the art of molecular biology.
  • Recombinant DNA methods can be used to prepare fusion proteins, for example, by making a DNA construct which comprises a coding sequence encoding an amino acid sequence according to the invention in proper reading frame with a nucleotide encoding the second protein segment and expressing the DNA construct in a host cell, as is known in the art.
  • Many kits for constructing fusion proteins are available from companies that supply research labs with tools for experiments, including, for example, Promega Corporation (Madison, Wl), Stratagene (La Jolla, CA), Clontech (Mountain View, CA), Santa Cruz Biotechnology (Santa Cruz, CA), MBL
  • Proteins, fusion proteins, or polypeptides ofthe invention can be produced by recombinant DNA methods.
  • a sequence listing encoding one ofthe subject colon or colorectal proteins can be expressed in prokaryotic or eukaryotic host cells using expression systems known in the art. These expression systems include bacterial, yeast, insect, and mammalian cells.
  • the resulting expressed protein can then be purified from the culture medium or from extracts ofthe cultured cells using purification procedures known in the art. For example, for proteins fully secreted into the culture medium, cell-free medium can be diluted with sodium acetate and contacted with a cation exchange resin, followed by hydrophobic interaction chromatography. Using this method, the desired protein or polypeptide is typically greater than 95% pure. Further purification can be undertaken, using, for example, any ofthe techniques listed above.
  • Proteins can be further modified, for example by phosphorylation or glycosylation of the appropriate sites, in order to obtain a functional protein.
  • Covalent attachments can be made using known chemical or enzymatic methods.
  • Human or non-human primate proteins according to the invention or polypeptide of the invention can also be expressed in cultured host cells in a form that facilitates purification.
  • a protein or polypeptide can be expressed as a fusion protein comprising, for example, maltose binding protein, glutathione-S-transferase, or thioredoxin, and purified using a commercially available kit. Kits for expression and purification of such fusion proteins are available from companies such as New England BioLabs, Pharmacia, and Invifrogen. Proteins, fusion proteins, or polypeptides can also be tagged with an epitope, such as a "Flag" epitope (Kodak), and purified using an antibody which specifically binds to that epitope.
  • an epitope such as a "Flag" epitope (Kodak)
  • transgenic animals such as mice, rats, guinea pigs, cows, goats, pigs, or sheep.
  • Female transgenic animals can then produce proteins, polypeptides, or fusion proteins ofthe invention in their milk. Methods for constructing such animals are known and widely used in the art.
  • synthetic chemical methods such as solid phase peptide synthesis, can be used to synthesize a secreted protein or polypeptide.
  • General means for the production of peptides, analogs or derivatives are outlined in Chemistry and Biochemistry of Amino Acids, Peptides, and Proteins - A Survey of Recent Developments, B. Weinstein, ed. (1983).
  • Substitution of D-amino acids for the normal L-stereoisomer can be carried out to increase the half-life ofthe molecule.
  • homologous polynucleotide sequences can be confirmed by hybridization under stringent conditions, as is known in the art.
  • homologous sequences can be identified which contain at most about 25-30% base pair mismatches.
  • homologous nucleic acids can contain 15-25% base pair mismatches or fewer, for example about 5-15% base pair mismatches.
  • the invention also provides polynucleotide probes which can be used to detect complementary nucleotide sequences, for example, in hybridization protocols such as Northern or Southern blotting or in situ hybridizations.
  • Polynucleotide probes ofthe invention comprise at least 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, or 40 or more contiguous nucleotides ofthe gene A and gene B nucleic acid sequences provided herein.
  • Polynucleotide probes ofthe invention can comprise a detectable label, such as a radioisotopic, fluorescent, enzymatic, or chemiluminescent label.
  • Isolated genes corresponding to the cDNA sequences disclosed herein are also provided. Standard molecular biology methods can be used to isolate the corresponding genes using the cDNA sequences provided herein. These methods include preparation of probes or primers based on the disclosed sequences for use in identifying or amplifying the genes from mammalian, including human, genomic libraries or other sources of human genomic DNA.
  • Polynucleotide molecules ofthe invention can also be used as primers to obtain additional copies of the polynucleotides, using polynucleotide amplification methods.
  • Polynucleotide molecules can be propagated in vectors and cell lines using techniques well known in the art. Polynucleotide molecules can be on linear or circular molecules. They can be on autonomously replicating molecules or on molecules without replication sequences. They can be regulated by their own or by other regulatory sequences, as is known in the art.
  • Polynucleotide molecules comprising the coding sequences disclosed herein can be used in a polynucleotide construct, such as a DNA or RNA construct.
  • Polynucleotide molecules ofthe invention can be used, for example, in an expression construct to express all or a portion of a protein, variant, fusion protein, or single-chain antibody in a host cell.
  • An expression construct comprises a promoter which is functional in a chosen host cell. The skilled artisan can readily select an appropriate promoter from the large number of cell type- specific promoters known and used in the art.
  • the expression construct can also contain a transcription terminator which is functional in the host cell.
  • the expression construct comprises a polynucleotide segment which encodes all or a portion ofthe desired protein. The polynucleotide segment is located downstream from the promoter. Transcription ofthe polynucleotide segment initiates at the promoter.
  • the expression construct can be linear or circular and can contain sequences, if desired, for
  • polynucleotide molecules comprising human or non-human primate gene promoter and UTR sequences, operably linked to either protein coding sequences or other sequences encoding a detectable or selectable marker.
  • Promoter and/or UTR-based constructs are useful for studying the transcriptional and translational regulation of protein expression, and for identifying activating and/or inhibitory regulatory proteins.
  • An expression construct can be introduced into a host cell.
  • the host cell comprising the expression construct can be any suitable prokaryotic or eukaryotic cell.
  • Expression systems in bacteria include those described in Chang et al, Nature 275:615 (1978); Goeddel et al, Nature 281: 544 (1979); Goeddel et al, Nucleic Acids Res. 8:4057 (1980); EP 36,776;
  • Mammalian expression can be accomplished as described in Dijkema et al, EMBO J. 4: 761(1985); Gormanetal, Proc. Natl. Acad. Sci. USA 79: 6777 (1982b); Boshart et al, Cell 41: 521 (1985); and U.S. 4,399,216.
  • Other features of mammalian expression can be facilitated as described in Ham and Wallace, Meth Enz. 58: 44 (1979); Barnes and Sato, Anal. Biochem. 102: 255 (1980); U.S. 4,767,704; U.S. 4,657,866; U.S. 4,927,762; U.S. 4,560,655; WO 90/103430, WO 87/00195, and U.S. RE 30,985.
  • Expression constructs can be introduced into host cells using any technique known in the art. These techniques include transferrin-polycation-mediated DNA transfer, transfection with naked or encapsulated nucleic acids, liposome-mediated cellular fusion, intracellular transportation of DNA-coated latex beads, protoplast fusion, viral infection, electroporation, "gene gun,” and calcium phosphate-mediated transfection.
  • Expression of an endogenous gene encoding a protein ofthe invention can also be manipulated by introducing by homologous recombination a DNA construct comprising a transcription unit in frame with the endogenous gene, to form a homologously recombinant cell comprising the transcription unit.
  • the transcription unit comprises a targeting sequence, a regulatory sequence, an exon, and an unpaired splice donor site.
  • the new transcription unit can be used to turn the endogenous gene on or off as desired. This method of affecting endogenous gene expression is taught in U.S. Patent 5,641,670.
  • the targeting sequence is a segment of at least 10, 12, 15, 20, or 50 contiguous nucleotides ofthe nucleotide sequences disclosed herein.
  • the transcription unit is located upstream to a coding sequence ofthe endogenous gene.
  • the exogenous regulatory sequence directs transcription ofthe coding sequence ofthe endogenous gene.
  • Human or non-human primate protein can also include hybrid and modified forms thereof including fusion proteins, fragments and hybrid and modified forms in which certain amino acids have been deleted or replaced, modifications such as where one or more amino acids have been changed to a modified amino acid or unusual amino acid.
  • any human or non-human primate protein which shows cross-reactivity with antibodies to a gene described herein or whose encoding nucleotide sequences including genomic DNA, mRNA or cDNA are isolated through hybridization with the complementary sequence of genomic or subgenomic nucleotide sequences or cDNA of a gene disclosed herein or a fragment thereof.
  • Degenerate DNA sequences that encode human or non-human primate proteins are also included within the present invention as are allelic variants of.
  • Colon or colorectal proteins ofthe invention can be prepared using recombinant DNA techniques.
  • pure form or “purified form” or “substantially purified form” it is meant that a protein composition is substantially free of other proteins which are not protein.
  • the present invention also includes therapeutic or pharmaceutical compositions comprising human or non-human primate proteins, fragments or variants according to the invention in an effective amount for treating patients with disease, and a method comprising administering a therapeutically effective amount of a protein according to the invention.
  • These compositions and methods are useful for treating cancers associated with a protein according to the invention, e.g. colon cancer.
  • One skilled in the art can readily use a variety of assays known in the art to determine whether a protein according to the invention would be useful in promoting survival or functioning in a particular cell type.
  • anti-sense oligonucleotides can be made specific to genes disclosed herein and a method utilized for diminishing the level of expression a protein according to the invention by a cell comprising administering one or more gene anti-sense oligonucleotides.
  • gene specific anti-sense oligonucleotides reference is made to oligonucleotides that have a nucleotide sequence that interacts through base pairing with a specific complementary nucleic acid sequence involved in the expression of a gene according to the invention that the expression ofthe gene is reduced.
  • Nucleic acids involved in the expression ofthe subject gene include genomic DNA and mRNA that encode a colon or colorectal gene disclosed herein.
  • This genomic DNA molecule can comprise regulatory regions ofthe gene, or the coding sequence for mature gene encoded by the gene.
  • the term complementary to a nucleotide sequence in the context of antisense oligonucleotides and methods therefor means sufficiently complementary to such a sequence as to allow hybridization to that sequence in a cell, i.e., under physiological conditions.
  • the antisense oligonucleotides can comprise a sequence containing from about 8 to about 100 nucleotides, including antisense oligonucleotides that comprise from about 15 to about 30 nucleotides.
  • the antisense oligonucleotides can also contain a variety of modifications that confer resistance to nucleolytic degradation such as, for example, modified intemucleoside linages [Uhlmann and Peyman, Chemical Reviews 90:543-548 (1990); Schneider and Banner, Tetrahedron Lett. 31:335, (1990) which are incorporated by reference], modified nucleic acid bases as disclosed in 5,958,773 and patents disclosed therein, and/or sugars and the like.
  • modified intemucleoside linages Uhlmann and Peyman, Chemical Reviews 90:543-548 (1990); Schneider and Banner, Tetrahedron Lett. 31:335, (1990) which are incorporated by reference
  • modified nucleic acid bases as disclosed in 5,958,773 and patents disclosed therein, and/or sugars and the like.
  • the antisense compounds ofthe invention can include modified bases.
  • the antisense oligonucleotides ofthe invention can also be modified by chemically linking the oligonucleotide to one or more moieties or conjugates to enhance the activity, cellular distribution, or cellular uptake ofthe antisense oligonucleotide.
  • Representative moieties or conjugates include lipids such as cholesterol, cholic acid, thioether, aliphatic chains, phospholipids, polyamines, polyethylene glycol (PEG), palmityl moieties, and others as disclosed in, for example, U.S. Patents 5,514,758, 5,565,552, 5,567,810, 5,574,142, 5,585,481, 5,587,371, 5,597,696 and 5,958,773.
  • Chimeric antisense oligonucleotides are also within the scope ofthe invention, and can be prepared from the present inventive oligonucleotides using the methods described in, for example, U.S. Patents 5,013,830, 5,149,797, 5,403,711, 5,491,133, 5,565,350, 5,652,355, 5,700,922 and 5,958,773.
  • Select of optimal antisense molecules for particular targets typically involves routine screening of a number of candidate molecules.
  • An antisense molecule can be targeted to an accessible, or exposed, portion ofthe target RNA molecule. Although in some cases information is available about the stracture of target mRNA molecules, the current approach to inhibition using antisense is via experimentation.
  • mRNA levels in the cell can be measured routinely in treated and control cells by reverse transcription ofthe mRNA and assaying the cDNA levels.
  • the biological effect can be determined routinely by measuring cell growth or viability as is known in the art. Measuring the specificity of antisense activity by assaying and analyzing cDNA levels is an art-recognized method of validating antisense results. It has been suggested that RNA from treated and control cells should be reverse-transcribed and the resulting cDNA populations analyzed. [Branch, A. D., T.I.B.S. 23:45-50 (1998)].
  • compositions ofthe present invention can be administered by any suitable route known in the art including for example intravenous, subcutaneous, intramuscular, transdermal, intrathecal or infracerebral. Administration can be either rapid as by injection or over a period of time as by slow infusion or administration of slow release formulation.
  • a human or non-human primate protein according to the invention can also be linked or conjugated with agents that provide desirable pharmaceutical or pharmacodynamic properties.
  • the protein can be coupled to any substance known in the art to promote penetration or transport across the blood-brain barrier such as an antibody to the transferrin receptor, and administered by intravenous injection (see, for example, Friden et al., Science 259:373-377 (1993) which is incorporated by reference).
  • the subject protein can be stably linked to a polymer such as polyethylene glycol to obtain desirable properties of solubility, stability, half-life and other pharmaceutically advantageous properties.
  • a polymer such as polyethylene glycol
  • compositions are usually employed in the form of pharmaceutical preparations, which are made in a manner well known in the pharmaceutical art. See, e.g. Remington Pharmaceutical Science, 18th Ed., Merck Publishing Co. Eastern PA, (1990).
  • Physiological saline solutions can be used, as well as other pharmaceutically acceptable carriers such as physiological concentrations of other non-toxic salts, five percent aqueous glucose solution, sterile water and the like.
  • Compositions ofthe invention can also include a suitable buffer.
  • such solutions can be lyophilized and stored in a sterile ampoule ready for reconstitution by the addition of sterile water for ready injection.
  • the primary solvent can be aqueous or alternatively non-aqueous.
  • the subject human or primate protein, fragment or variant thereof can also be incorporated into a solid or semi-solid biologically compatible matrix which can be implanted into tissues requiring treatment.
  • the carrier can also contain other pharmaceutically-acceptable excipients for modifying or maintaining the pH, osmolarity, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor ofthe formulation.
  • the carrier can contain still other pharmaceutically-acceptable excipients for modifying or maintaining release or absorption or penetration across the blood-brain barrier.
  • Excipients are those substances usually and customarily employed to formulate dosages for parenteral administration in either unit dosage or multi-dose form or for direct infusion into the cerebrospinal fluid by continuous or periodic infusion.
  • Dose administration can be repeated depending upon the pharmacokinetic parameters ofthe dosage formulation and the route of administration used. It is also contemplated that certain formulations containing a protein according to the invention or variant or fragment thereof are to be administered orally. Protein formulations can be encapsulated and formulated with suitable carriers in solid dosage forms.
  • suitable carriers, excipients, and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, calcium silicate, microcrystalline cellulose, polyvinylpyrrohdone, cellulose, gelatin, syrap, methyl cellulose, methyl- and propylhydroxybenzoates, talc, magnesium, stearate, water, mineral oil, and the like.
  • the formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents or flavoring agents.
  • compositions can be formulated so as to provide rapid, sustained, or delayed release of the active ingredients after administration to the patient by employing procedures well known in the art.
  • the formulations can also contain substances that diminish proteolytic degradation and promote absorption such as, for example, surface active agents.
  • the specific dose is calculated according to the approximate body weight or body surface area ofthe patient or the volume of body space to be occupied. The dose also depends on the particular route of admimsfration selected. Further refinement of the calculations necessary to determine the appropriate dosage for treatment is routinely made by those of ordinary skill in the art. Following a review ofthe present disclosure, an effective dosage can be determined without undue experimentation. Exact dosages are determined in conjunction with standard dose-response studies. The amount ofthe composition actually administered can be determined by a practitioner, in the light ofthe relevant circumstances including the condition or conditions to be treated, the choice of composition to be administered, the age, weight, and response ofthe individual patient, the severity ofthe patient's symptoms, and the chosen route of administration.
  • a protein ofthe present invention is therapeutically administered by implanting into patients vectors or cells capable of producing a biologically-active form of the protein or a precursor ofthe protein, i.e., a molecule that can be readily converted to a biological-active form ofthe by the body.
  • cells that secrete the protein can be encapsulated into semipermeable membranes for implantation into a patient.
  • the cells can be cells that normally express the protein or a precursor thereof or the cells can be transformed to express the protein or a precursor thereof.
  • a human protein can be used, or a non-human primate protein homolog of a human protein can be used.
  • detection as used herein in the context of detecting the presence of a cancer gene according to the invention in a patient is intended to include the determining of the amount of protein according to the invention or the ability to express an amount of this protein in a patient, the estimation of prognosis in terms of probable outcome of a disease and prospect for recovery, the monitoring of these protein levels over a period of time as a measure of status ofthe condition, and the monitoring of colon or colorectal protein according to the invention for determining an effective therapeutic regimen for the patient, e.g. one with colon cancer.
  • a sample is obtained from the patient.
  • the sample can be a tissue biopsy sample or a sample of blood, plasma, serum, CSF or the like. It has been found that the subject genes are expressed at high levels in some cancers, e.g., colon or colorectal cancers. Samples for detecting protein can be taken from these tissue. When assessing peripheral levels of protein, a sample of blood, plasma or serum can be used. When assessing the levels of protein in the central nervous system, samples can be obtained from cerebrospinal fluid or neural tissue. In some instances, it is desirable to determine whether a gene according to the invention is intact in the patient or in a tissue or cell line within the patient.
  • a method for detecting and characterizing any alterations in the gene.
  • the method comprises providing an oligonucleotide that contains the gene corresponding cDNA, genomic DNA or a fragment thereof or a derivative thereof.
  • a derivative of an oligonucleotide it is meant that the derived oligonucleotide is substantially the same as the sequence from which it is derived in that the derived sequence has sufficient sequence complementarily to the sequence from which it is derived to hybridize specifically to the gene.
  • a nucleic acid ofthe invention can be isolated, chemically synthesized, of recombinantly produced (e.g., using in vitro DNA replication, reverse transcription, or transcription).
  • patient genomic DNA is isolated from a cell sample from the patient and digested with one or more restriction endonucleases such as, for example, Taql and Alul.
  • this assay determines whether a patient or a particular tissue in a patient has an intact gene according to the invention or a gene abnormality.
  • Hybridization to a gene according to the invention would involve denaturing the chromosomal DNA to obtain a single-stranded DNA; contacting the single-stranded DNA with a gene probe associated with the gene sequence; and identifying the hybridized DNA- probe to detect chromosomal DNA containing at least a portion of a human gene according to the invention.
  • probe refers to a stracture comprised of a polynucleotide that forms a hybrid stracture with a target sequence, due to complementarity of probe sequence with a sequence in the target region.
  • Oligomers suitable for use as probes typically contain at least about 8-12 contiguous nucleotides which are complementary to the targeted sequence, for example 20 nucleotides.
  • Probes ofthe present invention can be DNA or RNA oligonucleotides and can be made by any method known in the art such as, for example, excision, transcription or chemical synthesis. Probes can be labeled with any detectable label known in the art such as, for example, radioactive or fluorescent labels or enzymatic marker.
  • Labeling ofthe probe can be accomplished by any method known in the art such as by PCR, random priming, end labeling, nick translation or the like. Methods that do not employ a labeled probe can also be used to determine the hybridization. Representative techniques include Southern blotting, fluorescence in situ hybridization, and single-strand conformation polymo ⁇ hism with PCR amplification.
  • Hybridization is typically carried out at about 25° - 45° C, or at about 32° -40° C, or at about 37° - 38° C. Hybridization can proceed for about 0.25 hour to about 96 hours, or from about 1 (one) hour to about 72 hours, or from about 4 hours to about 24 hours. Gene abnormalities can also be detected by using the PCR method and primers that flank or lie within the particular gene.
  • the PCR method is well known in the art. Briefly, this method is performed using two oligonucleotide primers which are capable of hybridizing to the nucleic acid sequences flanking a target sequence that lies within gene and amplifying the target sequence.
  • oligonucleotide primer refers to a short strand of DNA or RNA ranging in length from about 8 to about 30 bases.
  • the upstream and downstream primers are typically from about 20 to about 30 base pairs in length and hybridize to the flanking regions for replication ofthe nucleotide sequence.
  • the polymerization is catalyzed by a DNA-polymerase in the presence of deoxynucleotide triphosphates or nucleotide analogs to produce double-stranded DNA molecules.
  • the double strands are then separated by any denaturing method including physical, chemical or enzymatic.
  • a method of physical denaturation involving heating the nucleic acid, typically to temperatures from about 80°C to 105°C for times ranging from about 1 to about 10 minutes. The process is repeated for the desired number of cycles.
  • the primers are selected to be substantially complementary to the strand of DNA being amplified. Therefore, the primers need not reflect the exact sequence ofthe template, but must be sufficiently complementary to selectively hybridize with the sfrand being amplified.
  • the DNA sequence comprising a gene ofthe invention or a fragment thereof is then directly sequenced and analyzed by comparison ofthe sequence with the sequences disclosed herein to identify alterations which might change activity or expression levels or the like.
  • a method for detecting protein a colon according to the invention is provided based upon an analysis of tissue expressing the gene. Certain tissues such as breast, lung, colon and others can be analyzed. The method comprises hybridizing a polynucleotide to mRNA from a sample of tissue that normally expresses the gene. The sample is obtained from a patient suspected of having an abnormality in the gene. To detect the presence of mRNA encoding protein a colon or colorectal protein according to the invention is obtained from a patient. The sample can be from blood or from a tissue biopsy sample. The sample can be treated to extract the nucleic acids contained therein. The resulting nucleic acid from the sample is subjected to gel electrophoresis or other size separation techniques. The mRNA ofthe sample is contacted with a DNA sequence serving as a probe to form hybrid duplexes. The use of a labeled probes as discussed above allows detection ofthe resulting duplex.
  • high stringency conditions can be used in order to prevent false positives, that is the hybridization and apparent detection ofthe gene nucleotide sequences when in fact an intact and functioning gene is not present.
  • sequences derived from the gene or cDNA less stringent conditions could be used, however, are less preferred because ofthe likelihood of false positives.
  • the stringency of hybridization is determined by a number of factors during hybridization and during the washing procedure, including temperature, ionic strength, length of time and concentration of formamide. These factors are outlined in, for example, Sambrook et al. [Sambrook et al. (1989), supra].
  • RT/PCR reverse transcription polymerization chain reaction
  • the method of RT/PCR is well known in the art, and can be performed as follows.
  • Total cellular RNA is isolated by, for example, the standard guanidium isothiocyanate method and the total RNA is reverse transcribed.
  • the reverse transcription method involves synthesis of DNA on a template of RNA using a reverse transcriptase enzyme and a 3' end primer. Typically, the primer contains an oligo(dT) sequence.
  • the cDNA thus produced is then amplified using the PCR method and specific primers.
  • the polymerase chain reaction method is performed as described above using two oligonucleotide primers that are substantially complementary to the two flanking regions of the DNA segment to be amplified. Following amplification, the PCR product is then electrophoresed and detected by ethidium bromide staining or by phosphoimaging.
  • the present invention further provides for methods to detect the presence of a colon or colorectal protein in a sample obtained from a patient.
  • any method known in the art for detecting proteins can be used. Representative methods include, but are not limited to immunodiffusion, immunoelectrophoresis, immunochemical methods, binder-ligand assays, immunohistochemical techniques, agglutination and complement assays. [Basic and Clinical Immunology, 217-262, Sites and Terr, eds., Appleton & Lange, Norwalk, CT, (1991), which is inco ⁇ orated by reference].
  • binder-ligand immunoassays can be used, which involve reacting antibodies with an epitope or epitopes of a colon protein ofthe invention and competitively displacing a labeled protein or derivative thereof.
  • a derivative of a protein according to the invention is intended to include a polypeptide in which certain amino acids have been deleted or replaced or changed to modified or unusual amino acids wherein the derivative is biologically equivalent to the gene and wherein the polypeptide derivative cross-reacts with antibodies raised against the protein.
  • cross-reaction it is meant that an antibody reacts with an antigen other than the one that induced its formation.
  • Numerous competitive and non-competitive protein-binding immunoassays are well known in the art. Antibodies employed in such assays can be unlabeled, for example as used in agglutination tests, or labeled for use in a wide variety of assay methods.
  • Labels that can be used include radionuclides, enzymes, fluorescers, chemiluminescers, enzyme substrates or co-factors, enzyme inhibitors, particles, dyes and the like for use in radioinununoassay (RIA), enzyme immunoassays, e.g., enzyme-linked immunosorbent assay (ELISA), fluorescent immunoassays and the like.
  • RIA radioinununoassay
  • ELISA enzyme-linked immunosorbent assay
  • fluorescent immunoassays and the like.
  • polyclonal or monoclonal antibodies to the subject non-human primate or human proteins or according to the invention an epitope thereof can be made for use in immunoassays by any of a number of methods known in the art.
  • epitope reference is made to an antigenic determinant of a polypeptide.
  • An epitope could comprise 3 amino acids in a spatial conformation which is unique to the epitope. Generally an epitope consists of at least 5 such amino acids. Methods of determining the spatial conformation of amino acids are known in the art, and include, for example, x-ray crystallography and 2 dimensional nuclear magnetic resonance.
  • One approach for preparing antibodies to a protein is the selection and preparation of an amino acid sequence of all or part ofthe protein, chemically synthesizing the sequence and injecting it into an appropriate animal, typically a rabbit, hamster or a mouse.
  • Oligopeptides can be selected as candidates for the production of an antibody to the subject colon or colorectal protein based upon the oligopeptides lying in hydrophilic regions, which are thus likely to be exposed in the mature protein.
  • Additional oligopeptides can be determined using, for example, the Antigenicity Index, Welling, G.W. et al., FEBSLett. 188:215-218 (1985), inco ⁇ orated herein by reference.
  • humanized monoclonal antibodies are provided, wherein the antibodies are specific for a protein according to the invention.
  • the phrase "humanized antibody” refers to an antibody derived from a non-human antibody, typically a mouse monoclonal antibody.
  • a humanized antibody can be derived from a chimeric antibody that retains or substantially retains the antigen-binding properties of the parental, non-human, antibody but which exhibits diminished immunogenicity as compared to the parental antibody when administered to humans.
  • chimeric antibody refers to an antibody containing sequence derived from two different antibodies (see, e.g., U.S. Patent No. 4,816,567) which typically originate from different species. Most typically, chimeric antibodies comprise human and murine antibody fragments generally human constant and mouse variable regions.
  • humanized antibodies are far less immunogenic in humans than the parental mouse monoclonal antibodies, they can be used for the treatment of humans with far less risk of anaphylaxis. Thus, these antibodies are useful in therapeutic applications that involve in vivo administration to a human such as, e.g., use as radiation sensitizers for the treatment of neoplastic disease or use in methods to reduce the side effects of, e.g., cancer therapy.
  • Humanized antibodies can be prepared using a variety of techniques including, for example: (1) grafting the non-human complementarity determining regions (CDRs) onto a human framework and constant region (a process referred to in the art as “humanizing”), or, alternatively, (2) transplanting the entire non-human variable domains, but “cloaking” them with a human-like surface by replacement of surface residues (a process referred to in the art as “veneering”).
  • humanized antibodies include both “humanized” and “veneered” antibodies. These methods are disclosed in, e.g., Jones et al., Nature 321:522-525 (1986); Morrison et al., Proc. Natl. Acad.
  • complementarity determining region refers to amino acid sequences which together define the binding affinity and specificity ofthe natural Fv region of a native immunoglobulin-binding site. See, e.g., Chothia et al., J. Mol. Biol 196:901-917 (1987);
  • constant region refers to the portion ofthe antibody molecule that confers effector functions.
  • mouse constant regions are substituted by human constant regions.
  • the constant regions ofthe subject-humanized antibodies are derived from human immunoglobulins.
  • the heavy chain constant region can be selected from any ofthe five isotypes: alpha, delta, epsilon, gamma or mu.
  • One method of humanizing antibodies comprises aligning the non-human heavy and light chain sequences to human heavy and light chain sequences, selecting and replacing the non-human framework with a human framework based on such alignment, molecular modeling to predict the conformation ofthe humanized sequence and comparing to the conformation ofthe parent antibody. This process is followed by repeated back mutation of residues in the CDR region which disturb the stracture ofthe CDRs until the predicted conformation ofthe humanized sequence model closely approximates the conformation of the non-human CDRs ofthe parent non-human antibody.
  • Humanized antibodies can be further derivatized to facilitate uptake and clearance, e.g, via Ashwell receptors. See, e.g.,
  • Humanized antibodies to proteins according to the invention can also be produced using transgenic animals that are engineered to contain human immunoglobulin loci.
  • transgenic animals that are engineered to contain human immunoglobulin loci.
  • WO 98/24893 discloses transgenic animals having a human Ig locus wherein the animals do not produce functional endogenous immunoglobulins due to the inactivation of endogenous heavy and light chain loci.
  • WO 91/10741 also discloses transgenic non-primate mammalian hosts capable of mounting an immune response to an immunogen, wherein the antibodies have primate constant and/or variable regions, and wherein the endogenous immunoglobulin-encoding loci are substituted or inactivated.
  • WO 96/30498 discloses the use ofthe Cre/Lox system to modify the immunoglobulin locus in a mammal, such as to replace all or a portion ofthe constant or variable region to form a modified antibody molecule.
  • WO 94/02602 discloses non-human mammalian hosts having inactivated endogenous Ig loci and functional human Ig loci.
  • U.S. Patent No. 5,939,598 discloses methods of making transgenic mice in which the mice lack endogenous heavy claims, and express an exogenous immunoglobulin locus comprising one or more xenogeneic constant regions.
  • an immune response can be produced to a selected antigenic molecule, and antibody-producing cells can be removed from the animal and used to produce hybridomas that secrete human monoclonal antibodies.
  • Immunization protocols, adjuvants, and the like are known in the art, and are used in immunization of, for example, a transgenic mouse as described in WO 96/33735.
  • This publication discloses monoclonal antibodies against a variety of antigenic molecules including IL-6, IL-8, TNF, human CD4, L-selectin, gp39, and tetanus toxin.
  • the monoclonal antibodies can be tested for the ability to inhibit or neutralize the biological activity or physiological effect ofthe corresponding protein.
  • WO 96/33735 discloses that monoclonal antibodies against IL-8, derived from immune cells of transgenic mice immunized with IL-8, blocked IL-8-induced functions of neutrophils. Human monoclonal antibodies with specificity for the antigen used to immunize transgenic animals are also disclosed in WO 96/34096.
  • proteins and variants thereof according to the invention are used to immunize a transgenic animal as described above.
  • Monoclonal antibodies are made using methods known in the art, and the specificity ofthe antibodies is tested using isolated colon or colorectal proteins according to the invention.
  • Methods for preparation ofthe human or primate protein according to the invention or an epitope thereof include, but are not limited to chemical synthesis, recombinant DNA techniques or isolation from biological samples. Chemical synthesis of a peptide can be performed, for example, by the classical Merrifeld method of solid phase peptide synthesis (Merrifeld, J. Am. Chem. Soc.
  • Polyclonal antibodies can be prepared by immunizing rabbits or other animals by injecting antigen followed by subsequent boosts at appropriate intervals. The animals are bled and sera assayed against purified protein usually by ELISA or by bioassay based upon the ability to block the action of a gene according to the invention.
  • the antibody can be isolated from the yolk ofthe egg.
  • Monoclonal antibodies can be prepared after the method of Milstein and Kohler by fusing splenocytes from immunized mice with continuously replicating tumor cells such as myeloma or lymphoma cells. [Milstein and Kohler, Nature 256:495-491 (1975); Gulfre and Milstein, Methods in Enzymology: Immunochemical Techniques 75:1-46, Langone and Banatis eds., Academic Press, (1981) which are inco ⁇ orated by reference]. The hybridoma cells so formed are then cloned by limiting dilution methods and supemates assayed for antibody production by ELISA, RIA or bioassay.
  • Another aspect of the present invention provides for a method for preventing or treating diseases involving overexpression ofthe a protein according to the invention by treatment of a patient with antibodies to specific tumor antigen according to the invention.
  • antibodies either polyclonal or monoclonal, to the protein can be produced by any suitable method known in the art as discussed above.
  • murine or human monoclonal antibodies can be produced by hybridoma technology or, alternatively, the tumor protein, or an immunologically active fragment thereof, or an anti-idiotypic antibody, or fragment thereof can be administered to an animal to elicit the production of antibodies capable of recognizing and binding to the tumor protein.
  • Antibodies can be of any class or subclass, e.g., IgG, IgA, lgM, IgD, and IgE or in the case of avian species, IgY, and subclasses thereof.
  • HTS high- throughput screening methods
  • Model systems are available that can be adapted for use in high throughput screening for compounds that inhibit the interaction of a protein with its ligand, for example by competing with the protein for ligand binding.
  • Sarabbi et al, Anal. Biochem. 237:10-15 (1996) describe cell-free, non-isotopic assays for discovering molecules that compete with natural ligands for binding to the active site of IL-1 receptor.
  • Martens, C. et al, Anal Biochem. 273:20-31 (1999) describe a generic particle-based nonradioactive method in which a labeled ligand binds to its receptor immobilized on a particle; label on the particle decreases in the presence of a molecule that competes with the labeled ligand for receptor binding.
  • the therapeutic gene polynucleotides and polypeptides ofthe present invention can be utilized in gene delivery vehicles.
  • the gene delivery vehicle can be of viral or non- viral origin (see generally, Jolly, Cancer Gene Therapy 1:51-64 (1994); Kimura, Human Gene Therapy 5:845-852 (1994); Connelly, Human Gene Therapy 1:185-193 (1995); and Kaplitt, Nature Genetics 6:148-153 (1994)).
  • Gene therapy vehicles for delivery of constructs including a coding sequence of a therapeutic according to the invention can be administered either locally or systemically. These constructs can utilize viral or non- viral vector approaches. Expression of such coding sequences can be induced using endogenous mammalian or heterologous promoters. Expression ofthe coding sequence can be either constitutive or regulated.
  • the present invention can employ recombinant retrovirases which are constructed to carry or express a selected nucleic acid molecule of interest.
  • Retroviras vectors that can be employed include those described in EP 0415 731; WO 90/07936; WO 94/03622; WO 93/25698; WO 93/25234; U.S. Patent No. 5,219,740; WO 93/11230; WO 93/10218; Vile and Hart, Cancer Res. 53:3860-3864 (1993); Vile and Hart, Cancer Res. 53:962-967 (1993); Ram et al., Cancer Res. 53:83-88 (1993); Takamiya et al., J. Neurosci. Res.
  • Recombinant retrovirases useful in accordance with the present invention include those described in WO 91/02805.
  • Packaging cell lines suitable for use with the above-described retroviral vector constructs can be readily prepared (see PCT publications WO 95/3 0763 and WO 92/05266), and used to create producer cell lines (also termed vector cell lines) for the production of recombinant vector particles.
  • producer cell lines also termed vector cell lines
  • packaging cell lines can be prepared from human (such as HT1080 cells) or mink parent cell lines, thereby allowing production of recombinant retrovirases that can survive inactivation in human serum.
  • the present invention also employs alphaviras-based vectors that can function as gene delivery vehicles.
  • Vectors can be constructed from a wide variety of alphavirases, including, for example, Sindbis virus vectors, Semliki forest virus (ATCC VR-67; ATCC VR-1247), Ross River virus (ATCC VR-373; ATCC VR-1246) and Venezuelan equine encephalitis virus (ATCC VR-923; ATCC VR-1250; ATCC VR 1249; ATCC VR-532).
  • Representative examples of such vector systems include those described in U.S. Patent Nos. 5,091,309; 5,217,879; and 5,185,440; and PCT Publication Nos. WO 92/10578; WO 94/21792; WO 95/27069; WO 95/27044; and WO 95/07994.
  • Gene delivery vehicles ofthe present invention can also employ parvovirus such as adeno-associated virus (AAV) vectors.
  • AAV adeno-associated virus
  • Representative examples include the AAV vectors disclosed by Srivastava in WO 93/09239, Samulski et al., J. Vir. 63: 3822-3828 (1989); Mendelson et al., Virol 166: 154-165 (1988); and Flotte et al., P.NA.S. 90: 10613-10617 (1993).
  • adenoviral vectors include those described by Berkner, Biotechniques 6:616-621 (Biotechniques); Rosenfeld et al, Science 252:431-434 (1991); WO 93/19191; Kolls et al, P.N.A.S. 215-219 (1994); Kass-Bisleret al., P. ⁇ .A.S. 90: 11498- 11502 (1993); Guzman et al., Circulation 88: 2838-2848 (1993); Guzman et al, Cir. Res. 73: 1202-1207 (1993); Zabner et al., Cell 75: 207-216 (1993); Li et al., Hum. Gene Ther.
  • adenoviral gene therapy vectors employable in this invention also include those described in WO 94/12649, WO 93/03769; WO 93/19191 ; WO 94/28938; WO 95/11984 and WO 95/00655.
  • Administration of D ⁇ A linked to kill adenoviras as described in Curiel, Hum. Gene Ther. 3: 147-154 (1992) can be employed.
  • eukaryotic cell delivery vehicles cells for example see U.S. Serial No. 08/240,030, filed May 9, 1994, and U.S. Serial No. 08/404,796; deposition of photopolymerized hydrogel materials; hand-held gene transfer particle gun, as described in U.S. Patent No. 5,149,655; ionizing radiation as described in U.S. Patent No. 5,206,152 and in WO 92/11033; nucleic charge neutralization or fusion with cell membranes. Additional approaches are described in Philip, Mol. Cell Biol. -7 ⁇ :2411-2418 (1994), and in Woffendin, Proc. Natl Acad. Sci. 97:1581-1585 (1994).
  • Naked DNA can also be administered directly to a subject.
  • Exemplary naked DNA introduction methods are described in WO 90/11092 and U.S. Patent No. 5,580,859. Uptake efficiency may be improved using biodegradable latex beads. DNA coated latex beads are efficiently transported into cells after endocytosis initiation by the beads. The method may be improved further by treatment of the beads to increase hydrophobicity and thereby facilitate disruption ofthe endosome and release ofthe DNA into the cytoplasm.
  • Liposomes that can act as gene delivery vehicles are described in U.S. Patent No. 5,422,120, PCT Patent Publication Nos. WO 95/13 796, WO 94/23697, and WO 9 1/14445, and EP No. 0 524968.
  • non- viral delivery suitable for use includes mechanical delivery systems such as the approach described in Woffendin et al., Proc. Natl. Acad. Sci. USA 91(24): 11581- 11585 (1994).
  • the coding sequence and the product of expression of such can be delivered through deposition of photopolymerized hydrogel materials.
  • Other conventional methods for gene delivery that can be used for delivery ofthe coding sequence include, for example, use of hand-held gene transfer particle gun, as described in U.S. Patent No. 5,149,655; use of ionizing radiation for activating transferred gene, as described in U.S. Patent No. 5,206,152 and PCT Patent Publication No. WO 92/11033.
  • Example 1 Identification of CICO1-CICO3 Genes Through a collaboration with Analytical Pathology Medical Group (at Grossmont Hospital), IDEC obtained pairs of snap frozen normal and malignant colon tissue removed during surgery.
  • human tentative human consensus sequence (THC) 684921 was identified from the BLAST database.
  • chrl_70_2399.f mRNA Sequence (coding sequence in CAPITALS, no ATG at start) aagttgccccacctctctgagcattggcttccccatctgtgaaagaggag tgctgatgtttgccttctaggggcctagtgaggcttaagggtgagcagca ggcacacagaaagctagaaatacaggatcactgtgggacggtggggctgg ccacctgggcaggccacttacccagcggccccctctgtctccaggtgttc atcggcgtaaactgtctgagcacagacttttcctcacaaaagggggtgaa gggtgtceccctgaacctgcagattgacacctatgactgtggcttg
  • nucleotide sequences of each candidate gene are listed below. The first sequence listed for each candidate gene was obtained directly from the public NCBI database
  • Candidate 1 GENBANK Accession No. W91975 W91975/IMAGE Clone 415310 3' mRNA Sequence
  • Candidate 3 GENBANK Accession No. AI680111 AI680111/IMAGE Clone 22520293' mRNA Sequence TTTTTTTTTTGT- ⁇ teATAAATATATTAGC-AAATGAAT ⁇
  • GAPDH Glyceraldehyde 3 -phosphate dehydrogenase
  • the following primers were used to amplify a 507 base pair product ofthe candidate 3 gene: 5 ' TCCCACCCGCTGTACCTGTGC 3 ' (SEQ ID NO : 58) 5 ' CCTGCAGCTGGCCTGGTACCT 3 ' (SEQ ID NO : 59)
  • RT reverse transcriptase
  • GAPDH GAPDH
  • the positive control for candidate 3 was IMAGE 2324560, obtained from the ATCC.
  • the following primers were used to amplify a 415 base pair product ofthe candidate 3 gene:
  • Example 2 Using the same technology employed in Example 1 to identify the CICO genes, the following sequences were identified as differentially expressed in colon cancer:
  • This protein contains a transmembrane domain as determined by SMART (shown below), SOSUI, and TmPred. SMART also predicts that this protein contains a RING domain, which is a zinc finger domain involved in protein: protein interactions. The stracture ofthe protein is depicted schematically below:
  • EXAMPLE 4 Using the Gene Logic database and the methods described generally in Example 2, the following additional DNA sequences were identified as being overexpressed in colon tumor tissue:
  • Fragment AA781143 was upregulated 4.16-fold in the colon samples when compared to mixed normal tissue. E-Northem analysis of this fragment demonstrates that it is expressed in 69% ofthe colon tumors with greater than 50% malignant cells and shows little or no expression in normal tissues. See Figure 8.
  • the GeneLogic database calls this protein "hypothetical protein from EUROIMAGE 2021883.”
  • EUROIMAGE 2021883 Nucleotide Sequence CCAGAGTTTGTCTTCTACGACCAGCTGAAGCAAGTGATGAATGCGTACAGAGTCAAGCCGGC CGTCTTTGACCTGCTCCTGGCTGTTGGCATTGCTGCCTACCTCGGCATGGCCTACGTGGCTG TCCAGCACTTCAGCCTCCTCTACAAGACCGTCCAGAGGCTGCTCGTGAAGGCCAAGACACAG TGACACAGCCACCCCCACAGCCGGAGCCCCCGCCGCTCCACAGTCCCTGGGGCCGAGCACGA GTGAGTGGACACTGCCCCGCCGCGGGCGGCCCTGCAGGGACAGGGGCCCTCTCTCCCTCCGG CGGTGGTTGGAACACTGAATTACAGAGCTTTTTTCTGTTGCTCTCCGAGACTGGGGGGGGAT TGTTTCTTCTTCCTTGTCTTTGAACTTCCTTGGAGGAGCTTGGGAGACGTCCCGGGGC CAGGCTACGGACTTGCGGACGAGCCCCCCAGTCCTGGGAGCCGGCCAGTCCTGGGG
  • the protein set forth above contains one TM (transmembrane domain) by SMART, SOSUI, and TmPred prediction programs.
  • TM transmembrane domain
  • SOSUI SOSUI
  • TmPred prediction programs TmPred prediction programs.
  • BLAST database and EST sequences suggest that the following alternative nucleotide and protein sequences correspond to AA781143:
  • GENBANK also identifies RefSeq Loc56926 as corresponding to AA781143, which nucleotide and protein sequences are set forth below:
  • Loc56926 in normal and malignant human tissues was further investigated by PCR experiments using commercially available human cDNA panels and cDNA samples prepared in-house from human tissues and cell lines. See Figures 9A-9B, 10A-10B, 11 A- 1 IB, and 12A-12B. Expression of Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was measured in these experiments as a control for cDNA integrity.
  • GAPDH is a housekeeping gene expressed abundantly in all human tissues. The following primers were used to amplify a 482 base pair product ofthe GAPDH gene:
  • the positive confrol for Loc56926 IMAGE clone 4428206 was obtained from the ATCC.
  • Primers used to amplify a 283 base pair product of Loc56926 were: 5 ⁇ AATGCAGTGCTGAACACGGAG 3 ' (SEQ ID NO:64)
  • fragment AW779536 was upregulated 3.7 fold.
  • E- Northern analysis shown in Figure 13 demonstrates that the fragment is expressed in 77% of the tumors and poorly expressed in normal tissue.
  • This gene encodes a protein having the following predicted stracture:
  • fragment AL531683 was found to be upregulated 3.76- fold.
  • the E-Northem analysis shown in Figure 14 demonstrates that the fragment is expressed in 100% ofthe tumors analyzed and poorly expressed in normal tissue.
  • fragment AI202201 was upregulated 3.18-fold.
  • E- Northern analysis shown in Figure 15 demonstrates that the fragment is expressed in 77% of the tumors and poorly expressed in normal tissue.
  • fragment AL389942 was upregulated 3.83-fold.
  • E- Northern analysis shown in Figure 16 demonstrates that the fragment is expressed in 55% of the tumors and poorly expressed in normal tissue.
  • DNA fragment NM_021246 is 5 -fold upregulated as shown by hybridization in the malignant colon when compared with mixed normal samples, greater than 3 -fold upregulated compared with normal kidney, liver and lung, and greater than 2-fold upregulated in all other tissues.
  • NM_021246 Nucleotide Sequence
  • the amino acid sequence for Ly6G6D is set forth below:
  • AI821606 is in the 3 'UTR of predicted genes corresponding to both strands of a chromosome. Based thereon, this fragment could be part ofthe following genes:
  • SMART analysis identified three transmembrane domains (rectangles) and a signal sequence.
  • the predicted stracture ofthe protein is depicted schematically below:

Abstract

Nucleic acids and proteins that are overexpressed in colon or colorectal tumor tissues, and which are useful diagnostic and therapeutic targets.

Description

NOVEL GENE TARGETS AND LIGANDS THAT BIND THERETO FOR TREATMENT AND DIAGNOSIS OF COLON CARCINOMAS
RELATED APPLICATIONS This application relates to U.S. Provisional Patent Application Serial No. 60/367,727 filed March 28, 2002 , U.S. Provisional Patent Application Serial No. 60/381,328 filed May 20, 2002, U.S. Provisional Patent Application Serial No. 60/386, 747 filed June 10, 2002, and U.S. Provisional Patent Application Serial No. 60/427,564 filed November 20, 2002, each of which are incorporated by reference in their entirety herein.
FIELD OF THE INVENTION The present invention relates the identification of gene targets for treatment and diagnosis of neoplastic diseases, such as colon or colorectal cancer, and other cancers wherein the subject genes are upregulated and the use thereof to express the corresponding antigen, and to produce ligands that specifically bind such antigen, e.g. monoclonal antibodies and small molecules.
DESCRIPTION OF RELATED ART Colorectal cancers are among the most common cancers in men and women in the U.S. and are one ofthe leading causes of death. Other than surgical resection no other systemic or adjuvant therapy is available. Vogelstein and colleagues have described the sequence of genetic events that appear to be associated with the multistep process of colon cancer development in humans (Fearon and Vogelstein, 1990). An understanding ofthe molecular genetics of carcinogenesis, however, has not led to preventative or therapeutic measures. It can be expected that advances in molecular genetics will lead to better risk assessment and early diagnosis but colorectal cancers will remain a deadly disease for a majority of patients due to the lack of an adjuvant therapy.
Endogenous gastrins and exogenous gastrins (other than tetragastrin) seem to promote the growth of established colon cancers in mice (Singh, et al., 1986; Singh, et al., 1987; et al., 1984; Smith and Solomon, 1988; Singh, et al., 1990; Rehfeld and van Solinge, 1994) and promote carcinogen induced colon cancers in rats (Williamson et al., 1978; Karlin et al., 1985; Lamoste and Willems; 1988). Recent studies of Montag et al (1993) further support a possible co-carcinogenic role of gastrin in the initiation of tumors.
Many colon cancer cells express and secrete gastrin gene products (Dai et al., 1992; Kochinan et al., 1992; Finley et al., 1993; Van Solinge et al., 1993; Xu et al., 1994; Singh et al., 1994a; Hoosein et al., 1988; Hoosein et al., 1990) and bind gastrin-like peptides (Singh et al., 1986; Singh et al., 1987; Weinstock and Baldwin, 1988; Watson and Steele, 1994; Upp et al., 1989; Singh et al., 1985). In previous reports gastrin antibodies were either reported to inhibit (Hoosein et al., 1988; Hoosein et al, 1990) the growth of colon cancer cell lines in vitro.
However other investigators have had inconclusive results with colon cancer cell lines. A number of studies testing the effects of gastrin on cell proliferation of cancer cells have been performed (Sirinek et al., 1985; Kusyk et al., 1986; Watson et al, 1989). The results have varied widely. In one study, four different human cancer cell lines were tested for growth stimulation by pentagastrin and only one showed growth stimulation (Eggstein et al., 1991). Similarly in majority ofthe studies conducted to-date, mitogenic effects of gastrin have been demonstrated only on a very small percentage of colon cancer cell lines (Hoosein et al., 1988; Hoosein et al, 1990; Shrink et al, 1985; Kusyk et al, 1986; Guo et al, 1990; Ishizuka et al, 1994).
Since only a small percentage of established human colon cancer cell lines demonstrated a growth response to exogenous gastrins, investigators in this field came to believe that gastrin probably did not play a significant role in the growth of colon cancers. The recent discovery that human colon cancer cell lines and primary human colon cancers express the gastrin gene has sparked a renewed interest in a possible autocrine role of gastrin- like peptides in colon cancers. However, significant skepticism remains in the field, to date, regarding the importance of gastrin gene expression to the continued growth and tumorigenicity of colon cancers.
Thus, to-date, no systemic or adjuvant therapies have been developed for colon cancers, based on the knowledge that a significant percentage of human colon cancers express the gastrin gene. In fact, no adjuvant or systemic therapy has been developed for colon cancers that is based on the knowledge ofthe expression of other growth factors such as TGF-alpha. or IGF-II, since none ofthe growth factors demonstrate a significant growth effect on majority ofthe colon cancer cell lines in culture. At the present time the only systemic treatment available for colon cancer is chemotherapy. However, chemotherapy has not proven to be very effective for the treatment of colon cancers for several reasons, in part because colon cancers express high levels ofthe MDR gene (that codes for multi-drug resistance gene products). The MDR gene products actively transport the toxic substances out ofthe cell before the chemotherapeutic agents can damage the DNA machinery ofthe cell. These toxic substances harm the normal cell populations more than they harm the colon cancer cells for the above reasons. There is no effective systemic treatment for treating colon cancers other than surgically removing the cancers. In the case of several other cancers, including breast cancers, the knowledge of growth promoting factors (such as EGF, estradiol, IGF-II) that appear to be expressed or effect the growth ofthe cancer cells, has been translated for treatment purposes. But in the case of colon cancers this knowledge has not been applied and therefore the treatment outcome for colon cancers remains bleak.
Antisense RNA technology has been developed as an approach to inhibiting gene expression, including oncogene expression. An "antisense" RNA molecule is one which contains the complement of, and can therefore hybridize with, protein-encoding RNAs ofthe cell. It is believed that the hybridization of antisense RNA to its cellular RNA complement can prevent expression ofthe cellular RNA, perhaps by limiting its franslatability. While various studies have involved the processing of RNA or direct introduction of antisense RNA oligonucleotides to cells for the inhibition of gene expression (Brown, et al., 1989; Wicksfrom, et al., 1988; Smith, et al., 1986; Buvoli, et al., 1987), the more common means of cellular introduction of antisense RNAs has been through the construction of recombinant vectors that express antisense RNA once the vector is introduced into the cell.
A principle application of antisense RNA technology has been in connection with attempts to affect the expression of specific genes. For example, Delauney, et al. have reported the use antisense transcripts to inhibit gene expression in transgenic plants (Delauney, et al., 1988). These authors report the down-regulation of chloramphenicol acetyl transferase activity in tobacco plants transformed with CAT sequences through the application of antisense technology.
Antisense technology has also been applied in attempts to inhibit the expression of various oncogenes. For example, Kasid, et al., 1989, report the preparation of recombinant vector construct employing Craf-1 cDNA fragments in an antisense orientation, brought under the control of an adenoviras 2 late promoter. These authors report that the introduction of this recombinant construct into a human squamous carcinoma resulted in a greatly reduced tumorigenic potential relative to cells transfected faith control sense transfectants. Similarly, Prochownik, et al., 1988, have reported the use of Cmiyc antisense constructs to accelerate differentiation and inhibit G.sub.l progression in Friend Murine Erythroleukemia cells. In contrast, Khokha, et al., 1989, discloses the use of antisense RNAs to confer oncogenicity on 3T3 cells, through the use of antisense RNA to reduce murine tissue inhibitor or metalloproteinases levels.
Antisense methodology takes advantage ofthe fact that nucleic acids tend to pair with "complementary" sequences. By complementary, it is meant that polynucleotides are those which are capable of base-pairing according to the standard Watson-Crick complementary rales. That is, the larger purines base pair with the smaller pyrimidines to form combinations of guanine paired with cytosine (G:C) and adenine paired with either thymine (A:T) in the case of DNA, or adenine paired with uracil (A:U) in the case of RNA. Inclusion of less common bases such as inosine, 5-methylcytosine, 6-methyladenine, hypoxanthine and others in hybridizing sequences does not interfere with pairing.
Targeting double-stranded (ds) DNA with polynucleotides leads to triple-helix formation; targeting RNA leads to double-helix formation. Antisense polynucleotides, when introduced into a target cell, specifically bind to their target polynucleotide and interfere with transcription, RNA processing, transport, translation and/or stability. Antisense RNA constructs, or DNA encoding such antisense RNAs, can be employed to inhibit gene transcription or translation or both within a host cell, either in vitro or in vivo, such as within a host animal, including a human subject.
Throughout this application, the term "expression vector or construct" is meant to include any type of genetic construct containing a nucleic acid coding for a gene product in which part or all ofthe nucleic acid encoding sequence is capable of being transcribed. The transcript can be translated into a protein but it need not be. Thus, in certain embodiments, expression includes both transcription of a gene and translation of mRNA into a gene product. In other embodiments, expression only includes transcription ofthe nucleic acid encoding a gene of interest.
The nucleic acid encoding a gene product is under transcriptional control of a promoter. A "promoter" refers to a DNA sequence recognized by the synthetic machinery of the cell, or introduced synthetic machinery, required to initiate the specific transcription of a gene. The phrase "under transcriptional control" means that the promoter is in the correct location and orientation in relation to the nucleic acid to control RNA polymerase initiation and expression ofthe gene.
The term promoter is used to refer to a group of transcriptional control modules that are clustered around the initiation site for RNA polymerase II. Much ofthe thinking about how promoters are organized derives from analyses of several viral promoters, including those for the HSV thymidine kinase (tk) and SV40 early transcription units. These studies, augmented by more recent work, have shown that promoters are composed of discrete functional modules, each consisting of approximately 7-20 base pairs of DNA, and containing one or more recognition sites for transcriptional activator or repressor proteins. At least one module in each promoter functions to position the start site for RNA synthesis. The best known example of this is the TATA box, but in some promoters lacking a TATA box, such as the promoter for the mammalian terminal deoxynucleotidyl transferase gene and the promoter for the SV40 late genes, a discrete element overlying the start site itself helps to fix the place of initiation.
Additional promoter elements regulate the frequency of transcriptional initiation. Typically, these are located in the region 30-110 base pairs upstream ofthe start site, although a number of promoters have recently been shown to contain functional elements downstream ofthe start site as well. The spacing between promoter elements frequently is flexible, so that promoter function is preserved when elements are inverted or moved relative to one another. In the tk promoter, the spacing between promoter elements can be increased to 50 base pairs apart before activity begins to decline. Depending on the promoter, it appears that individual elements can function either cooperatively or independently to activate transcription.
A promoter is selected based on its capability to direct gene expression in the targeted cell. Thus, where a human cell is targeted, the nucleic acid coding region can be positioned adjacent to and under the control of a promoter that is capable of being expressed in a human cell. Generally speaking, such a promoter might include either a human or viral promoter.
In various instances, the human cytomegalovirus (CMV) immediate early gene promoter, the S V40 early promoter and the Rous sarcoma virus long terminal repeat can be used to obtain high-level expression ofthe gene of interest. The use of other viral or mammalian cellular or bacterial phage promoters which are well known in the art to achieve expression of a gene of interest is contemplated as well, provided that the levels of expression are sufficient for a given purpose. By employing a promoter with well-known properties, the level and pattern of expression ofthe gene product following transfection can be optimized. Further, selection of a promoter that is regulated in response to specific physiologic signals can permit inducible expression ofthe gene product. Representative elements/promoters useful in accordance with the present invention include but are not limited to those listed below.
Enhancers were originally detected as genetic elements that increased transcription from a promoter located at a distant position on the same molecule of DNA. This ability to act over a large distance had little precedent in classic studies of prokaryotic transcriptional regulation. Subsequent work showed that regions of DNA with enhancer activity are organized much like promoters. That is, they are composed of many individual elements, each of which binds to one or more transcriptional proteins.
The basic distinction between enhancers and promoters is operational. An enhancer region as a whole must be able to stimulate transcription at a distance; this need not be true of a promoter region or its component elements. A promoter includes one or more elements that direct initiation of RNA synthesis at a particular site and in a particular orientation, whereas enhancers lack these specificities. Promoters and enhancers are often overlapping and contiguous, often seeming to have a very similar modular organization.
Viral promoters, cellular promoters/enhancers and inducible promoters/enhancers that could be used in combination with the nucleic acid encoding a gene of interest in an expression construct. Some examples of enhancers include Immunoglobulin Heavy Chain; Immunoglobulin Light Chain; T-Cell Receptor; HLA DQ a and DQ b b-Interferon; Interleukin-2; Interleukin-2 Receptor: Gibbon Ape Leukemia Virus; MHC Class II 5 or HLA- DRa; b-Actin; Muscle Creatine Kinase; Prealbumin (Transthyretin); Elastase I; Metallothionein; Collagenase, Albumin Gene; α-Fetoprotein; α-Globin; β-Globin; c-fos: c- HA-ras; Insulin Neural Cell Adhesion Molecule (NCAM); al-Antitrypsin; H2B (TH2B) Histone; Mouse or Type I Collagen; Glucose-Regulated Proteins (GRP94 and GRP78); Rat Growth Hormone; Human Serum Amyloid A (SAA); Troponin I (TN I); Platelet-Derived Growth Factor; Duchenne Muscular Dystrophy; SV40 or CMV; Polyoma; Retrovirases; Papilloma Virus; Hepatitis B Virus; Human Immunodeficiency Virus. Inducers such as phorbol ester (TFA) heavy metals; glucocorticoids; poly (rl)X; poly(rc); Ela; H2O2; IL 1 ; Interferon, Newcastle Disease Vims; A23187; IL-6; Serum; SV40 Large T Antigen; FMA; thyroid Hormone; could be used. Additionally, any promoter/enhancer combination (as per the Eukaryotic Promoter Data Base EPDB) could also be used to drive expression ofthe gene. Eukaryotic cells can support cytoplasmic transcription from certain bacterial promoters if the appropriate bacterial polymerase is provided, either as part ofthe delivery complex or as an additional genetic expression construct. In certain instances, the expression construct can comprise a virus or engineered construct derived from a viral genome. The ability of certain viruses to enter cells via receptor-mediated endocytosis and to integrate into host cell genome and express viral genes stably and efficiently have made them attractive candidates for the transfer of foreign genes into mammalian cells (Ridgeway, 1988; Nicolas and Rubenstein, 1988; Baichwal et al., 1986: Temin, 1986). The first viruses used as gene vectors were DNA viruses including the papo viruses (simian virus 40, bovine papilloma virus, and polyoma) (Ridgeway, 1988; Baichwal et al., 1986) and adenovirases (Ridgeway, 1988; Baichwal et al., 1986). These have a relatively low capacity for foreign DNA sequences and have a restricted host spectrum. Furthermore, their oncogenic potential and cytopathic effects in permissive cells raise safety concerns. They can accommodate only up to 8 kB of foreign genetic material but can be readily introduced in a variety of cell lines and laboratory animals (Nicolas and Rubenstein, 1988; Temin, 1986).
Where a cDNA insert is employed, a polyadenylation signal is typically inserted to effect proper polyadenylation ofthe gene transcript. Any suitable polyadenylation sequence can be used. An expression cassette can also include a terminator sequence. These elements enhance message levels and minimize read through from the cassette into other sequences.
It is understood in the art that to bring a coding sequence under the control of a promoter, or operatively linking a sequence to a promoter, one positions the 5' end ofthe transcription initiation site ofthe transcriptional reading frame ofthe protein between about land about 50 nucleotides "downstream" of (i.e., 3' of) the chosen promoter. In addition, where eukaryotic expression is contemplated, an appropriate polyadenylation site (e.g., 5'- AATAAA-3' (SEQ ID NO:66)) can be included if absent from the original cloned segment. Typically, the poly A addition site is placed about 30 to 2000 nucleotides "downstream" of the termination site ofthe protein at a position prior to transcription termination. The above background references are part ofthe present invention insofar as they are applicable to the invention described herein. Hence there are no effective and specific ways of treating or diminishing the growth of colorectal cancer to date. Therefore, there exists a significant need for the identification of novel gene targets for the treatment and diagnosis of colon or colorectal cancer, especially given the huge human toll caused by this disease annually.
SUMMARY OF THE INVENTION
It is an aspect ofthe invention to identify novel gene targets for freatment and the diagnosis of cancer, such as colon or colorectal cancer.
It is a specific aspect ofthe invention to develop novel therapies for treatment of cancer, such as colon cancer, involving the administration of anti-sense oligonucleotides corresponding to gene targets that are expressed by certain colon or colorectal cancers. It is another specific aspect ofthe invention to provide the antigens expressed by genes that are expressed by malignant tissues, e.g., colon or colorectal cancers.
It is another specific aspect ofthe invention to produce ligands that bind antigens expressed by certain cancers, such as colon or colorectal cancers. Representative ligands include monoclonal antibodies.
It is another specific aspect ofthe invention to provide novel therapeutic regimens for the treatment of cancer, for example colon cancer, that involve the administration of antigens expressed by certain colon or colorectal cancers, alone or in combination with adjuvants that elicit an antigen-specific cytotoxic T-cell lymphocyte response against cancer cells that express such antigen.
It is another aspect ofthe invention to provide novel therapeutic regimens for the treatment of cancer, such as colon or colorectal cancer, that involve the administration of ligands, for example, monoclonal antibodies that specifically bind novel antigens that are expressed by certain cancer tissues including colon cancer tissues. It is another aspect ofthe invention to provide a novel method for diagnosis of cancer, for example colon or colorectal cancer, by using ligands, e.g., monoclonal antibodies, that specifically bind to antigens that are expressed by cancers including certain colon or colorectal cancers, in order to detect whether a subject has or is at increased risk of developing colon or colorectal cancer. It is another aspect ofthe invention to provide a novel method of detecting persons having, or at increased risk of developing certain types of cancers, including colon cancer by use of labeled DNAs that hybridize to novel gene targets expressed by certain cancers, including colon cancers.
It is yet another aspect ofthe invention to provide diagnostic test kits for the detection of persons having or at increased risk of developing certain cancer, including colon cancer that comprise a ligand, e.g., monoclonal antibody that specifically binds to an antigen expressed by certain colon cancers, and a detectable label, e.g., a radiolabel or fluorophore.
It is another aspect ofthe invention to provide diagnostic kits for detection of persons having or at risk of developing certain cancers, including colon cancer that comprise DNA primers or probes specific for novel gene targets expressed by colon cancers, and a detectable label, e.g. radiolabel or fluorophore.
BRIEF DESCRIPTION OF THE DRAWINGS Figure 1 summarizes expression data for the CICO1, CICO2 and CICO3, which were identified based on overexpression in colon cancer as described in Example 1. Figures 2-5 depict gene expression profiles determined using the Gene Logic datasuite as described in Example 2. The values along the y-axis represent expression intensities in Gene Logic units. Each blue circle represents an individual patient sample. The bar graph on the left ofthe figure depicts the percentage of each tissue type found to express the gene fragment. The total number of samples for each tissue type is as follows: colon tumor, tumor % above 50, 31 ; colon tumors, 45; normal breast, 37; normal colon, 30; normal esophagus, 18, normal kidney, 28; normal liver, 21; normal lung, 35; normal lymph node 10; normal ovary, 25; normal pancreas, 20; normal prostate, 20; normal rectum, 22; normal stomach, 25. "Colon tumor, tumor % above 50" refers to tumor samples for which at least 50% of each sample comprises malignant tissue, as determined by a pathologist. This sample set is a subset of colon tumors, which comprises all colon tumor samples contained within the Gene Logic database.
Figure 2 depicts the gene expression profile of Candidate 1, which was determined using the Gene Logic datasuite for GENBANK Accession No. W91975 as described in Example 2. Candidate 1 is overexpressed in colon tumor tissue. Figure 3 depicts the gene expression profile of Candidate 2, which was determined using the Gene Logic datasuite for GENBANK Accession No. Al 694242 as described in Example 2. Candidate 2 is overexpressed in colon tumor tissue. Figure 4 contains the gene expression profile of Candidate 3, which was determined using the Gene Logic datasuite for GENBANK Accession No. AI680111 as described in Example 2. Candidate 3 is overexpressed in colon tumor tissue.
Figure 5 depicts the gene expression profile of Candidate 4, which was determined using the Gene Logic datasuite for GENBANK Accession No. AA813827 as described in Example 2. Candidate 4 is overexpressed in colon tumor tissue.
Figures 6 A and 6B show PCR data of Candidate 3 expression (Figure 6 A) and GAPDH expression (Figure 6B) in normal human tissues. Candidate 3 was screened against Human Multiple Tissue cDNA panels I & II (Clontech #K1420-1 & # K1421-1 ) according to the manufacturer's instructions. GAPDH was not tested against the prostate sample. The positive confrol for Candidate 3 was IMAGE 2324560, obtained from the American Tissue Type Collection (Manassas, Virginia). The cDNA samples present in each lane are as follows: lane 1, heart; lane 2, brain; lane 3, placenta; lane 4, lung; lane 5, liver; lane 6, skeletal muscle; lane 7, kidney; lane 8, pancreas; lane 9, spleen; lane 10, thymus; lane 11, prostate; lane 12, testis; lane 13, ovary; lane 14, small intestine; lane 15, colon; lane 16, peripheral blood leukocytes; lane 17, positive control; lane 18, negative control. Arrow denotes the anticipated size ofthe PCR product for candidate 3. The results shown in this figure indicate that candidate 3 is not expressed at detectable levels in any ofthe normal tissues tested.
Figures 7A and 7B show PCR data of Candidate 3 expression (Figure 7A) and GAPDH expression (Figure 7B) in colon tumor samples. The cDNA samples present in each lane are as follows: lane 1, grade 3 adenocarcinoma; lane 2, grade 2 adenocarcinoma; lane 3, grade 1 adenocarcinoma; lane 4, grade 2 adenocarcinoma; lane 5, colorectal cancer cell line HCT116; lane 6, positive control (IMAGE clone); lane 7, negative confrol. Arrow denotes the anticipated size ofthe PCR product for candidate 3. The results shown in this figure indicate that candidate 3 is expressed in at least 3 of 4 colon tumor samples in addition to colorectal tumor cell line HCT116.
Figure 8 depicts E-Northern expression data for Loc 56926, which is overexpressed in colon cancer, as described in Example 4.
Figures 9A and 9B are PCR panels showing expression of Loc56926 (Figure 9A) and GAPDH (Figure 9B) in malignant colon samples. The cDNA samples present in each lane are as follows: lane M, marker; lane 1, no template control; lane 2 colon cancer 8T; lane 3, colon cancer DT; lane 4, colon cancer FT; lane 5, colon cancer GT; lane 6, colon cancer HT; lane 7, colon cancer IT; lane 8, colon cancer QT; lane 9, prostate cancer OT; lane 10, colon cancer RT; lane 11, colon cancer cell line HCT116; lane 12, positive control EST. The results from this figure demonstrate that Loc56926 expression is present in cDNA from three of eight tested colon cancer samples. Figures 10A and 10B are PCR panels showing expression of Loc56926 (Figure 10 A) and GAPDH (Figure 10B) in normal human tissues. Hybridization was performed using Human Multiple Tissue cDNA panel I (Clontech #K1420-1) according to the manufacturer's instructions. The cDNA samples present in each lane are as follows: lane M, marker; lane 1, no template control; lane 2, colon tumor 8T; lane 3, colon tumor HT; lane 4, colon tumor RT; lane 5, colon cancer cell line HCT116; lane 6, normal colon; lane 7, normal brain; lane 8, normal heart; lane 9, kidney; lane 10, normal liver; lane 11, normal lung; lane 12, skeletal muscle; lane 13, normal pancreas; lane 14, normal placenta lane 15; EST control. These results demonstrate that Loc56926 is present in colon tumors with light expression in the normal pancreas (note the increase in GAPDH in the pancreas lane compared to the colon tumor lanes) and not expressed at detectable levels the other tested normal human tissues.
Figures 11 A and 1 IB are PCR panels showing expression of Loc56926 (Figure 11 A) and GAPDH (Figure 1 IB) in human tissues. Hybridization was performed using Human Multiple Tissue cDNA panel II (Clontech # K1421-1) according to the manufacturer's instructions. The cDNA samples present in each lane are as follows: lane M, marker; lane 1, no template control; lane 2, colon tumor 8T; lane 3, colon tumor HT; lane 4, colon tumor RT; lane 5, colon cancer cell line HCT116; lane 6, normal colon; lane 7, normal peripheral blood leukocytes; lane 8, small intestine; lane 9, normal ovary; lane 10, normal prostate; lane 11, normal spleen; lane 12, normal testis; lane 13, normal thymus; lane 14, EST confrol. These results demonstrate that Loc56926 is not expressed at detectable levels in these normal tissues.
Figures 12A and 12B are PCR panels showing expression of Loc56926 (Figure 12A) and GAPDH (Figure 12B) in normal brain tissue samples. Hybridization was performed using Normal Neural System cDNA panel (Biochain, C8234503, C8234504, C8234505). The cDNA samples present in each lane are as follows: lane M, marker; lane 1, no template confrol; lane 2, cerebellum; lane 3, cerebral cortex; lane 4, medulla oblongata; lane 5, pons; lane 6, frontal lobe; lane 7, occipital lobe; lane 8, parietal lobe; lane 9, temporal lobe; lane 10, placental neural system; lane 11, EST control. These results demonstrate that Lco56926 is not expressed at detectable levels in the normal brain.
Figure 13 depicts E-Northern expression data for the AW779536 gene, which is overexpressed in colon cancer, as described in Example 4. Figure 14 depicts E-Northern expression data for the AL531683 gene, which is overexpressed in colon cancer, as described in Example 4.
Figure 15 depicts E-Northern expression data for the AI202201 gene, which is overexpressed in colon cancer, as described in Example 4.
Figure 16 depicts E-Northern expression data for the AL389942 gene, which is overexpressed in colon cancer, as described in Example 4.
Figure 17 depicts E-Northern expression results for the Ly6G6Dgene, also described in Example 5.
Figure 18 depicts E-Northem expression results for FLJ32334, also described in Example 6.
DETAILED DESCRIPTION OF THE INVENTION The present invention relates to the identification of genes which are to be specifically expressed and upregulated in certain cancers, including colon or colorectal tumors. This was determined using the Gene Logic (Gaithersburg, Maryland) datasuite or Celera (Rockville, Maryland) database and by screening malignant colon tumor tissues as described in detail herein.
In particular, the present invention involves the discovery that certain genes, the nucleic acid sequences and predicted coding sequences of which are identified herein are specifically expressed in certain malignant tissues including colon or colorectal tumor tissues. The disclosed therapies involve the synthesis of oligonucleotides having sequences in the antisense orientation relative to the genes identified by the present inventors which are specifically expressed by malignant tissues, including colon or colorectal tumors. Suitable therapeutic antisense oligonucleotides typically vary in length from two to several hundred nucleotides in length, more typically about 50-70 nucleotides in length. These antisense oligonucleotides can be administered as naked DNAs or in protected forms, e.g., encapsulated in liposomes. The use of liposomal or other protected forms may enhance in vivo stability and delivery to target sites, i.e., colon tumor cells. Also, the subject novel genes can be used to design novel ribozymes that target the cleavage ofthe corresponding mRNAs in colon and other tumor cells. Similarly, these ribozymes can be administered in free (naked) form or by the use of delivery systems that enhance stability and/or targeting, e.g., liposomes. Ribozymal and antisense therapies used to target genes that are selectively expressed by cancer cells are well known in the art. Also, the present invention embraces the administration of use of DNAs that hybridize to the novel gene targets identified herein, attached to therapeutic effector moieties, for example radiolabels, including metallic and halogen isotopes (e.g., 90yttrium, I31iodine), cytotoxins, cytotoxic enzymes, in order to selectively target and kill cells that express these genes, . e. , colon tumor cells.
Still further, the present invention encompasses non-nucleic acid based therapies, for example antigens encoded by the nucleic acids disclosed herein. It is anticipated that these antigens can be used as therapeutic or prophylactic anti-tumor vaccines. For example, antigens ofthe present invention can be admimstered with adjuvants that induce a cytotoxic T lymphocyte response. Representative adjuvants include those disclosed in U.S. Patent Nos. 5,709,860, 5,695,770, and 5,585,103, which promote CTL responses against prostate and papillomavirus related human colon cancer. The disclosures of U.S. Patent Nos. 5,709,860, 5,695,770, and 5,585,103 are incorporated by reference in their entirety.
The disclosed antigens can be administered in combination with an adjuvant to elicit a humoral immune response against such antigens, thereby delaying or preventing the development of cancers (e.g., a colon cancer) associated with the overexpression ofthe antigens.
Embodiments ofthe invention comprise administration of one or more novel colon cancer antigens, for example in combination with an adjuvant. A representative adjuvant is PRO VAX®, which comprises a microfluidized adjuvant containing Squalene, TWEEN® and PLURONIC®, in an amount sufficient to be therapeutically or prophylactically effective. See U.S. Patent Nos. 5,709,860, 5,695,770, and 5,585,103. A typical dosage of formulated antigen ranges from about 50 to about 20,000 mg/kg body weight, or from about 100 to about 5000 mg/kg body weight. Alternatively, the subject tumor-associated antigens can be administered with other adjuvants, e.g., ISCOM®, DETOX™, SAF®, Freund's adjuvant, Alum, Saponin, among others. In another embodiment, the present invention provides methods for preparing monoclonal antibodies against the antigens encoded by the DNA sequences disclosed in the examples which are expressed specifically by certain malignant tissues including colon or colorectal tumor tissues. Monoclonal antibodies are produced by conventional methods and include human monoclonal antibodies, humanized monoclonal antibodies, chimeric monoclonal antibodies, single chain antibodies, including scFv's and antigen-binding antibody fragments such as Fabs, 2 Fabs, and Fab' fragments. Methods for the preparation of monoclonal antibodies and fragments thereof, for example by pepsin or papain-mediated cleavage, are well known in the art. In general, an appropriate (non-homologous) host is immunized with the subject colon cancer antigens, immune cells are isolated from the host and used to prepare hybridomas. Monoclonal antibodies that specifically bind to either of such antigens are identified by routine screening techniques. Useful monoclonal antibodies typically bind the target antigens with high affinity, e.g., possess a binding affinity (Kd) on the order of lO"6 to lO"10 M. Monoclonal antibodies and fragments ofthe invention are useful for anti-tumor immunotherapy. Optionally, therapeutic effector moieties (e.g., radiolabels, cytotoxins, therapeutic enzymes, agents that induce apoptosis) can be attached to the antibodies to provide for targeted cytotoxicity, i.e., killing of human colon tumor cells. Given the fact that the subject genes are apparently not significantly expressed by many normal tissues this should not result in significant adverse side effects (toxicity to non-target tissues).
Antibodies and/or antibody fragments are administered to a subject in labeled or unlabeled form, alone or in combination with other therapeutics, such as chemotherapeutics such as progestin, EGFR, TAXOL®, and the like. The administered composition can include a pharmaceutically acceptable carrier, and optionally adjuvants, stabilizers, etc., used in antibody compositions for therapeutic use.
The present invention also provides diagnostic methods for detection ofthe colon or colorectal tumor-specific genes disclosed herein. Diagnostic methods include detecting the expression of one or more of these genes at the DNA level or at the protein level. Patients who test positive for the disclosed tumor-specific genes diagnosed are identified as having or being at increased risk of developing colon cancer. Additionally, the levels of antigen expression can be useful in determining patient status, i.e., how far the disease has advanced. For example, the expression or expression level of a tumor-specific gene can indicate a particular stage of tumor progression.
At the DNA level, gene expression is detected by known DNA detection methods, including but not limited to Northern blot hybridization, strand displacement amplification (SDA), catalytic hybridization amplification (CHA), PCR amplification (for example, using primers corresponding to the novel genes disclosed herein), and other known DNA detection methods. For example, the presence or absence of cancer associated with the genes disclosed herein can be determined based on whether PCR products are obtained, and the level of expression. Expression levels can also be monitored to determine the prognosis of a colon cancer patient as the levels of expression ofthe PCR product likely increase as the disease progresses. Suitable controls and quantification is are performed for diagnostic methods as known in the art.
At the protein level, the status of a subject to be tested for colon cancer, or other cancer associated by overexpression of a gene disclosed herein, can be evaluated by testing biological fluids, such as blood, urine, colon tissue, with an antibody or antibodies or fragment that specifically binds to the novel colon tumor antigens disclosed herein. Methods of using antibodies to detect antigen expression are well known and include ELISA, competitive binding assays, and the like. Representative assays use an antibody or antibody fragment that specifically binds the target antigen directly or indirectly bound to a label that provides for detection, for example, a radiolabel, an enzyme, or a fluorophore.
As noted, the present invention provides novel genes and corresponding antigens that correlate to human colon cancer. The present invention also embraces variants thereof. By "variants" is intended sequences that are at least 75% identical thereto, for example at least 85% identical, or at least 90% identical when these DNA sequences are aligned to the subject DNAs or a fragment thereof having a size of at least 50 nucleotides. Representative variants include allelic variants.
The present invention also provides primers for amplification of nucleic acids encoding the subject novel genes or a portion thereof, which are present is a biological sample, for example, an mRNA library obtained from a desired cell source, including human colon cell or tissue samples. Typically, such primers are about 12 to 50 nucleotides in length and are constmcted such that they provide for amplification ofthe entire or most ofthe target gene. The present invention further provides antigens encoded by the disclosed DNAs or fragments thereof that bind to or elicit antibodies specific to the full-length antigens. Typically, such fragments are at least 10 amino acids in length, more typically at least 25 amino acids in length. The colon or colorectal tumor-specific genes ofthe invention are expressed in a majority of colon tumor samples tested. Some of these genes are also upregulated in other cancers. Thus, the present invention further contemplates identification of other cancers wherein the expression ofthe disclosed genes or variants thereof correlate to a cancer or an increased likelihood of cancer, for example breast, pancreas, lung or colon cancers. Also provided are compositions and methods to detect and treat such cancers.
"Isolated" refers to any human protein that is not in its normal cellular millieu. This includes by way of example compositions comprising recombinant protein, pharmaceutical compositions comprising purified protein, diagnostic compositions comprising purified protein, and isolated protein compositions comprising protein. In representative embodiments ofthe invention, an isolated protein comprises a substantially pure protein, in that it is substantially free of other proteins, for example, at least 90% pure, that comprises the amino acid sequence disclosed herein or natural homologues or mutants having essentially the same sequence. A naturally occurring mutant might be found, for instance, in tumor cells expressing a gene encoding a mutated protein sequence. "Native human protein" refers to a protein that comprises the amino acid sequence of the protein expressed in its endogenous environment, i.e., a human colon or colorectal tumor tissue.
"Native non-human primate protein" refers to a protein that is a non-human primate homologue ofthe protein having the amino acid sequence discussed in the examples. Given the phylogenetic closeness of humans to other primates, it is anticipated that human and non- human proteins expressed by the genes disclosed in the examples have non-human primate counterparts that possess amino acid sequences that are highly similar, such as 95% sequence identity or higher.
"Isolated human or non-human primate nucleic acid molecule or sequence" refers to a nucleic acid molecule that encodes human protein which is not in its normal human cellular millieu, e.g., is not comprised in the human or non-human primate chromosomal DNA. This includes by way of example vectors that comprise a nucleic acid molecule, a probe that comprises a gene nucleic acid sequence directly or indirectly attached to a detectable moiety, e.g. a fluorescent or radioactive label, or a DNA fusion that comprises a nucleic acid molecule encoding a colon antigen according to the invention fused at its 5' or 3' end to a different DNA, e.g. a promoter or a DNA encoding a detectable marker or effector moiety. Representative nucleic acid sequence encoding human proteins are disclosed herein. Also included are natural homologues or mutants having substantially the same sequence. Naturally occurring homologies that are degenerate would encode the same protein as discussed herein in the examples, but would include nucleotide differences that do not change the corresponding amino acid sequence. Naturally occurring mutants might be found in tumor cells, wherein such nucleotide differences result in a mutant protein. Naturally occurring homologues containing conservative substitutions are also encompassed.
"Variant of human or non-human primate protein" refers to a protein possessing an amino acid sequence that possess at least 90% sequence identity, such as at least 91% sequence identity, or at least 92% sequence identity, or at least 93% sequence identity, or at least 94% sequence identity, or at least 95% sequence identity, or at least 96% sequence identity, or at least 97% sequence identity, or at least 98% sequence identity, and including at least 99% sequence identity, to the corresponding native human or non-human primate protein wherein sequence identity is as defined herein. Preferably, a variant possesses at least one biological property in common with the human or non-human protein.
"Variant of human or non-human primate nucleic acid molecule or sequence" refers to a nucleic acid sequence that possesses at least 90% sequence identity, such as at least 91%, or at least 92%, or at least 93%, or at least 94%, or at least 95%, or at least 96%, or at least 97%, or at least 98% sequence identity, and including at least 99% sequence identity, to the corresponding native human or non-human primate nucleic acid sequence, wherein "sequence identity" is as defined herein.
"Fragment of human or non-human primate nucleic acid molecule or sequence" refers to a nucleic acid sequence corresponding to a portion ofthe native human nucleic acid sequence discussed herein in the examples or a primate native non-human homolog molecule, wherein said portion is at least about 50 nucleotides in length, or 100, for example, at least 200 or 300 nucleotides in length.
"Antigenic fragments of colon or colorectal" refer to polypeptides corresponding to a fragmentt o < f colon antigen encoded by any ofthe genes disclosed herein or a variant or homologue thereof that when used itself or attached to an immunogenic carrier that elicits antibodies that specifically bind the protein. Typically, antigenic fragments are at least 20 amino acids in length.
Sequence identity or percent identity is intended to mean the percentage ofthe same residues shared between two sequences, referenced to the human DNA or amino acid sequences disclosed herein, when the two sequences are aligned using the Clustal method [Higgins et al, Cabios 8:189-191 (1992)] of multiple sequence alignment in the Lasergene biocomputing software (DNASTAR, INC. of Madison, Wisconsin). In this method, multiple alignments are carried out in a progressive manner, in which larger and larger alignment groups are assembled using similarity scores calculated from a series of pairwise alignments. Optimal sequence alignments are obtained by finding the maximum alignment score, which is the average of all scores between the separate residues in the alignment, determined from a residue weight table representing the probability of a given amino acid change occurring in two related proteins over a given evolutionary interval. Penalties for opening and lengthening gaps in the alignment contribute to the score. The default parameters used with this program are as follows: gap penalty for multiple alignment=10; gap length penalty for multiple alignment=10; k-tuple value in pairwise alignments ; gap penalty in pairwise alignment=3; window value in pairwise alignment=5; diagonals saved in pairwise alignment=5. The residue weight table used for the alignment program is PAM25O [Dayhoffet al., in Atlas of Protein Sequence and Stracture, Dayhoff, Ed., NDRF, Washington, Vol. 5, suppl. 3, p. 345, (1978)].
Percent conservation is calculated from the above alignment by adding the percentage of identical residues to the percentage of positions at which the two residues represent a conservative substitution (defined as having a log odds value of greater than or equal to 0.3 in the PAM250 residue weight table). Conservation is referenced to a human gene ofthe invention when determining percent conservation with a non-human gene and when determining percent conservation. Conservative amino acid changes satisfying this requirement include: R-K; E-D, Y-F, L-M; V-I, Q-H.
Polypeptide Fragments
The invention provides polypeptide fragments ofthe disclosed proteins. Polypeptide fragments ofthe invention can comprise at least 8 amino acid residues, such as at least 25 or at least 50 amino acid residues of human or non-human primate gene according to the invention or an analogue thereof. Polypeptide fragments can also comprise at least 75, 100, 125, 150, 175, 200, 225, 250, or 275 residues ofthe polypeptide encoded by gene the subject genes which are specifically expressed by certain human colon or colorectal as well as some other tumor tissues. In one embodiment ofthe invention, a protein fragment can also comprise a majority ofthe native protein colon or colorectal protein, i.e. at least about 100 contiguous residues ofthe native colon or colorectal protein antigen.
Biologically Active Variants The invention also encompasses biologically active mutants of protein colon or colorectal proteins according to the invention, which comprise an amino acid sequence that is at least 80%, for example, 90% or 95-99% similar to the subject tumor-associated proteins. Guidance in determining which amino acid residues can be substituted, inserted, or deleted without abolishing biological or immunological activity can be found using computer programs well known in the art, such as DNASTAR software. Protein variants can include conoservative amino acid changes, i.e., substitutions of similarly charged or uncharged amino acids. A conservative amino acid change involves substitution of one of a family of amino acids which are related in their side chains. Naturally occurring amino acids are generally divided into four families: acidic (aspartate, glutamate), basic (lysine, arginine, histidine), non-polar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), and uncharged polar (glycine, asparagine, glutamine, cystine, serine, threonine, tyrosine) amino acids. Phenylalanine, tryptophan, and tyrosine are sometimes classified jointly as aromatic amino acids.
A subset of mutants, called muteins, is a group of polypeptides in which neutral amino acids, such as serines, are substituted for cysteine residues which do not participate in disulfide bonds. These mutants may be stable over a broader temperature range than native secreted proteins. See Mark et al, U.S. Patent 4,959,314.
It is reasonable to expect that an isolated replacement of a leucine with an isoleucine or valine, an aspartate with a glutamate, a threonine with a serine, or a similar replacement of an amino acid with a structurally related amino acid can be made without affecting the biological properties ofthe resulting secreted protein or polypeptide variant.
Human or non-human primate protein variants include glycosylated forms, aggregative conjugates with other molecules, and covalent conjugates with unrelated chemical moieties. Also, protein variants also include allelic variants, species variants, and muteins. Truncations or deletions of regions which do not affect the differential expression ofthe protein gene are also variants. Covalent variants can be prepared by linking functionalities to groups which are found in the amino acid chain or at the N- or C-terminal residue, as is known in the art.
Some amino acid sequence ofthe proteins ofthe invention can be varied without significant effect on the stracture or function ofthe protein. If such differences in sequence are contemplated, it should be remembered that there are critical areas on the protein which determine activity. In general, it is possible to replace residues that form the tertiary stracture, provided that residues performing a similar function are used. Numerous substitutions at non-critical regions ofthe protein are well tolerated. The replacement of amino acids can also change the selectivity of binding to cell surface receptors. Ostade et al., Nature 361:266-268 (1993) describes certain mutations resulting in selective binding of TNF- alpha to only one ofthe two known types of TNF receptors. Thus, the polypeptides ofthe present invention can include one or more amino acid substitutions, deletions or additions, either from natural mutations or human manipulation.
The invention further includes variations ofthe protein subject colon or colorectal which show comparable expression patterns or which include antigenic regions. Protein mutants include deletions, insertions, inversions, repeats, and type substitutions. Guidance concerning which amino acid changes are likely to be phenotypically silent can be found in Bowie, J.U., et al., "Deciphering the Message in Protein Sequences: Tolerance to Amino Acid Substitutions," Science 247:1306-1310 (1990).
For example, charged amino acids can be substituted with another charged amino acid, or with neutral or negatively charged amino acids. The latter results in proteins with reduced positive charge to improve the characteristics ofthe disclosed protein. The prevention of aggregation is highly desirable. Aggregation of proteins not only results in a loss of activity but can also be problematic when preparing pharmaceutical formulations, because they can be immunogenic. (Pinckard et al., Clin. Exp. Immunol. 2:331-340 (1967); Robbins et al., Diabetes 36:838-845 (1987); Cleland et al., Crit. Rev. Therapeutic Drug Carrier Systems 10:307-377 (1993)).
Amino acids in the polypeptides ofthe present invention that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine- scanning mutagenesis (Cunningham and Wells, Science 244: 1081-1085 (1989)). The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity such as binding to a natural or synthetic binding partner. Sites that are critical for ligand-receptor binding can also be determined by structural analysis such as crystallization, nuclear magnetic resonance or photoaffinity labeling (Smith et al., JMol. Biol. 224:899-904 (1992) and de Vos et al. Science 255: 306-312 (1992)).
Conservative amino acid substitutions often do not significantly affect the folding or activity ofthe protein. A skilled artisan could determine an appropriate number and nature of amino acid substitutions based on factors as described above. Generally speaking, the number of substitutions for any given polypeptide are fewer than 50, 40, 30, 25, 20, 15, 10, 5 or 3 residues.
Fusion Proteins
Fusion proteins comprising proteins or polypeptide fragments ofthe subject colon or colorectal proteins can also be constructed. Fusion proteins are useful for generating antibodies against amino acid sequences and for use in various assay systems. For example, fusion proteins can be used to identify proteins which interact with a protein ofthe invention or which interfere with its biological function. Physical methods, such as protein affinity chromatography, or library-based assays for protein-protein interactions, such as the yeast two-hybrid or phage display systems, can also be used for this purpose. The foregoing can also be adapted as a screening technique. Fusion proteins comprising a signal sequence and or a transmembrane domain of a protein according to the invention or a fragment thereof can be used to target other protein domains to cellular locations in which the domains are not normally found, such as bound to a cellular membrane or secreted extracellularly.
A fusion protein comprises two protein segments fused together by means of a peptide bond. Amino acid sequences for use in fusion proteins ofthe invention can utilize any ofthe amino acid sequences or encoded by the nucleotide sequences disclosed herein, or can be prepared from biologically active variants or fragment of said protein sequence, such as those described above. The first protein segment can consist of a full-length protein or a variant or fragment thereof. These fragments can range in size from about 8 amino acids up to the full length ofthe protein.
The second protein segment can be a full-length protein or a polypeptide fragment. Proteins commonly used in fusion protein construction include β-galactosidase, β- glucuronidase, green fluorescent protein (GFP), autofluorescent proteins, including blue fluorescent protein (BFP), glutathione-S-transferase (GST), luciferase, horseradish peroxidase (HRP), and chloramphenicol acetyltransferase (CAT). Additionally, epitope tags can be used in fusion protein constructions, including histidine (His) tags, FLAG tags, influenza hemagglutinin (HA) tags, Myc tags, VSV-G tags, and thioredoxin (Trx) tags. Other fusion constructions can include maltose binding protein (MBP), S-tag, Lex a DNA binding domain (DBD) fusions, GAL4 DNA binding domain fusions, and herpes simplex virus (HSV) BP16 protein fusions.
These fusions can be made, for example, by covalently linking two protein segments or by standard procedures in the art of molecular biology. Recombinant DNA methods can be used to prepare fusion proteins, for example, by making a DNA construct which comprises a coding sequence encoding an amino acid sequence according to the invention in proper reading frame with a nucleotide encoding the second protein segment and expressing the DNA construct in a host cell, as is known in the art. Many kits for constructing fusion proteins are available from companies that supply research labs with tools for experiments, including, for example, Promega Corporation (Madison, Wl), Stratagene (La Jolla, CA), Clontech (Mountain View, CA), Santa Cruz Biotechnology (Santa Cruz, CA), MBL
International Corporation (MIC; Watertown, MA), and Quantum Biotechnologies (Montreal, Canada; 1-888-DNA-KITS).
Proteins, fusion proteins, or polypeptides ofthe invention can be produced by recombinant DNA methods. For production of recombinant proteins, fusion proteins, or polypeptides, a sequence listing encoding one ofthe subject colon or colorectal proteins can be expressed in prokaryotic or eukaryotic host cells using expression systems known in the art. These expression systems include bacterial, yeast, insect, and mammalian cells.
The resulting expressed protein can then be purified from the culture medium or from extracts ofthe cultured cells using purification procedures known in the art. For example, for proteins fully secreted into the culture medium, cell-free medium can be diluted with sodium acetate and contacted with a cation exchange resin, followed by hydrophobic interaction chromatography. Using this method, the desired protein or polypeptide is typically greater than 95% pure. Further purification can be undertaken, using, for example, any ofthe techniques listed above.
Proteins can be further modified, for example by phosphorylation or glycosylation of the appropriate sites, in order to obtain a functional protein. Covalent attachments can be made using known chemical or enzymatic methods.
Human or non-human primate proteins according to the invention or polypeptide of the invention can also be expressed in cultured host cells in a form that facilitates purification. For example, a protein or polypeptide can be expressed as a fusion protein comprising, for example, maltose binding protein, glutathione-S-transferase, or thioredoxin, and purified using a commercially available kit. Kits for expression and purification of such fusion proteins are available from companies such as New England BioLabs, Pharmacia, and Invifrogen. Proteins, fusion proteins, or polypeptides can also be tagged with an epitope, such as a "Flag" epitope (Kodak), and purified using an antibody which specifically binds to that epitope. The coding sequence disclosed herein can also be used to construct transgenic animals, such as mice, rats, guinea pigs, cows, goats, pigs, or sheep. Female transgenic animals can then produce proteins, polypeptides, or fusion proteins ofthe invention in their milk. Methods for constructing such animals are known and widely used in the art.
Alternatively, synthetic chemical methods, such as solid phase peptide synthesis, can be used to synthesize a secreted protein or polypeptide. General means for the production of peptides, analogs or derivatives are outlined in Chemistry and Biochemistry of Amino Acids, Peptides, and Proteins - A Survey of Recent Developments, B. Weinstein, ed. (1983). Substitution of D-amino acids for the normal L-stereoisomer can be carried out to increase the half-life ofthe molecule. Typically, homologous polynucleotide sequences can be confirmed by hybridization under stringent conditions, as is known in the art. For example, using the following wash conditions: 2X SSC (0.3 M NaCl, 0.03 M sodium citrate, pH 7.0), 0.1% SDS, room temperature twice, 30 minutes each; then 2X SSC, 0.1% SDS, 50°C once, 30 minutes; then 2X SSC, room temperature twice, 10 minutes each, homologous sequences can be identified which contain at most about 25-30% base pair mismatches. Homologous nucleic acids can contain 15-25% base pair mismatches or fewer, for example about 5-15% base pair mismatches. The invention also provides polynucleotide probes which can be used to detect complementary nucleotide sequences, for example, in hybridization protocols such as Northern or Southern blotting or in situ hybridizations. Polynucleotide probes ofthe invention comprise at least 12, 13, 14, 15, 16, 17, 18, 19, 20, 30, or 40 or more contiguous nucleotides ofthe gene A and gene B nucleic acid sequences provided herein. Polynucleotide probes ofthe invention can comprise a detectable label, such as a radioisotopic, fluorescent, enzymatic, or chemiluminescent label.
Isolated genes corresponding to the cDNA sequences disclosed herein are also provided. Standard molecular biology methods can be used to isolate the corresponding genes using the cDNA sequences provided herein. These methods include preparation of probes or primers based on the disclosed sequences for use in identifying or amplifying the genes from mammalian, including human, genomic libraries or other sources of human genomic DNA.
Polynucleotide molecules ofthe invention can also be used as primers to obtain additional copies of the polynucleotides, using polynucleotide amplification methods.
Polynucleotide molecules can be propagated in vectors and cell lines using techniques well known in the art. Polynucleotide molecules can be on linear or circular molecules. They can be on autonomously replicating molecules or on molecules without replication sequences. They can be regulated by their own or by other regulatory sequences, as is known in the art.
Polynucleotide Constructs
Polynucleotide molecules comprising the coding sequences disclosed herein can be used in a polynucleotide construct, such as a DNA or RNA construct. Polynucleotide molecules ofthe invention can be used, for example, in an expression construct to express all or a portion of a protein, variant, fusion protein, or single-chain antibody in a host cell. An expression construct comprises a promoter which is functional in a chosen host cell. The skilled artisan can readily select an appropriate promoter from the large number of cell type- specific promoters known and used in the art. The expression construct can also contain a transcription terminator which is functional in the host cell. The expression construct comprises a polynucleotide segment which encodes all or a portion ofthe desired protein. The polynucleotide segment is located downstream from the promoter. Transcription ofthe polynucleotide segment initiates at the promoter. The expression construct can be linear or circular and can contain sequences, if desired, for autonomous replication.
Also included are polynucleotide molecules comprising human or non-human primate gene promoter and UTR sequences, operably linked to either protein coding sequences or other sequences encoding a detectable or selectable marker. Promoter and/or UTR-based constructs are useful for studying the transcriptional and translational regulation of protein expression, and for identifying activating and/or inhibitory regulatory proteins.
Host Cells
An expression construct can be introduced into a host cell. The host cell comprising the expression construct can be any suitable prokaryotic or eukaryotic cell. Expression systems in bacteria include those described in Chang et al, Nature 275:615 (1978); Goeddel et al, Nature 281: 544 (1979); Goeddel et al, Nucleic Acids Res. 8:4057 (1980); EP 36,776;
U.S. 4,551,433; deBoer et al, Proc. Natl Acad Sci. USA 80: 21-25 (1983); and Siebenlist et al. Cell 20: 269 (1980). Expression systems in yeast include those described in Hinnnen et al, Proc. Natl.
Acad. Sci. USA 75: 1929 (1978); Ito et al, JBacteήol 153: 163 (1983); Kurtz et al, Mol.
Cell. Biol. 6: 142 (1986); Kunze et al, J Basic Microbiol. 25: 141 (1985); Gleeson et al, J.
Gen. Microbiol. 132: 3459 (1986), Roggenkamp et al, Mol Gen. Genet. 202: 302 (1986));
Das et al, J Bacteriol. 158: 1165 (1984); De Louvencourt et al, JBacteriol 154:737 (1983), Van den Berg et al, Bio/Technology 8: 135 (1990); Kunze et al., J. Basic Microbiol 25: 141
(1985); Cregg et al, Mol Cell Biol 5: 3376 (1985); U.S. 4,837,148; U.S. 4,929,555; Beach and Nurse, Nature 300: 706 (1981); Davidow et al, Curr. Genet. 10: 380 (1985); Gaillardin et al, Curr. Genet. 10: 49 (1985); Ballance et al, Biochem. Biophys. Res. Commun. 112: 284-
289 (1983); Tilburn et al, Gene 26: 205-22 (1983); Yelton et al, Proc. Natl. Acad, Sci. USA 81 : 1470-1474 (1984); Kelly and Hynes, EMBO J. 4: 475479 (1985); EP 244,234; and WO
91/00357.
Expression of heterologous genes in insects can be accomplished as described in U.S.
4,745,051; Friesen et al (1986) "The Regulation of Baculoviras Gene Expression" in: THE
MOLECULAR BIOLOGY OF BACULOVIRUSES (W. Doerfler, ed.); EP 127,839; EP 155,476; Vlak et al, J. Gen. Virol 69: 765-776 (1988); Miller et al, Ann. Rev. Microbiol. 42:
177 (1988); Carbonell et al, Gene 73: 409 (1988); Maeda et al, Nature 315: 592-594 (1985);
Lebacq-Verheyden et al, Mol. Cell Biol 8: 3129 (1988); Smith et al, Proc. Natl. Acad. Sci. USA 82: 8404 (1985); Miyajima et al, Gene 58: 273 (1987); and Martin et al, DNA 7:99 (1988). Numerous baculoviral strains and variants and corresponding permissive insect host cells from hosts are described in Luckow et al, Bio/Technology (1988) 6: 47-55, Miller et al, in GENETIC ENGINEERING (Setlow, J.K. et al. eds.), Vol. 8, pp. 277-279 (Plenum Publishing, 1986); and Maeda et al, Nature, 315: 592-594 (1985).
Mammalian expression can be accomplished as described in Dijkema et al, EMBO J. 4: 761(1985); Gormanetal, Proc. Natl. Acad. Sci. USA 79: 6777 (1982b); Boshart et al, Cell 41: 521 (1985); and U.S. 4,399,216. Other features of mammalian expression can be facilitated as described in Ham and Wallace, Meth Enz. 58: 44 (1979); Barnes and Sato, Anal. Biochem. 102: 255 (1980); U.S. 4,767,704; U.S. 4,657,866; U.S. 4,927,762; U.S. 4,560,655; WO 90/103430, WO 87/00195, and U.S. RE 30,985.
Expression constructs can be introduced into host cells using any technique known in the art. These techniques include transferrin-polycation-mediated DNA transfer, transfection with naked or encapsulated nucleic acids, liposome-mediated cellular fusion, intracellular transportation of DNA-coated latex beads, protoplast fusion, viral infection, electroporation, "gene gun," and calcium phosphate-mediated transfection.
Expression of an endogenous gene encoding a protein ofthe invention can also be manipulated by introducing by homologous recombination a DNA construct comprising a transcription unit in frame with the endogenous gene, to form a homologously recombinant cell comprising the transcription unit. The transcription unit comprises a targeting sequence, a regulatory sequence, an exon, and an unpaired splice donor site. The new transcription unit can be used to turn the endogenous gene on or off as desired. This method of affecting endogenous gene expression is taught in U.S. Patent 5,641,670.
The targeting sequence is a segment of at least 10, 12, 15, 20, or 50 contiguous nucleotides ofthe nucleotide sequences disclosed herein. The transcription unit is located upstream to a coding sequence ofthe endogenous gene. The exogenous regulatory sequence directs transcription ofthe coding sequence ofthe endogenous gene.
Human or non-human primate protein can also include hybrid and modified forms thereof including fusion proteins, fragments and hybrid and modified forms in which certain amino acids have been deleted or replaced, modifications such as where one or more amino acids have been changed to a modified amino acid or unusual amino acid.
Also included within the meaning of substantially homologous is any human or non- human primate protein which shows cross-reactivity with antibodies to a gene described herein or whose encoding nucleotide sequences including genomic DNA, mRNA or cDNA are isolated through hybridization with the complementary sequence of genomic or subgenomic nucleotide sequences or cDNA of a gene disclosed herein or a fragment thereof. Degenerate DNA sequences that encode human or non-human primate proteins are also included within the present invention as are allelic variants of.
Colon or colorectal proteins ofthe invention can be prepared using recombinant DNA techniques. By "pure form" or "purified form" or "substantially purified form" it is meant that a protein composition is substantially free of other proteins which are not protein. The present invention also includes therapeutic or pharmaceutical compositions comprising human or non-human primate proteins, fragments or variants according to the invention in an effective amount for treating patients with disease, and a method comprising administering a therapeutically effective amount of a protein according to the invention. These compositions and methods are useful for treating cancers associated with a protein according to the invention, e.g. colon cancer. One skilled in the art can readily use a variety of assays known in the art to determine whether a protein according to the invention would be useful in promoting survival or functioning in a particular cell type.
In certain circumstances, it may be desirable to modulate or decrease the amount of the subject colon or colorectal protein expressed. Thus, in another aspect ofthe present invention, anti-sense oligonucleotides can be made specific to genes disclosed herein and a method utilized for diminishing the level of expression a protein according to the invention by a cell comprising administering one or more gene anti-sense oligonucleotides. By gene specific anti-sense oligonucleotides reference is made to oligonucleotides that have a nucleotide sequence that interacts through base pairing with a specific complementary nucleic acid sequence involved in the expression of a gene according to the invention that the expression ofthe gene is reduced. Nucleic acids involved in the expression ofthe subject gene include genomic DNA and mRNA that encode a colon or colorectal gene disclosed herein. This genomic DNA molecule can comprise regulatory regions ofthe gene, or the coding sequence for mature gene encoded by the gene. The term complementary to a nucleotide sequence in the context of antisense oligonucleotides and methods therefor means sufficiently complementary to such a sequence as to allow hybridization to that sequence in a cell, i.e., under physiological conditions. The antisense oligonucleotides can comprise a sequence containing from about 8 to about 100 nucleotides, including antisense oligonucleotides that comprise from about 15 to about 30 nucleotides. The antisense oligonucleotides can also contain a variety of modifications that confer resistance to nucleolytic degradation such as, for example, modified intemucleoside linages [Uhlmann and Peyman, Chemical Reviews 90:543-548 (1990); Schneider and Banner, Tetrahedron Lett. 31:335, (1990) which are incorporated by reference], modified nucleic acid bases as disclosed in 5,958,773 and patents disclosed therein, and/or sugars and the like.
Any modifications or variations ofthe antisense molecule which are known in the art to be broadly applicable to antisense technology are included within the scope ofthe invention. Representative modifications include preparation of phosphorus-containing linkages as disclosed in U.S. Patents 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361, 5,625,050 and 5,958,773.
The antisense compounds ofthe invention can include modified bases. The antisense oligonucleotides ofthe invention can also be modified by chemically linking the oligonucleotide to one or more moieties or conjugates to enhance the activity, cellular distribution, or cellular uptake ofthe antisense oligonucleotide. Representative moieties or conjugates include lipids such as cholesterol, cholic acid, thioether, aliphatic chains, phospholipids, polyamines, polyethylene glycol (PEG), palmityl moieties, and others as disclosed in, for example, U.S. Patents 5,514,758, 5,565,552, 5,567,810, 5,574,142, 5,585,481, 5,587,371, 5,597,696 and 5,958,773.
Chimeric antisense oligonucleotides are also within the scope ofthe invention, and can be prepared from the present inventive oligonucleotides using the methods described in, for example, U.S. Patents 5,013,830, 5,149,797, 5,403,711, 5,491,133, 5,565,350, 5,652,355, 5,700,922 and 5,958,773. Select of optimal antisense molecules for particular targets typically involves routine screening of a number of candidate molecules. An antisense molecule can be targeted to an accessible, or exposed, portion ofthe target RNA molecule. Although in some cases information is available about the stracture of target mRNA molecules, the current approach to inhibition using antisense is via experimentation. mRNA levels in the cell can be measured routinely in treated and control cells by reverse transcription ofthe mRNA and assaying the cDNA levels. The biological effect can be determined routinely by measuring cell growth or viability as is known in the art. Measuring the specificity of antisense activity by assaying and analyzing cDNA levels is an art-recognized method of validating antisense results. It has been suggested that RNA from treated and control cells should be reverse-transcribed and the resulting cDNA populations analyzed. [Branch, A. D., T.I.B.S. 23:45-50 (1998)]. The therapeutic or pharmaceutical compositions ofthe present invention can be administered by any suitable route known in the art including for example intravenous, subcutaneous, intramuscular, transdermal, intrathecal or infracerebral. Administration can be either rapid as by injection or over a period of time as by slow infusion or administration of slow release formulation. Additionally, a human or non-human primate protein according to the invention can also be linked or conjugated with agents that provide desirable pharmaceutical or pharmacodynamic properties. For example, the protein can be coupled to any substance known in the art to promote penetration or transport across the blood-brain barrier such as an antibody to the transferrin receptor, and administered by intravenous injection (see, for example, Friden et al., Science 259:373-377 (1993) which is incorporated by reference). Furthermore, the subject protein can be stably linked to a polymer such as polyethylene glycol to obtain desirable properties of solubility, stability, half-life and other pharmaceutically advantageous properties. [See, for example, Davis et al., Enzyme Eng. 4:169-73 (1978); Buruham, Λm. J. Hosp. Pharm. 51:210-218 (1994) which are incorporated by reference].
The compositions are usually employed in the form of pharmaceutical preparations, which are made in a manner well known in the pharmaceutical art. See, e.g. Remington Pharmaceutical Science, 18th Ed., Merck Publishing Co. Eastern PA, (1990). Physiological saline solutions can be used, as well as other pharmaceutically acceptable carriers such as physiological concentrations of other non-toxic salts, five percent aqueous glucose solution, sterile water and the like. Compositions ofthe invention can also include a suitable buffer. Optionally, such solutions can be lyophilized and stored in a sterile ampoule ready for reconstitution by the addition of sterile water for ready injection. The primary solvent can be aqueous or alternatively non-aqueous. The subject human or primate protein, fragment or variant thereof can also be incorporated into a solid or semi-solid biologically compatible matrix which can be implanted into tissues requiring treatment.
The carrier can also contain other pharmaceutically-acceptable excipients for modifying or maintaining the pH, osmolarity, viscosity, clarity, color, sterility, stability, rate of dissolution, or odor ofthe formulation. Similarly, the carrier can contain still other pharmaceutically-acceptable excipients for modifying or maintaining release or absorption or penetration across the blood-brain barrier. Excipients are those substances usually and customarily employed to formulate dosages for parenteral administration in either unit dosage or multi-dose form or for direct infusion into the cerebrospinal fluid by continuous or periodic infusion.
Dose administration can be repeated depending upon the pharmacokinetic parameters ofthe dosage formulation and the route of administration used. It is also contemplated that certain formulations containing a protein according to the invention or variant or fragment thereof are to be administered orally. Protein formulations can be encapsulated and formulated with suitable carriers in solid dosage forms. Some examples of suitable carriers, excipients, and diluents include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, calcium silicate, microcrystalline cellulose, polyvinylpyrrohdone, cellulose, gelatin, syrap, methyl cellulose, methyl- and propylhydroxybenzoates, talc, magnesium, stearate, water, mineral oil, and the like. The formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents or flavoring agents. The compositions can be formulated so as to provide rapid, sustained, or delayed release of the active ingredients after administration to the patient by employing procedures well known in the art. The formulations can also contain substances that diminish proteolytic degradation and promote absorption such as, for example, surface active agents.
The specific dose is calculated according to the approximate body weight or body surface area ofthe patient or the volume of body space to be occupied. The dose also depends on the particular route of admimsfration selected. Further refinement of the calculations necessary to determine the appropriate dosage for treatment is routinely made by those of ordinary skill in the art. Following a review ofthe present disclosure, an effective dosage can be determined without undue experimentation. Exact dosages are determined in conjunction with standard dose-response studies. The amount ofthe composition actually administered can be determined by a practitioner, in the light ofthe relevant circumstances including the condition or conditions to be treated, the choice of composition to be administered, the age, weight, and response ofthe individual patient, the severity ofthe patient's symptoms, and the chosen route of administration.
In one embodiment, a protein ofthe present invention is therapeutically administered by implanting into patients vectors or cells capable of producing a biologically-active form of the protein or a precursor ofthe protein, i.e., a molecule that can be readily converted to a biological-active form ofthe by the body. For example, cells that secrete the protein can be encapsulated into semipermeable membranes for implantation into a patient. The cells can be cells that normally express the protein or a precursor thereof or the cells can be transformed to express the protein or a precursor thereof. For human subjects, a human protein can be used, or a non-human primate protein homolog of a human protein can be used. In a number of circumstances it would be desirable to determine the levels of protein or corresponding mRNA encoding a protein according to the invention in a patient. The identification ofthe subject genes which are specifically expressed by colon or colorectal tumors suggests these proteins are expressed at different levels during some diseases, e.g., cancers, provides the basis for the conclusion that the presence of these proteins serves a normal physiological function related to cell growth and survival. Endogenously produced human colon or colorectal antigen according to the invention may also play a role in certain disease conditions.
The term "detection" as used herein in the context of detecting the presence of a cancer gene according to the invention in a patient is intended to include the determining of the amount of protein according to the invention or the ability to express an amount of this protein in a patient, the estimation of prognosis in terms of probable outcome of a disease and prospect for recovery, the monitoring of these protein levels over a period of time as a measure of status ofthe condition, and the monitoring of colon or colorectal protein according to the invention for determining an effective therapeutic regimen for the patient, e.g. one with colon cancer.
To detect the presence of a gene according to the invention in a patient, a sample is obtained from the patient. The sample can be a tissue biopsy sample or a sample of blood, plasma, serum, CSF or the like. It has been found that the subject genes are expressed at high levels in some cancers, e.g., colon or colorectal cancers. Samples for detecting protein can be taken from these tissue. When assessing peripheral levels of protein, a sample of blood, plasma or serum can be used. When assessing the levels of protein in the central nervous system, samples can be obtained from cerebrospinal fluid or neural tissue. In some instances, it is desirable to determine whether a gene according to the invention is intact in the patient or in a tissue or cell line within the patient. By an intact gene, it is meant that there are no alterations in the gene such as point mutations, deletions, insertions, chromosomal breakage, chromosomal rearrangements and the like wherein such alteration might alter the production of gene or alter its biological activity, stability or the like to lead to disease processes. Thus, in one embodiment ofthe present invention a method is provided for detecting and characterizing any alterations in the gene. The method comprises providing an oligonucleotide that contains the gene corresponding cDNA, genomic DNA or a fragment thereof or a derivative thereof. By a derivative of an oligonucleotide, it is meant that the derived oligonucleotide is substantially the same as the sequence from which it is derived in that the derived sequence has sufficient sequence complementarily to the sequence from which it is derived to hybridize specifically to the gene. A nucleic acid ofthe invention can be isolated, chemically synthesized, of recombinantly produced (e.g., using in vitro DNA replication, reverse transcription, or transcription). Typically, patient genomic DNA is isolated from a cell sample from the patient and digested with one or more restriction endonucleases such as, for example, Taql and Alul. Using the Southern blot protocol, which is well known in the art, this assay determines whether a patient or a particular tissue in a patient has an intact gene according to the invention or a gene abnormality. Hybridization to a gene according to the invention would involve denaturing the chromosomal DNA to obtain a single-stranded DNA; contacting the single-stranded DNA with a gene probe associated with the gene sequence; and identifying the hybridized DNA- probe to detect chromosomal DNA containing at least a portion of a human gene according to the invention. The term "probe" as used herein refers to a stracture comprised of a polynucleotide that forms a hybrid stracture with a target sequence, due to complementarity of probe sequence with a sequence in the target region. Oligomers suitable for use as probes typically contain at least about 8-12 contiguous nucleotides which are complementary to the targeted sequence, for example 20 nucleotides. Probes ofthe present invention can be DNA or RNA oligonucleotides and can be made by any method known in the art such as, for example, excision, transcription or chemical synthesis. Probes can be labeled with any detectable label known in the art such as, for example, radioactive or fluorescent labels or enzymatic marker. Labeling ofthe probe can be accomplished by any method known in the art such as by PCR, random priming, end labeling, nick translation or the like. Methods that do not employ a labeled probe can also be used to determine the hybridization. Representative techniques include Southern blotting, fluorescence in situ hybridization, and single-strand conformation polymoφhism with PCR amplification.
Hybridization is typically carried out at about 25° - 45° C, or at about 32° -40° C, or at about 37° - 38° C. Hybridization can proceed for about 0.25 hour to about 96 hours, or from about 1 (one) hour to about 72 hours, or from about 4 hours to about 24 hours. Gene abnormalities can also be detected by using the PCR method and primers that flank or lie within the particular gene. The PCR method is well known in the art. Briefly, this method is performed using two oligonucleotide primers which are capable of hybridizing to the nucleic acid sequences flanking a target sequence that lies within gene and amplifying the target sequence. The terms "oligonucleotide primer" as used herein refers to a short strand of DNA or RNA ranging in length from about 8 to about 30 bases. The upstream and downstream primers are typically from about 20 to about 30 base pairs in length and hybridize to the flanking regions for replication ofthe nucleotide sequence. The polymerization is catalyzed by a DNA-polymerase in the presence of deoxynucleotide triphosphates or nucleotide analogs to produce double-stranded DNA molecules. The double strands are then separated by any denaturing method including physical, chemical or enzymatic. Commonly, a method of physical denaturation is used involving heating the nucleic acid, typically to temperatures from about 80°C to 105°C for times ranging from about 1 to about 10 minutes. The process is repeated for the desired number of cycles. The primers are selected to be substantially complementary to the strand of DNA being amplified. Therefore, the primers need not reflect the exact sequence ofthe template, but must be sufficiently complementary to selectively hybridize with the sfrand being amplified.
After PCR amplification, the DNA sequence comprising a gene ofthe invention or a fragment thereof is then directly sequenced and analyzed by comparison ofthe sequence with the sequences disclosed herein to identify alterations which might change activity or expression levels or the like.
In another embodiment, a method for detecting protein a colon according to the invention is provided based upon an analysis of tissue expressing the gene. Certain tissues such as breast, lung, colon and others can be analyzed. The method comprises hybridizing a polynucleotide to mRNA from a sample of tissue that normally expresses the gene. The sample is obtained from a patient suspected of having an abnormality in the gene. To detect the presence of mRNA encoding protein a colon or colorectal protein according to the invention is obtained from a patient. The sample can be from blood or from a tissue biopsy sample. The sample can be treated to extract the nucleic acids contained therein. The resulting nucleic acid from the sample is subjected to gel electrophoresis or other size separation techniques. The mRNA ofthe sample is contacted with a DNA sequence serving as a probe to form hybrid duplexes. The use of a labeled probes as discussed above allows detection ofthe resulting duplex.
When using the cDNA encoding a colon or colorectal protein according to the invention or a derivative ofthe cDNA as a probe, high stringency conditions can be used in order to prevent false positives, that is the hybridization and apparent detection ofthe gene nucleotide sequences when in fact an intact and functioning gene is not present. When using sequences derived from the gene or cDNA, less stringent conditions could be used, however, are less preferred because ofthe likelihood of false positives. The stringency of hybridization is determined by a number of factors during hybridization and during the washing procedure, including temperature, ionic strength, length of time and concentration of formamide. These factors are outlined in, for example, Sambrook et al. [Sambrook et al. (1989), supra].
In order to increase the sensitivity ofthe detection in a sample of mRNA encoding the protein, the technique of reverse transcription polymerization chain reaction (RT/PCR) can be used to amplify cDNA transcribed from mRNA encoding the protein. The method of RT/PCR is well known in the art, and can be performed as follows. Total cellular RNA is isolated by, for example, the standard guanidium isothiocyanate method and the total RNA is reverse transcribed. The reverse transcription method involves synthesis of DNA on a template of RNA using a reverse transcriptase enzyme and a 3' end primer. Typically, the primer contains an oligo(dT) sequence. The cDNA thus produced is then amplified using the PCR method and specific primers. [Belyavsky et al., Nwc/. Acid Res. 17:2919-2932 (1989); Krug and Berger, Methods in Enzymology, 152:316-325, Academic Press, ΝY (1987) which are incoφorated by reference]. The polymerase chain reaction method is performed as described above using two oligonucleotide primers that are substantially complementary to the two flanking regions of the DNA segment to be amplified. Following amplification, the PCR product is then electrophoresed and detected by ethidium bromide staining or by phosphoimaging. The present invention further provides for methods to detect the presence of a colon or colorectal protein in a sample obtained from a patient. Any method known in the art for detecting proteins can be used. Representative methods include, but are not limited to immunodiffusion, immunoelectrophoresis, immunochemical methods, binder-ligand assays, immunohistochemical techniques, agglutination and complement assays. [Basic and Clinical Immunology, 217-262, Sites and Terr, eds., Appleton & Lange, Norwalk, CT, (1991), which is incoφorated by reference]. For example, binder-ligand immunoassays can be used, which involve reacting antibodies with an epitope or epitopes of a colon protein ofthe invention and competitively displacing a labeled protein or derivative thereof.
As used herein, a derivative of a protein according to the invention is intended to include a polypeptide in which certain amino acids have been deleted or replaced or changed to modified or unusual amino acids wherein the derivative is biologically equivalent to the gene and wherein the polypeptide derivative cross-reacts with antibodies raised against the protein. By cross-reaction it is meant that an antibody reacts with an antigen other than the one that induced its formation. Numerous competitive and non-competitive protein-binding immunoassays are well known in the art. Antibodies employed in such assays can be unlabeled, for example as used in agglutination tests, or labeled for use in a wide variety of assay methods. Labels that can be used include radionuclides, enzymes, fluorescers, chemiluminescers, enzyme substrates or co-factors, enzyme inhibitors, particles, dyes and the like for use in radioinununoassay (RIA), enzyme immunoassays, e.g., enzyme-linked immunosorbent assay (ELISA), fluorescent immunoassays and the like.
Polyclonal or monoclonal antibodies to the subject non-human primate or human proteins or according to the invention an epitope thereof can be made for use in immunoassays by any of a number of methods known in the art. By epitope reference is made to an antigenic determinant of a polypeptide. An epitope could comprise 3 amino acids in a spatial conformation which is unique to the epitope. Generally an epitope consists of at least 5 such amino acids. Methods of determining the spatial conformation of amino acids are known in the art, and include, for example, x-ray crystallography and 2 dimensional nuclear magnetic resonance.
One approach for preparing antibodies to a protein is the selection and preparation of an amino acid sequence of all or part ofthe protein, chemically synthesizing the sequence and injecting it into an appropriate animal, typically a rabbit, hamster or a mouse.
Oligopeptides can be selected as candidates for the production of an antibody to the subject colon or colorectal protein based upon the oligopeptides lying in hydrophilic regions, which are thus likely to be exposed in the mature protein.
Additional oligopeptides can be determined using, for example, the Antigenicity Index, Welling, G.W. et al., FEBSLett. 188:215-218 (1985), incoφorated herein by reference.
In other embodiments ofthe present invention, humanized monoclonal antibodies are provided, wherein the antibodies are specific for a protein according to the invention. The phrase "humanized antibody" refers to an antibody derived from a non-human antibody, typically a mouse monoclonal antibody. Alternatively, a humanized antibody can be derived from a chimeric antibody that retains or substantially retains the antigen-binding properties of the parental, non-human, antibody but which exhibits diminished immunogenicity as compared to the parental antibody when administered to humans. The phrase "chimeric antibody," as used herein, refers to an antibody containing sequence derived from two different antibodies (see, e.g., U.S. Patent No. 4,816,567) which typically originate from different species. Most typically, chimeric antibodies comprise human and murine antibody fragments generally human constant and mouse variable regions.
Because humanized antibodies are far less immunogenic in humans than the parental mouse monoclonal antibodies, they can be used for the treatment of humans with far less risk of anaphylaxis. Thus, these antibodies are useful in therapeutic applications that involve in vivo administration to a human such as, e.g., use as radiation sensitizers for the treatment of neoplastic disease or use in methods to reduce the side effects of, e.g., cancer therapy.
Humanized antibodies can be prepared using a variety of techniques including, for example: (1) grafting the non-human complementarity determining regions (CDRs) onto a human framework and constant region (a process referred to in the art as "humanizing"), or, alternatively, (2) transplanting the entire non-human variable domains, but "cloaking" them with a human-like surface by replacement of surface residues (a process referred to in the art as "veneering"). In the present invention, humanized antibodies include both "humanized" and "veneered" antibodies. These methods are disclosed in, e.g., Jones et al., Nature 321:522-525 (1986); Morrison et al., Proc. Natl. Acad. Sci, USA., 81:6851-6855 (1984); Morrison and Oi, Adv. Immunol, 44:65-92 (1988); Verhoeyer et al., Science 239: 1534- 1536 (1988); Padlan, Molec. Immun. 28:489-498 (1991); Padlan, Molec. Immunol 31(3): 169-217 (1994); and Kettleborough, CA. et al., Protein Eng. 4(7):773-83 (1991) each of which is incoφorated herein by reference.
The phrase "complementarity determining region" refers to amino acid sequences which together define the binding affinity and specificity ofthe natural Fv region of a native immunoglobulin-binding site. See, e.g., Chothia et al., J. Mol. Biol 196:901-917 (1987);
Kabat et al., U.S. Dept. of Health and Human Services NIH Publication No. 91-3242 (1991). The phrase "constant region" refers to the portion ofthe antibody molecule that confers effector functions. In the present invention, mouse constant regions are substituted by human constant regions. The constant regions ofthe subject-humanized antibodies are derived from human immunoglobulins. The heavy chain constant region can be selected from any ofthe five isotypes: alpha, delta, epsilon, gamma or mu.
One method of humanizing antibodies comprises aligning the non-human heavy and light chain sequences to human heavy and light chain sequences, selecting and replacing the non-human framework with a human framework based on such alignment, molecular modeling to predict the conformation ofthe humanized sequence and comparing to the conformation ofthe parent antibody. This process is followed by repeated back mutation of residues in the CDR region which disturb the stracture ofthe CDRs until the predicted conformation ofthe humanized sequence model closely approximates the conformation of the non-human CDRs ofthe parent non-human antibody. Humanized antibodies can be further derivatized to facilitate uptake and clearance, e.g, via Ashwell receptors. See, e.g.,
U.S. Patent Nos. 5,530,101 and 5,585,089 which patents are incoφorated herein by reference.
Humanized antibodies to proteins according to the invention can also be produced using transgenic animals that are engineered to contain human immunoglobulin loci. For example, WO 98/24893 discloses transgenic animals having a human Ig locus wherein the animals do not produce functional endogenous immunoglobulins due to the inactivation of endogenous heavy and light chain loci. WO 91/10741 also discloses transgenic non-primate mammalian hosts capable of mounting an immune response to an immunogen, wherein the antibodies have primate constant and/or variable regions, and wherein the endogenous immunoglobulin-encoding loci are substituted or inactivated. WO 96/30498 discloses the use ofthe Cre/Lox system to modify the immunoglobulin locus in a mammal, such as to replace all or a portion ofthe constant or variable region to form a modified antibody molecule. WO 94/02602 discloses non-human mammalian hosts having inactivated endogenous Ig loci and functional human Ig loci. U.S. Patent No. 5,939,598 discloses methods of making transgenic mice in which the mice lack endogenous heavy claims, and express an exogenous immunoglobulin locus comprising one or more xenogeneic constant regions.
Using a transgenic animal described above, an immune response can be produced to a selected antigenic molecule, and antibody-producing cells can be removed from the animal and used to produce hybridomas that secrete human monoclonal antibodies. Immunization protocols, adjuvants, and the like are known in the art, and are used in immunization of, for example, a transgenic mouse as described in WO 96/33735. This publication discloses monoclonal antibodies against a variety of antigenic molecules including IL-6, IL-8, TNF, human CD4, L-selectin, gp39, and tetanus toxin. The monoclonal antibodies can be tested for the ability to inhibit or neutralize the biological activity or physiological effect ofthe corresponding protein. WO 96/33735 discloses that monoclonal antibodies against IL-8, derived from immune cells of transgenic mice immunized with IL-8, blocked IL-8-induced functions of neutrophils. Human monoclonal antibodies with specificity for the antigen used to immunize transgenic animals are also disclosed in WO 96/34096.
In the present invention, proteins and variants thereof according to the invention are used to immunize a transgenic animal as described above. Monoclonal antibodies are made using methods known in the art, and the specificity ofthe antibodies is tested using isolated colon or colorectal proteins according to the invention. Methods for preparation ofthe human or primate protein according to the invention or an epitope thereof include, but are not limited to chemical synthesis, recombinant DNA techniques or isolation from biological samples. Chemical synthesis of a peptide can be performed, for example, by the classical Merrifeld method of solid phase peptide synthesis (Merrifeld, J. Am. Chem. Soc. 55:2149, 1963 which is incoφorated by reference) or the FMOC strategy on a Rapid Automated Multiple Peptide Synthesis system [E. I. du Pont de Nemours Company, Wilmington, DE) (Caprino and Han, J. Org. Chem. 37:3404 (1972) which is incoφorated by reference]. Polyclonal antibodies can be prepared by immunizing rabbits or other animals by injecting antigen followed by subsequent boosts at appropriate intervals. The animals are bled and sera assayed against purified protein usually by ELISA or by bioassay based upon the ability to block the action of a gene according to the invention. When using avian species, e.g., chicken, turkey and the like, the antibody can be isolated from the yolk ofthe egg. Monoclonal antibodies can be prepared after the method of Milstein and Kohler by fusing splenocytes from immunized mice with continuously replicating tumor cells such as myeloma or lymphoma cells. [Milstein and Kohler, Nature 256:495-491 (1975); Gulfre and Milstein, Methods in Enzymology: Immunochemical Techniques 75:1-46, Langone and Banatis eds., Academic Press, (1981) which are incoφorated by reference]. The hybridoma cells so formed are then cloned by limiting dilution methods and supemates assayed for antibody production by ELISA, RIA or bioassay.
The unique ability of antibodies to recognize and specifically bind to target proteins provides an approach for treating an overexpression ofthe protein. Thus, another aspect of the present invention provides for a method for preventing or treating diseases involving overexpression ofthe a protein according to the invention by treatment of a patient with antibodies to specific tumor antigen according to the invention.
Specific antibodies, either polyclonal or monoclonal, to the protein can be produced by any suitable method known in the art as discussed above. For example, murine or human monoclonal antibodies can be produced by hybridoma technology or, alternatively, the tumor protein, or an immunologically active fragment thereof, or an anti-idiotypic antibody, or fragment thereof can be administered to an animal to elicit the production of antibodies capable of recognizing and binding to the tumor protein. Antibodies can be of any class or subclass, e.g., IgG, IgA, lgM, IgD, and IgE or in the case of avian species, IgY, and subclasses thereof.
The availability of isolated human or primate protein according to the invention allows for the identification of small molecules and low molecular weight compounds that inhibit the binding ofthe protein to binding partners, through routine application of high- throughput screening methods (HTS). HTS methods generally refer to technologies that permit the rapid assaying of lead compounds for therapeutic potential. HTS techniques employ robotic handling of test materials, detection of positive signals, and inteφretation of data. Lead compounds can be identified via the incoφoration of radioactivity or through optical assays that rely on absorbance, fluorescence or luminescence as read-outs. [Gonzalez, J.E. et al, Curr. Opin. Biotech. 9:624-63 1 (1998)].
Model systems are available that can be adapted for use in high throughput screening for compounds that inhibit the interaction of a protein with its ligand, for example by competing with the protein for ligand binding. Sarabbi et al, Anal. Biochem. 237:10-15 (1996) describe cell-free, non-isotopic assays for discovering molecules that compete with natural ligands for binding to the active site of IL-1 receptor. Martens, C. et al, Anal Biochem. 273:20-31 (1999) describe a generic particle-based nonradioactive method in which a labeled ligand binds to its receptor immobilized on a particle; label on the particle decreases in the presence of a molecule that competes with the labeled ligand for receptor binding.
The therapeutic gene polynucleotides and polypeptides ofthe present invention can be utilized in gene delivery vehicles. The gene delivery vehicle can be of viral or non- viral origin (see generally, Jolly, Cancer Gene Therapy 1:51-64 (1994); Kimura, Human Gene Therapy 5:845-852 (1994); Connelly, Human Gene Therapy 1:185-193 (1995); and Kaplitt, Nature Genetics 6:148-153 (1994)). Gene therapy vehicles for delivery of constructs including a coding sequence of a therapeutic according to the invention can be administered either locally or systemically. These constructs can utilize viral or non- viral vector approaches. Expression of such coding sequences can be induced using endogenous mammalian or heterologous promoters. Expression ofthe coding sequence can be either constitutive or regulated.
The present invention can employ recombinant retrovirases which are constructed to carry or express a selected nucleic acid molecule of interest. Retroviras vectors that can be employed include those described in EP 0415 731; WO 90/07936; WO 94/03622; WO 93/25698; WO 93/25234; U.S. Patent No. 5,219,740; WO 93/11230; WO 93/10218; Vile and Hart, Cancer Res. 53:3860-3864 (1993); Vile and Hart, Cancer Res. 53:962-967 (1993); Ram et al., Cancer Res. 53:83-88 (1993); Takamiya et al., J. Neurosci. Res. 33:493-503 (1992); Baba et al., J. Neurosurg. 79:729-735 (1993); U.S. Patent No. 4,777,127; GB Patent No. 2,200,651; and EP 0 345 242. Recombinant retrovirases useful in accordance with the present invention include those described in WO 91/02805. Packaging cell lines suitable for use with the above-described retroviral vector constructs can be readily prepared (see PCT publications WO 95/3 0763 and WO 92/05266), and used to create producer cell lines (also termed vector cell lines) for the production of recombinant vector particles. For example, packaging cell lines can be prepared from human (such as HT1080 cells) or mink parent cell lines, thereby allowing production of recombinant retrovirases that can survive inactivation in human serum.
The present invention also employs alphaviras-based vectors that can function as gene delivery vehicles. Vectors can be constructed from a wide variety of alphavirases, including, for example, Sindbis virus vectors, Semliki forest virus (ATCC VR-67; ATCC VR-1247), Ross River virus (ATCC VR-373; ATCC VR-1246) and Venezuelan equine encephalitis virus (ATCC VR-923; ATCC VR-1250; ATCC VR 1249; ATCC VR-532). Representative examples of such vector systems include those described in U.S. Patent Nos. 5,091,309; 5,217,879; and 5,185,440; and PCT Publication Nos. WO 92/10578; WO 94/21792; WO 95/27069; WO 95/27044; and WO 95/07994.
Gene delivery vehicles ofthe present invention can also employ parvovirus such as adeno-associated virus (AAV) vectors. Representative examples include the AAV vectors disclosed by Srivastava in WO 93/09239, Samulski et al., J. Vir. 63: 3822-3828 (1989); Mendelson et al., Virol 166: 154-165 (1988); and Flotte et al., P.NA.S. 90: 10613-10617 (1993).
Representative examples of adenoviral vectors include those described by Berkner, Biotechniques 6:616-621 (Biotechniques); Rosenfeld et al, Science 252:431-434 (1991); WO 93/19191; Kolls et al, P.N.A.S. 215-219 (1994); Kass-Bisleret al., P.Ν.A.S. 90: 11498- 11502 (1993); Guzman et al., Circulation 88: 2838-2848 (1993); Guzman et al, Cir. Res. 73: 1202-1207 (1993); Zabner et al., Cell 75: 207-216 (1993); Li et al., Hum. Gene Ther. 4: 403- 409 (1993); Cailaud et al., Eur. J. Neurosci. 5: 1287-1291 (1993); Vincent et al., Nat. Genet. 5: 130-134 (1993); Jaffe et al., Nat. Genet. 1: 372-378 (1992); and Levrero et al., Gene 101: 195-202 (1992). Exemplary adenoviral gene therapy vectors employable in this invention also include those described in WO 94/12649, WO 93/03769; WO 93/19191 ; WO 94/28938; WO 95/11984 and WO 95/00655. Administration of DΝA linked to kill adenoviras as described in Curiel, Hum. Gene Ther. 3: 147-154 (1992) can be employed.
Other gene delivery vehicles and methods can be employed, including polycationic condensed DΝA linked or unlinked to kill adenoviras alone, for example Curiel, Hum. Gene Ther. 3: 147-154 (1992); ligand-linked DΝA, for example see Wu, J. Biol. Chem. 264:
16985-16987 (1989); eukaryotic cell delivery vehicles cells, for example see U.S. Serial No. 08/240,030, filed May 9, 1994, and U.S. Serial No. 08/404,796; deposition of photopolymerized hydrogel materials; hand-held gene transfer particle gun, as described in U.S. Patent No. 5,149,655; ionizing radiation as described in U.S. Patent No. 5,206,152 and in WO 92/11033; nucleic charge neutralization or fusion with cell membranes. Additional approaches are described in Philip, Mol. Cell Biol. -7^:2411-2418 (1994), and in Woffendin, Proc. Natl Acad. Sci. 97:1581-1585 (1994).
Naked DNA can also be administered directly to a subject. Exemplary naked DNA introduction methods are described in WO 90/11092 and U.S. Patent No. 5,580,859. Uptake efficiency may be improved using biodegradable latex beads. DNA coated latex beads are efficiently transported into cells after endocytosis initiation by the beads. The method may be improved further by treatment of the beads to increase hydrophobicity and thereby facilitate disruption ofthe endosome and release ofthe DNA into the cytoplasm. Liposomes that can act as gene delivery vehicles are described in U.S. Patent No. 5,422,120, PCT Patent Publication Nos. WO 95/13 796, WO 94/23697, and WO 9 1/14445, and EP No. 0 524968. Further non- viral delivery suitable for use includes mechanical delivery systems such as the approach described in Woffendin et al., Proc. Natl. Acad. Sci. USA 91(24): 11581- 11585 (1994). Moreover, the coding sequence and the product of expression of such can be delivered through deposition of photopolymerized hydrogel materials. Other conventional methods for gene delivery that can be used for delivery ofthe coding sequence include, for example, use of hand-held gene transfer particle gun, as described in U.S. Patent No. 5,149,655; use of ionizing radiation for activating transferred gene, as described in U.S. Patent No. 5,206,152 and PCT Patent Publication No. WO 92/11033.
EXAMPLES
The following Examples have been included to illustrate modes of the invention. Certain aspects of the following Examples are described in terms of techniques and procedures found or contemplated by the present co-inventors to work well in the practice of the invention. These Examples illustrate standard laboratory practices ofthe co-inventors. In light of the present disclosure and the general level of skill in the art, those of skill will appreciate that the following Examples are intended to be exemplary only and that numerous changes, modifications, and alterations can be employed without departing from the scope of the invention. Example 1 Identification of CICO1-CICO3 Genes Through a collaboration with Analytical Pathology Medical Group (at Grossmont Hospital), IDEC obtained pairs of snap frozen normal and malignant colon tissue removed during surgery. RNA was extracted from 10 pairs of those samples and submitted for GeneTag analysis at Celera/ Applied Bio Systems (ABI). In brief, the RNA was reverse transcribed into cDNA, digested with a restriction enzyme, and linkers were ligated to the cDNA library. The library was amplified using the linker sequences as a primer with an additional nucleotide (A, T, G, or C) (+1 PCR) to generate 16 libraries. The libraries were further amplified using the linker sequences as primers with an additional two nucleotides (+2 PCR) to generate 256 libraries. Fluorescently labeled products from these +2 PCR reactions were separated by capillary elecfrophoresis and the amplified sequences were quantitated. The expression profile obtained from malignant colon RNA was compared to that obtained using RNA from the normal colon. Several sequences were identified to be at least five-fold overexpressed in three of three tumors. The expression results are summarized in Figure 1. Overexpressed sequences were purified and amplified by PCR using the linkers with three additional nucleotides (+3 PCR). The +3 peaks were purified and sequenced. These sequences are set forth below:
CICO1 (Celera IDEC Colon Overexpressed D(bs213msl34-185)
Using 185 bases of +3 PCR sequence from GeneTag bs213msl34, human tentative human consensus sequence (THC) 684921 was identified from the BLAST database.
bs213msl43-185 Nucleotide Sequence GATCCAGGAGAGGAAGGAGTTTCAGAAGGCAGGAGCTGGTCCTCTATGTCATGAAATGTAGA GGGTGAGGCCAAGGAGGACCTGAGAGAAGGTAATTAGATTTGGTGTTTACAGGCTGGTCCCT GTGGCCAGCCACCCCACCCACTTTA(SEQ ID NO:l)
THC 684921 Nucleotide Sequence TGAGGAAACTGTGGCTTAGAGGAAAAGGTCATTAGTTCATTTTGGGATTT GTTGATTTTCAGATGTTTGAGATGTTGAGGATGGATTGTCCAGCAGGCTA TTAAGATGTGGTGAAGGCTAGAAATGTTGATTTAGGAGGTATTGCCTTCG AGAAGATAAAGGAGGAGAAGAGGAGAGCATCATGCAAGCTAGAGAAGAGA AAGAAGAAAAGTATTCTGGGGAATGTCTCCTTTGGGAGCAGAAAGAAGAC TCTGACGGAGCAGCCATCCAGGAAGTGGAATGAGATCCAGGAGAGGAAGG AGTTTCAGAAGGCAGGAGCTGGTCCTCTATGTCATGAAATGTAGAGGGTG AGGCCAAGGAGGACCTGAGAGAAGGTAATTAGATTTGGTGTTTACAGGCT GGTCCCTGTGGCCAGCCACCCCACCCACTTTAAAATATTTACTCTACAAA TGTTAATGTGTGAAGAGTTGCATGCCAGAATATTTATGGCATCAGTGTTG GTGGATACAGAACATTGGGAAACAACCCATTAATAGCAGAATGGTAAATC TGGCCAGTGAATAGTATAGCTTTTTAAAAGGAGGCTGATGTCTGAATTCA CTTTCAAAGTTGTTCACAATGTATTGCTAAAATACAAAAATGTTGCAGAA CCATATGTATGAGAGAAACCCCTTTTTCT (SEQ ID NO : 2 )
CICO 2 (bs222ms233-191)
191 bases ofthe +3 PCR sequence from GeneTag bs222ms233-191 overlapped with the 3'UTR of four different hypothetical proteins in the BLAST database.
bs222ms233-191 Nucleotide Sequence gatccccatggtatgcttgaatctgctccctgaacttcctgccagtgcctccccgtacccca aaacaatgtcaccatggttaccacctacccagaagactgttccctcctcccaagacccttgt ctgcagtggtgctcctgcaggctgcccgtta(SEQ ID NO: 3)
chrl_70_2399.c mRNA Sequence (coding sequence in CAPITALS, no ATG at start)
AGTGTGGTGATGGTTGTCTTCGACAATGAGAAGGTCCCAGTAGAGCAGCT GCGCTTCTGGAAGCACTGGCATTCCCGGCAACCCACTGCCAAGCAGCGGG TCATTGACGTGGCTGACTGCAAAGAAAACTTCAACACTGTGGAGCACATT GAGGAGGTGGCCTATAATGCACTGTCCTTTGTGTGGAACGTGAATGAAGA GGCCAAGGTGTTCATCGGCGTAAACTGTCTGAGCACAGACTTTTCCTCAC AAAAGGGGGTGAAGGGTGTCCCCCTGAACCTGCAGATTGACACCTATGAC TGTGGCTTGGGCACTGAGCGCCTGGTACACCGTGCTGTCTGCCAGATCAA GATCTTCTGTGACAAGGGAGCTGAGAGGAAGATGCGCGATGACGAGCGGA AGCAGTTCCGGAGGAAGGTCAAGTGCCCTGACTCCAGCAACAGTGGCGTC AAGGGCTGCCTGCTGTCGGGCTTCAGGGGCAATGAGACGACCTACCTTCG GCCAGAGACTGACCTGGAGACGCCACCCGTGCTGTTCATCCCCAATGTGC ACTTCTCCAGCCTGCAGCGGTCTGGAGGGGCAGCCCCCTCGGCAGGACCC AGCAGCTCCAACAGGCTGCCTCTGAAGCGTACCTGCTCGCCCTTCACTGA GGAGTTTGAGCCTCTGCCCTCCAAGCAGGCCAAGGAAGGCGACCTTCAGA GAGTTCTGCTGTATGTGCGGAGGGAGACTGAGGAGGTGTTTGACGCGCTC ATGTTGAAGACCCCAGACCTGAAGGGGCTGAGGAATGCGATCTCTGAGAA GTATGGGTTCCCTGAAGAGAACATTTACAAAGTCTACAAGAAATGCAAGC GAGGAATCTTAGTCAACATGGACAACAACATCATTCAGCATTACAGCAAC CACGTCGCCTTCCTGCTGGACATGGGGGAGCTGGACGGCAAAATTCAGAT CATCCTTAAGGAGCTGTAAggcctctcgagcatccaaaccctcacgacct gcaaggggccagcagggacgtggccccacgccacacacaacctctccaca tgcctcagcgctgttacttgaatgccttccctgagggaagaggcccttga gtcacagacccacagacgtcagggccagggagagacctagggggtcccct ggcctggatccccatggtatgcttgaatctgctccctgaacttcctgcca gtgcctccccgtaccccaaaacaatgtcaccatggttaccacctacccag aagactgttccctcctcccaagacccttgtctgcagtggtgctcctgcag gctgcccgttaagatggtggcggcacacgctccctcccgcagcaccacgc cagctggtgcggcccccactctctgtcttccttcaacttcagacaaagga tttctcaacctttggtcagttaacttgaaaactcttgattttcagtgcaa atgacttttaaaagacactatattggagtctctttctcagacttcctcag cgcaggatgtaaatagcactaacgatcgactggaacaaagtgaccgctgt gtaaaactactgccttgccactcactgttgtatacatttcttatttacga ttttcatttgttatatatatatataaatatactgtatatatatgcaacat tttatatttttcatggatatgtttttatcatttcaaaaaatgtgtatttc acatttcttggactttttttagctgttattcagtgatgcattttgtatac tcacgtggtatttagtaataaaaatctatctatgtattacgtcac (SEQ ID NO: 4)
chrl_70_2399.c Amino Acid Sequence
SWMWFDNEKVPVEQLRF KHWHSRQPTAKQRVIDVADCKENFNTVEHI EEVAYNALSFVWNVNEEAKVFIGVNCLSTDFSSQKGVKGVP NLQIDTYD CGLGTERLVHRAVCQIKIFCDKGAERKMRDDERKQFRRKVKCPDSSNSGV KGCL SGFRGNETTYLRPETDLETPPVLFIPNVHFSS QRSGGAAPSAGP SSSNRLPLKRTCSPFTEEFEPLPSKQAKEGDLQRVLLYVRRETEEVFDAL MLKTPDLKGLRNAISEKYGFPEENIYKVYKKCKRGILVNMDNNIIQHYSN HVAF D GE DGKIQIILKEL(SEQ ID NO: 5)
chrl_70_2399.f mRNA Sequence (coding sequence in CAPITALS, no ATG at start) aagttgccccacctctctgagcattggcttccccatctgtgaaagaggag tgctgatgtttgccttctaggggcctagtgaggcttaagggtgagcagca ggcacacagaaagctagaaatacaggatcactgtgggacggtggggctgg ccacctgggcaggccacttacccagcggccccctctgtctccaggtgttc atcggcgtaaactgtctgagcacagacttttcctcacaaaagggggtgaa gggtgtceccctgaacctgcagattgacacctatgactgtggcttgggca ctgagcgcctggtacaccgtgctgtctgccagatcaagatcttctgtgac aagggagctgagaggaagatgcgcgatgacgagcggaagcagttccggag gaaggtcaagtgccctgactccagcaacagtggcgtcaagggctgcctgc tgtcgggcttcaggggcaatgagacgacctaccttcggccagagactgac ctggagacgccacccgtgctgttcatccccaatgtgcacttctccagcct gcagcggtctggaggggcagccccctcggcaggacccagcagctccaaca ggctgcctctgaagcgtacctgctcgcccttcactgaggagtttgagcct ctgccctccaagcaggccaaggaaggcgaccttcagagagttctgctgta tgtgcggagggagactgaggaggtgtttgacgcgctcatgttgaagaccc cagacctgaaggggctgaggaatgcgatctctgagaagtatgggttccct gaaGAGAACATTTACAAAGTCTACAAGAAATGCAAGCGAGGAATCTTAGT CAACATGGACAACAACATCATTCAGCATTACAGCAACCACGTCGCCTTCC TGCTGGACATGGGGGAGCTGGACGGCAAAATTCAGATCATCCTTAAGGAG CTGTAAggcctctcgagcatccaaaccctcacgacctgcaaggggccagc agggacgtggccccacgccacacacaacctctccacatgcctcagcgctg ttacttgaatgccttccctgagggaagaggcccttgagtcacagacccac agacgtcagggccagggagagacctagggggtcccctggcctggatcccc atggtatgcttgaatctgctccctgaacttcctgccagtgcctccccgta ccccaaaacaatgtcaccatggttaccacctacccagaagactgttccct cctcccaagacccttgtctgcagtggtgctcctgcaggctgcccgttaag atggtggcggcacacgctccctcccgcagcaccacgccagctggtgcggc ccccactctctgtcttccttcaacttcagacaaaggatttctcaaccttt ggtcagttaacttgaaaactcttgattttcagtgcaaatgacttttaaaa gacactatattggagtctctttctcagacttcctcagcgcaggatgtaaa tagcactaacgatcgactggaacaaagtgaccgctgtgtaaaactactgc cttgccactcactgttgtatacatttcttatttacgattttcatttgtta tatatatatataaatatactgtatatatatgcaacattttatatttttca tggatatgtttttatcatttcaaaaaatgtgtatttcacatttcttggac tttttttagctgttattcagtgatgcattttgtatactcacgtggtattt agtaataaaaatctatctatgtattacgtcac (SEQ ID NO: 6)
chrl_70_2399.f Amino Acid Sequence
MRDDERKQFRRKVKCPDSSNSGVKGC LSGFRGNETTY RPETDLETPPV FIPNVHFSS QRSGGAAPSAGPSSSNRLPLKRTCSPFTEEFEPLPSKQA KEGD QRVLLYVRRETEEVFDALMLKTPDLKGLRNAISEKYGFPEENIYK VYKKCKRGILVNMDNNIIQHYSNHVAFLLDMGELDGKIQIILKEL(SEQ ID NO: 7)
C1000572 mRNA Sequence (coding) ATGAAAAGGTCTGTGCGGCTGCTAAAGAACGACCCAGTCAACTTGCAGAA ATTCTCTTACACTAGTGAGGATGAGGCCTGGAAGACGTACCTAGAAAACC CGTTGACAGCTGCCACAAAGGCCATGATGAGAGTCAATGGAGATGATGAG AGTGTTGCGGCCTTGAGCTTCCTCTATGATTACTACATGTCGATGCTCTT CCCAGATATCCTGAAAACCTCCCCGGAACCCCCATGTCCAGAGGACTACC CCAGCCTCAAAAGTGACTTTGAATACACCCTGGGCTCCCCCAAAGCCATC CACATCAAGTCAGGCGAGTCACCCATGGCCTACCTCAACAAAGGCCAGTT CTACCCCGTCACCCTGCGGACCCCAGCAGGTGGCAAAGGCCTTGCCTTGT CCTCCAACAAAGTCAAGAGTGTGGTGATGGTTGTCTTCGACAATGAGAAG GTCCCAGTAGAGCAGCTGCGCTTCTGGAAGCACTGGCATTCCCGGCAACC CACTGCCAAGCAGCGGGTCATTGACGTGGCTGACTGCAAAGAAAACTTCA ACACTGTGGAGCACATTGAGGAGGTGGCCTATAATGCACTGTCCTTTGTG TGGAACGTGAATGAAGAGGCCAAGGTGTTCATCGGCGTAAACTGTCTGAG CACAGACTTTTCCTCACAAAAGGGGGTGAAGGGTGTCCCCCTGAACCTGC AGATTGACACCTATGACTGTGGCTTGGGCACTGAGCGCCTGGTACACCGT GCTGTCTGCCAGATCAAGATCTTCTGTGACAAGGGAGCTGAGAGGAAGAT GCGCGATGACGAGCGGAAGCAGTTCCGGAGGAAGGTCAAGTGCCCTGACT CCAGCAACAGTGGCGTCAAGGGCTGCCTGCTGTCGGGCTTCAGGGGCAAT GAGACGACCTACCTTCGGCCAGAGACTGACCTGGAGACGCCACCCGTGCT GTTCATCCCCAATGTGCACTTCTCCAGCCTGCAGCGGTCTGGAGGGAGCC TCCAGCAGCCAGGGGCTCCTCTCATTTTCCTGCGTGTGATGGAAAATGTC TTTTTCACTTCATTGCAGGCAGCCCCCTCGGCAGGACCCAGCAGCTCCAA CAGGCTGCCTCTGAAGCGTACCTGCTCGCCCTTCACTGAGGAGTTTGAGC CTCTGCCCTCCAAGCAGGCCAAGGAAGGCGACCTTCAGAGAGTTCTGCTG TATGTGCGGAGGGAGACTGAGGAGGTGTTTGACGCGCTCATGTTGAAGAC CCCAGACCTGAAGGGGCTGAGGAATGCGATCTCTGAGAAGTATGGGTTCC CTGAAGAGAACATTTACAAAGTCTACAAGAAATGCAAGCGAGGAATCTTA GTCAACATGGACAACAACATCATTCAGCATTACAGCAACCACGTCGCCTT CCTGCTGGACATGGGGGAGCTGGACGGCAAAATTCAGATCATCCTTAAGG AGCTGTAA(SEQ ID NO: 8)
C 1000572 Amino Acid Sequence MKRSVRLLKNDPVNLQKFSYTSEDEAWKTYLENPLTAATKAMMRVNGDDE SVAA SFLYDYYMSMLFPDI KTSPEPPCPEDYPSLKSDFEYTLGSPKAI HIKSGESPMAYLNKGQFYPVTLRTPAGGKG ALSSNKVKSWMWFDNEK VPVEQLRFWKHWHSRQPTAKQRVIDVADCKENFNTVEHIEEVAYNALSFV NVNEEAKVFIGVNCLSTDFSSQKGVKGVPLNLQIDTYDCGLGTER VHR AVCQIKIFCDKGAERKMRDDERKQFRRKVKCPDSSNSGVKGCLLSGFRGN ETTYLRPETDLETPPVLFIPNVHFSSLQRSGGSLQQPGAPLIFLRVMENV FFTSLQAAPSAGPSSSNRLPLKRTCSPFTEEFEPLPSKQAKEGDLQRVLL YVRRETEEVFDALMLKTPD KGLRNAISEKYGFPEENIYKVYKKCKRGIL VNMDNNIIQHYSNHVAFLLDMGELDGKIQIILKEL(SEQ ID NO: 9)
ctgChr_lctg20.176 mRNA Sequence (coding) ATGGAGGCAGGGGAGAAAAGCGCTCTGGGTGCCTGGAGCCCGCAGCCCTG GGCAGCCCCGGGCTACCGCAGGGCGCAAGGGATCCTGGGCTGCGGCCGAG GGCGCCGGAAGTCGCCGCCGACCGCCTGGGTCTCGCAGGAAAACAGCCGG CGCCCGCGAGCTGCCCAGCGTCGGGTTTTCCTGAAGAGCCCAGCTCCTCA CACCTTGGGGCCTGGTGGGATGGGAGACACTGTCCTGGATGAAGCCGCTG GGAGAGCTGCCGCCTCCTGTATGCTGAGGTCTGTGCGGCTGCTAAAGAAC GACCCAGTCAACTTGCAGAAATTCTCTTACACTAGTGAGGATGAGGCCTG GAAGACGTACCTAGAAAACCCGTTGACAGCTGCCACAAAGGCCATGATGA GAGTCAATGGAGATGATGAGAGTGTTGCGGCCTTGAGCTTCCTCTATGAT TACTACATGGGTCCCAAGGAGAAGCGGATATTGTCCTCCAGCACTGGGGG CAGGAATGACCAAGGAAAGAGGTACTACCATGGCATGGAATATGAGACGG ACCTCACTCCCCTTGAAAGCCCCACACACCTCATGAAATTCCTGACAGAG AACGTGTCTGGAACCCCAGAGTACCCAGATTTGCTCAAGAAGAATAACCT GATGAGCTTGGAGGGGGCCTTGCCCACCCCTGGCAAGGCAGCTCCCCTCC CTGCAGGCCCCAGCAAGCTGGAGGCCGGCTCTGTGGACAGCTACCTGTTA CCCACCACTGATATGTATGATAATGGCTCCCTCAACTCCTTGTTTGAGAG CATTCATGGGGTGCCGCCCACACAGCGCTGGCAGCCAGACAGCACCTTCA AAGATGACCCACAGGAGTCGATGCTCTTCCCAGATATCCTGAAAACCTCC CCGGAACCCCCATGTCCAGAGGACTACCCCAGCCTCAAAAGTGACTTTGA ATACACCCTGGGCTCCCCCAAAGCCATCCACATCAAGTCAGGCGAGTCAC CCATGGCCTACCTCAACAAAGGCCAGTTCTACCCCGTCACCCTGCGGACC CCAGCAGGTGGCAAAGGCCTTGCCTTGTCCTCCAACAAAGTCAAGAGTGT GGTGATGGTTGTCTTCGACAATGAGAAGGTCCCAGTAGAGCAGCTGCGCT TCTGGAAGCACTGGCATTCCCGGCAACCCACTGCCAAGCAGCGGGTCATT GACGTGGCTGACTGCAAAGAAAACTTCAACACTGTGGAGCACATTGAGGA GGTGGCCTATAATGCACTGTCCTTTGTGTGGAACGTGAATGAAGAGGCCA AGGTGTTCATCGGCGTAAACTGTCTGAGCACAGACTTTTCCTCACAAAAG GGGGTGAAGGGTGTCCCCCTGAACCTGCAGATTGACACCTATGACTGTGG CTTGGGCACTGAGCGCCTGGTACACCGTGCTGTCTGCCAGATCAAGATCT TCTGTGACAAGGGAGCTGAGAGGAAGATGCGCGATGACGAGCGGAAGCAG TTCCGGAGGAAGGTCAAGTGCCCTGACTCCAGCAACAGTGGCGTCAAGGG CTGCCTGCTGTCGGGCTTCAGGGGCAATGAGACGACCTACCTTCGGCCAG AGACTGACCTGGAGACGCCACCCGTGCTGTTCATCCCCAATGTGCACTTC TCCAGCCTGCAGCGGTCTGGAGGGCTCCAACTGCCTAGTTACCGGCCGCA GGACCATCTGCAATTCCCAGCCCTTCTGGGCATGCTGGGGCCCAGGCTGC CTCTGAAGCGTACCTGCTCGCCCTTCACTGAGGAGTTTGAGCCTCTGCCC TCCAAGCAGGCCAAGGAAGGCGACCTTCAGAGAGTTCTGCTGTATGTGCG GAGGGAGACTGAGGAGGTGTTTGACGCGCTCATGTTGAAGACCCCAGACC TGAAGGGGCTGAGGAATGCGATCTCTGAGAAGTATGGGTTCCCTGAAGAG AACATTTACAAAGTCTACAAGAAATGCAAGCGAGGAATCTTAGTCAACAT GGACAACAACATCATTCAGCATTACAGCAACCACGTCGCCTTCCTGCTGG ACATGGGGGAGCTGGACGGCAAAATTCAGATCATCCTTAAGGAGCTGTAA (SEQ ID NO: 10)
ctgChr_lctg20.176 Amino Acid Sequence
MEAGEKSA GA SPQP AAPGYRRAQGILGCGRGRRKSPPTAWVSQENSR RPRAAQRRVFLKSPAPHTLGPGGMGDTVLDEAAGRAAASCM RSVR LKN DPVNLQKFSYTSEDEA KTYLENPLTAATKAMMRVNGDDESVAALSF YD YYMGPKEKRILSSSTGGRNDQGKRYYHGMEYETDLTPLESPTHLMKFLTE NVSGTPEYPD LKKNNL SLEGALPTPGKAAPLPAGPSKLEAGSVDSYLL PTTDMYDNGSLNSLFESIHGVPPTQRWQPDSTFKDDPQESMLFPDILKTS PEPPCPEDYPSLKSDFEYT GSPKAIHIKSGESPMAYLNKGQFYPVTLRT PAGGKGLALSSNKVKSVVMWFDNEKVPVEQLRFWKHWHSRQPTAKQRVI DVADCKENFNTVEHIEEVAYNALSFVWNVNEEAKVFIGVNCLSTDFSSQK GVKGVPLNLQIDTYDCGLGTERLVHRAVCQIKIFCDKGAERKMRDDERKQ FRRKVKCPDSSNSGVKGCLLSGFRGNETTYLRPETDLETPPVLFIPNVHF SSLQRSGGLQLPSYRPQDHLQFPAL GMLGPRLPLKRTCSPFTEEFEPLP SKQAKEGDLQRVLLYVRRETEEVFDALMLKTPDLKG RNAISEKYGFPEE (SEQ ID NO: 11)
CICO3 (bs432ms434-222)
The 222 bases ofthe +3 PCR sequence from GeneTag bs432ms434-222 overlapped with the 3'UTR oftwo different hypothetical proteins in the BLAST database. bs432ms434-222 Nucleotide Sequence
GATCTGCAATCAGAACTATTGAACTTCTCCATTCAGACCGCCACTCACACCTATGGGAAAAG GGTAATGTATCATCGGCTTAGCAACAGGGAATACTATTCGTATGATGGAAAATGGGGACAAA AGGCTTTGGTACATAAAACATTATTCCTTCCTTGGCCTAAAAACTCATCGCCACCTACATTA (SEQ ID NO: 12)
chrl9_53_399.c mRNA Sequence tctggagcagctgaaaaacaaggaagtgaaacagccaattcctgccttaa ctaattaacccaccttacgacattccaccattatgacgtgttcctgccct gccccaactgatcaatcgaccctgtgacattcttctggacaatgagtccc atcatctctccaccatgcaccttgtgactccctcctctgctgacaacaga taaccacctttaactgtaactttccacagcctaccccagccctataaagc tgcccctctcctatctcccttcgctgactctcttttcagactcagcccac ttgcacccaagtgaattaacagccttgttgctcacacaaagcctgtttag gtggtcttctatacggacatgcttgacacttggtgccaaaatctgggcca gggggactccttcgtgagaccggccccctgtcctggccctcattccgtga agagatccacctgcgacctcgggtcctcagaccagcccaaggaacatctc accaatttcaaatcggatctcctcggcttagtggctgaagactgatgctg cccgatcgcctcagaagccccttggaccatcacagatgccgagcttcggg taactcttacggtggaggattcccagccatatgaagacaccctagctgga cgatcagtccttgtcaaaagtctgacccctcaaactctacagcctcaatg gaccagaccctacccggtcatttatagcacaccaactgccgtccatctgc aggaccctctccattgggttcaccattccagaataaagccatgcccatca gacagccagcttgatctctcctcttcctcctggaagccacaagattaggc cgagagccgatcagacaaacaacctacaacccttaagctcctggcagcgc ccagccaaggccatgcttccttgcaacactccttccaaatggccatccca gcatgcttccaagcaggcttcatccgttcctctggaccctcatctcttaa gacctgccgcctataaaaaggattatatcttgagaccctatcctctaaaa ttttttccacacccaaaacaaaaaatctctgggtcaaaagtctaaaacgc ttaggctggcaaccatcagatccttgcccatggtgtcctcaagcctactc tcatgaaatggacaacagtacacgcatatggggccagttccacatatttg gcaaccagaccagcatccaggacaacacaaagatctgcaatcagaactat tgaacttctccattcagaccgccactcacacctatgggaaaagggtaatg tatcatcggcttagcaacagggaatactattcgtatgatggaaaatgggg acaaaaggctttggtacataaaacattattccttccttggcctaaaaact catcgccacctacattaaagctaatatgcctgattactgtttttagagaa cttattttattagggcagttccaagctcaaaaatacgctaactggcacct tgttagctacataaaaatgcaccctagacccgaaacttactagactcatt ataaaattttctttaaggtgtccacgcagtccctggtcacacttgaagca gtccggagaaatatcagccctaccccagtaatccccagaaggaacttaca cttttttttaatcttttcctacaacttcatattttataaataaaaagaca aaaatgtcaggcctgtgagctgaagcttagccattgtaacccctgtgacc tgcacatatccgtccaggtggcctgcaggagccaagaagtctggagcagc cgaaaaaccacaaagaagtgaaacagccagttcctgccttaactaattaa cccaccttacgacattccaccattatgacttgtccaccattatgacttgt tcctgccctgccccaactgatcaatcaaccctgtgacattcttctcctgg acaatgagtcccatcatctctccaccatgcaccttgtgaccccctcctct gctgaggataaccacctttaactgtaactttccacgcctacccaagccct ataaagctgcccctctcctatctcccttcactgactctcttttcggactc agcccacttgcacccaagtgaattaacagccttgttgctcacacaaagcc tgattgggtgtcttctatacggacacgcgtgacaggaacctcaacccaaa ggcagtctgatgaggtgtctaagataaaagtagcggcacaaaggcttttg taaacagaggcgtttcatgtggttttcctttcctttccttatatgtgaaa aggtgacagaaaagaaatcttcctaaaagagtc (SEQ ID NO: 13)
chrl9_53_399.c Amino Acid Sequence MGPVPHI QPDQHPGQHKDLQSELLNFSIQTATHTYGKRVMYHRLSNREY YSYDGKWGQKALVHKTLFLPWPKNSSPPTLKLICLITVFRELILLGQFQA QKYAN H VSYIKMHPRPETY(SEQ ID NO: 14)
chrl9_53_399.b mRNA Sequence tctggagcagctgaaaaacaaggaagtgaaacagccaattcctgccttaa ctaattaacccaccttacgacattccaccattatgacgtgttcctgccct gccccaactgatcaatcgaccctgtgacattcttctggacaatgagtccc atcatctctccaccatgcaccttgtgactccctcctctgctgacaacaga taaccacctttaactgtaactttccacagcctaccccagccctataaagc tgcccctctcctatctcccttcgctgactctcttttcagactcagcccac ttgcacccaagtgaattaacagccttgttgctcacacaaagcctgtttag gtggtcttctatacggacatgcttgacacttggtgccaaaatctgggcca gggggactccttcgtgagaccggccccctgtcctggccctcattccgtga agagatccacctgcgacctcgggtcctcagaccagcccaaggaacatctc accaatttcaaatcggatctcctcggcttagtggctgaagactgatgctg cccgatcgcctcagaagccccttggaccatcacagatgccgagcttcggg taactcttacggtggaggattcccagccatatgaagacaccctagctgga cgatcagtccttgtcaaaagtctgacccctcaaactctacagcctcaatg gaccagaccctacccggtcatttatagcacaccaactgccgtccatctgc aggaccctctccattgggttcaccattccagaataaagccatgcccatca gacagccagcttgatctctcctcttcctcctggaagccacaagattaggc cgagagccgatcagacaaacaacctacaacccttaagctcctggcagcgc ccagccaaggccatgcttccttgcaacactccttccaaatggccatccca gcatgcttccaagcaggcttca ccgttcctctggaccctcatctcttaa gacctgccgcctataaaaaggattatatcttgagaccctatcctctaaaa ttttttccacacccaaaacaaaaaatctctgggtcaaaagtctaaaacgc ttaggctggcaaccatcagatccttgcccatggtgtcctcaagcctactc tcatgaaatggacaacagtacacgcatatggggccagttccacatatttg gcaaccagaccagcatccaggacaacacaaagtatgttgtttgttgttag agggcttgggacatttcactctttgccagcctcagcttaatccaggagac aaagattattttccttattatctcttctgcataggatctgcaatcagaac tattgaacttctccattcagaccgccactcacacctatgggaaaagggta atgtatcatcggcttagcaacagggaatactattcgtatgatggaaaatg gggacaaaaggctttggtacataaaacattattccttccttggcctaaaa actcatcgccacctacattaaagctaatatgcctgattactgtttttaga gaacttattttattagggcagttccaagctcaaaaatacgctaactggca ccttgttagctacataaaaatgcaccctagacccgaaacttactagactc at ataaaattttctttaaggtgtccacgcagtccctggtcacacttgaa gcagtccggagaaatatcagccctaccccagtaatccccagaaggaactt acacttttttttaatcttttcctacaacttcatattttataaataaaaag acaaaaatgtcaggcctgtgagctgaagcttagccattgtaacccctgtg acctgcacatatccgtccaggtggcctgcaggagccaagaagtctggagc agccgaaaaaccacaaagaagtgaaacagccagttcctgccttaactaat taacccaccttacgacattccaccattatgacttgtccaccattatgact tgttcctgccctgccccaactgatcaatcaaccctgtgacattcttctcc tggacaatgagtcccatcatctctccaccatgcaccttgtgaccccctcc tctgctgaggataaccacctttaactgtaactttccacgcctacccaagc cctataaagctgcccctctcctatctcccttcactgactctcttttcgga ctcagcccacttgcacccaagtgaattaacagccttgttgctcacacaaa gcctgattgggtgtcttctatacggacacgcgtgacaggaacctcaaccc aaaggcagtctgatgaggtgtctaagataaaagtagcggcacaaaggctt ttgtaaacagaggcgtttcatgtggttttcctttcctttccttatatgtg aaaaggtgacagaaaagaaatcttcctaaaagagtc (SEQ ID NO: 15)
chrl9_53_399.b Amino Acid Sequence
CCPIASEAPWTITDAELRVT TVEDSQPYEDT AGRSVLVKS TPQTLQP QWTRPYPVIYSTPTAVHLQDPLH VHHSRIKPCPSDSQLDLSSSSWKPQD (SEQ ID NO: 16)
EXAMPLE 2 Identification of Candidate Genes 1-4 Four DNA sequences were identified as being overexpressed in colon carcinoma using the Gene Logic (Gaithersburg, Maryland) Gene Express Oncology Datasuite. The sequences were identified in a datasuite search, which compared gene expression in colon tumors with expression in normal tissues. These sequences represent genes and encode antigens which are targets for colon cancer therapeutics.
The nucleotide sequences of each candidate gene are listed below. The first sequence listed for each candidate gene was obtained directly from the public NCBI database
(www.ncbi.nlm.nih.gov) and corresponds to the GENBANK Accession No. number listed in the Gene Logic database. Additional sequence information was obtained by sequencing EST clones corresponding to each candidate gene.
Candidate 1: GENBANK Accession No. W91975 W91975/IMAGE Clone 415310 3' mRNA Sequence
GGCTTCTAAGGTACATTATGTTTTACTTTAATAAATAAAAATTAACTT GAAGAAAAATGCAGNGCCCTATTTAATTGCTCTGCATGAAATGTACAG AAACGGCAACCTCTGCGATTCTAAGCACTGTGAACGCCCCAGCCACAC CGTGTCAACAAACCGTGTGGCACTTGGGAGAAGGCAGGGGTGATTTAC GANTAGTCATGTTTCGCCTCCACCCGAGTCACTGCCAAGGAGTGGACA GTGACACTGAATAAGCATNCGGNGCACCTCCTTCGGGAAGGGACTTGG CTGACATGGTAGGCCTTCCCACTGGAGCCTGTACTTTGTCTTGCTGGG CAGCACTCCANTCATGGGAAGGAACAATGANCAAGGCGTGGTGGTGGG GGTGNGTAGGCCTGAGCGCCGTTTTCCATGGTGACCTTCACTGAGCAG GCAGCAGGCACTGATGGGCAGTTGAGNCTGGNAGGAGTCAGGTCCTGG
TCNTGCCTCTGGTGTAACGCAGCANGCCATCAAAGGT ( SEQ ID NO : 17 )
AGE Clone 194681 T3 & T7 Consensus Sequence
AGAATTCGGCACGAGNTTTTTTTTCTCTTAGATCTCCAGGTTCCCTTCCTTACCCCGGGA AGCCTTTCTTCATCCCACCGTCCTGGGGCGTTNCACAGTGCTTAGAATCGCAGAGGTTGC
CGTTTCTGTACATTTCATGCAGAGCAATTAAATAGGGCACTGCATTTTTCTTCAAGTTAA
TTTTTATTTATTAAAGTAAAACATAATGTACCTTAGAAGCCAGACAGTCCTACAAGCTTA
TTATGTTGTACAGCGGCGTTCCGTCCCCCTCCCCAGCCCTCTCTTTCTAGAGGCAGCCAA
TTTCAGCTGTCTCTCTCTGCTTACCTACATATTTCCATGTTTCTTGGTTCATCACCTGGT GGCACCTTCAGTCTGGAAACACCTGCCCTTCACTTTAGGGGAATTGGGCCCCTGTTCGTT
TGATAAGTTTTCCTACCATTTTCTGATTTGTTTTTTCTTTCTGGAAAATGTATTAGTCAG
ATGTAGGCTTTTCTGGATTAATCCTTCAACTTTCCTTTCTTTCTTTCCCTTCCTGCCTGT
CTCCCTGTTCTTTCTTACACTTTCTCAGGGAGATTCTTGACTGTATTTTCCAACTTTGTA
TCGACCATTTTACTTTTCCTGCCATATTTTCAATGTTTACTGATGTTTCTCTGCCCTTTC AGTGCATCCTGGTTTTATTTCATGTTAGACTGAATCCATGTGAAATTGATAACAGGTTTT
CAGCCCACACACACACACACAAAAAAAAAAAAAAAAAAAAAAAAAA (SEQ ID NO: 18) Candidate 2: GENBANK Accession No. AI694242 AI694242/IMAGE Clone 2327838 3' mRNA Sequence
TTTTGTTGGCTGAGGCGGTATTTTCCTTTTATTGCTGTTATGAGATT CAACATTTTTTCCAGAAATAACTTCTGAAAAGTGTGCCTAGATTTTG AACACTTGTGATCCTAACATGTGGTGAGAAAGGCTTTTCAAAACACA CACGTGTGGACAGAGGTCCACACACGGATACGTGTGCACACACGGGT GCCTTGGGCGTGCGTCTTCCAAAAGGGGCGAGTACAGCTATCAACTT GTGACTTCCAGGAGGCCTGGGTTTGCCTACGAAGGGGCCGTGTTCCC AGTTGGCGTTCACACGTGGTGTACACACACAGGCACAGGCACCGTGT CCCAAGGCCATCTCCCAAGGGCACCCGCAGACACTGGGCAGCCTTCT CCGAAGCTGTCAGTGTCCTTCCTCGTGAGAGGATGATGAAGAGGATG TGGTTTCCGCCGCCTCATCCACAGGCCGGCTG (SEQ ID NO: 19)
IMAGE Clone 2327838 T3 & T7 Consensus Sequence NAAAANGGCGCCNGNCCCANNTAAAATNNACCCNCCTAAAGGGGAAAAACTNNGGCGGCC GCCTTCGTTTTTTTTTTTTTTTTTTTGTGGTGGCTGAGGCGGTATTTTCCTTTTATTGCT GTTAAGAGATTCAACATTTTTTCCAGAAATAACTTCTGAAAAGGGGGCCTNAGATTTTGA ACACTTGGGATCCTAACAGGGGGTGAGAAAGGCTTTTCAAAACACACNACGGGTGGACAG AGGTCCACACACGGNATACGGGGGCACACACGGGTGCCTTGGGCGTGCGTCTTCCAAAAG GGGCGAGNTACAGCTATCAACTTGTGACTTCCAGGAGGCCTGGGTTTGCCTACGAAGGGG CCGNTGTTCCCAGTTGGCGTTCACACGTGGTGTACACACACAGGCACAGGCACCNGTGTC CCAANGGCCATCTNCCCAAGGGCACCCGCAGACACTGGGCAGCCTTCTCCGAAGCTGTCA GTGTCCTTCCTCGTGAGAGGATGATGAAGAGGATGTGGTTTCCGCCGCCTCATCCACAGG CCGGCTGCCCACGGAGCCTTAGACATCGAGGCCAGAGCGACAGAAGCCTGTGTGCTGACC GGCCTGGTCTCCTTTGACGTCTCGAGCAGCTTGGCAGGGTGGGAAAAGTAGCCTGAGAGT GATCCCCGGGCAGTGTCCGAGGCTCTGCCGTCCCCACCCCCACAGGCATCCAGGGGAGAG AAACAACCTGCGCCTGCGAGGCCGTGCGGACCCCGCTCCACTCACCCCGCCTGGGGGGCC AGAACCACCTCCCAGGGGCTTCCGCCAGTGCCGCAGTTGCTGACCCCAGGCAAACCTCGC CGCCTCCTGCCCCGGCGGGCCTGGGATTTGCGAATGTGTGAAGGCATTAGCTGCCAGTTG TAACTGGAACCCAGCCTAGAGGCCTCACTCCTCCAGCAGGAAGCCTTGTAATGCAGCGAA TCTGAACCCGGCCCAGCGTCCAGAGACAGGAAGCATTAATAGGAGCGAATGTGAACACTG TTCGCGCCCTGGCTGCGATTTATTGCCGATTGTGGGGAAAACATCAGTTGGTTGCAGAGT TTCATTCATCTTTAGGGACAGGACCGGTGTGTCTGGGTGGCAGTTTAGAGAGCTGGGACA GTCGGCATCACTCTGGGTGGCTCCTCTCAANCCCTGGTGCCTCGTGCCGAATTCTGGCCT CGAGGCATTCTNAGGGGCTNTATNC (SEQ ID NO: 20)
Candidate 3: GENBANK Accession No. AI680111 AI680111/IMAGE Clone 22520293' mRNA Sequence TTTTTTTTTTTTGT-ΞteATAAATATATTAGC-AAATGAAT^^
GATTCAAGCGTCTGTCTGGTTCAAATATAAATACCCATGTGGGTACCTAGGTGCTAGTC TCCCCACTAACTGAGGGAAAAAGGTTCCCAGGTGGGGTCCTCTGCCCACTTTGCCACCA CATTCACATTCCAAATGGGATAATGCCTGAGGGGCCATGAGTGGTCAGGCTGCCCTGGG GTGAATGTCACCCTGATGAGGCCCATCAGCTCTTGTCCACTCAGTGAGGCCAGACTTGT GCTCTAATCCACT (SEQ ID NO: 21)
IMAGE Clone 2324560 T7 Sequence CTNTGTANAAAGCTGGGTACGCGTAAGCTTGGGCCCCTCGAGGGATACTCTAGAGCGGC CGCCCTTTTTTTTTTTTTTTGTGGATAAATATATTAGCAAATAAATATATTTCTTAACA TAGTGCCTGATTCAAGCGTCTGTCTGGTTCAGATATAAATACCCATGTGGGTACCTAGG TGCTAGTCTCCCCACTAACTGAGGGAAAAAGGTTCCCAGGTGGGGTCCTCTGCCCACTT TGCCACCACATTCACATTCCAAATGGGATAATGCCTGAGGGGCCAAGAGTGGTCAGGCT GCCCTGGGGTGAATGTCACCCTGATGAGGCCCATCAGCTCTTGTCCACTCAGTGAGGCC AGACTTGTGCTCTAATCCACTCTCCTGTGGGTCCCTGGCCTGTATGGCTTATACTGGGG AGCTGGGCCTCTGGGCTGTCCAAACCCAAGGGTCACACTTTGCTTTTCCTTTGTTGTCC CCATTTTCCATCCTTGCTCTAAGACAAAACTTTTCCCAGAGAAGAACTCTTTGTTGTCC CCGCTCAGCTGTAATTCTGCCTTTTCTACCTTCATTCCATCCTTCCTCTGCCCAGATAA AGTCCAGCAGAAATTCCTCCTTTCTACCTCTCTGGGACTCTGAGACAGGAAATCTTCAA GGAGGAGTTTTTCCCTCCCCACTATTCTTATTCTCAACCCCCAGAAGAACCAANGGCTG CTGTACCCCCCTCAGGGACAGAACTCCACACTATANGGGGGAAAGNTTCANGGGACCCC TTCCTTTTANTGCTCANGGCTCCACCTATGCTACTGGNTCCTTTTGGCAAAAAAGGNAA ATGANAGAGCCAGGGGTTGCCCCNTGATGTAACANCCNTTACTGGGGANGGGNCCAANG NNGGTGNTCAAAGNNCCCCNAGGAGGGAGGNGANAAGGGGTCATGNGTTCTGCTNAANC CNCTGGTTGGTATAAANTTGANGNTTGGGGTGANGGAAACCAAAAANGGNTGGAAAAAG NAAAACACCTTTNNAAACCCTGGGTACCNNANATAAGNTTTTGGCCCNAAAAANTCNGC CNNCAAGGGATCCGCCCCNCCCCCCCAGGGAAAAANTTGGTTCCTNGGGNGAAAAGGAN TTTNCCCCCCNCAAATTTTNNCCNAAAAGNTTTGGAANTTGNAAAANAAAAGGANCCTT CCCCCCCCCNCCACAAAAAAAAAAAAAAAAAA (SEQ ID NO: 22)
IMAGE Clone 2324560 SP6 Sequence
CNNTTNCAAAAAGCAGGCTGGTACCGGTCCGGAATTCCCGGGATATCGTCGACCCACGC CGTCCGGTTTGCTGGTGTTGCTGAAATAACTCCAGCAGAAGGAAAATTAATGCAGTCCC ACCCGCTGTACCTGTGCAATGCCAGTGATGACGACAATCTGGAGCCTGGATTCATCAGC ATCGTCAAGCTGGAGAGTCCTCGACGGGCCCCCCGCCCCTGCCTGTCACTGGCTAGCAA GGCTCGGATGGCGGGTGAGCGAGGAGCCAGTGCTGTCCTCTTTGACATCACTGAGGATC GAGCTGCTGCTGAGCAGCTGCAGCAGCCGCTGGGGCTGACCTGGCCAGTGGTGTTGATC TGGGGTAATGACGCTGAGAAGCTGATGGAGTTTGTGTACAAGAACCAAAAGGCCCATGT GAGGATTGAGCTGAAGGAGCCCCCGGCCTGGCCAGATTATGATGTGTGGATCCTAATGA CAGTGGTGGGCACCATCTTTGTGATCATCCTGGCTTCGGTGCTGCGCATCCGGTGCCGC CCCCGCCACAGCAGGCCGGATCCGCTTCAGCAGAGAACAGCCTGGGCCATCAGCCAGCT GGCCACCAGGAGGTACCAGGCCAGCTGCAGGCAGGCCCGGGGTGAGTGGCCAGACTCAG GGAGCAGCTGCAGCTCAGCCCCTGTGTGTGCCATCTGTCTGGAGGGAGTTCTCTGAGGG GGCAGGAGCTACGGGTCATTTCCCTGCCTCCATGAGTTCCATCGTAACTGTGTGGACCC CTGGNTACATCAGCATCCGGACTTGCCCCCTCTTGCATGGTTCAACATCACANAGGGGA GATCCNTTTTCCCNGTCCCTGGGAACCTCTNCNATCTTACCAAGAACCAGGGTCGGAAG ACTCCCCCCTCATTTCNCCAGCATCCCCGGCATGNCCCACTACACCNTCCCTGGTNGCC TACCTGTTNGGGCCCTTCCCCGGAATGCAGGGGNTNGGGCCCCCNCNAACTGGGTCCTT TCCTGCCNTCCAGGNAGCCAGGCATGGGCCCCCCGAATCACCCCTTCCCCNAANATGGA NNATCCCCCGGGTTCCAGGAAAACAAACAACCNCTGGAAGGAANCCNNNACCCCNTNNC CCNAAGGCTGGGGAANGNAACNCCCCCNATTCCCCNTNNANGANCCCTNNGTTTNCNCN AGGCCCCTNACCCGGGCCNNGCCCCCNAAACAAAGGGANTTGANAAANT (SEQ ID NO: 23)
These sequences correspond to hypothetical gene FLJ20315/GENBANK Accession No. No. AK000322. AK000322 Nucleotide Sequence
AAAAAAAAAAAACTTTAGAGAAAGGAAGGGCCAAAACTACGACTTGGCTTTCTGAAACG GAAGCATAAATGTTCTTTTCCTCCATTTGTCTGGATCTGAGAACCTGCATTTGGTATTA GCTAGTGGAAGCAGTATGTATGGTTGAAGTGCATTGCTGCAGCTGGTAGCATGAGTGGT GGCCACCAGCTGCAGCTGGCTGCCCTCTGGCCCTGGCTGCTGATGGCTACCCTGCAGGC AGGCTTTGGACGCACAGGACTGGTACTGGCAGCAGCGGTGGAGTCTGAAAGATCAGCAG AACAGAAAGCTGTTATCAGAGTGATCCCCTTGAAAATGGACCCCACAGGAAAACTGAAT CTCACTTTGGAAGGTGTGTTTGCTGGTGTTGCTGAAATAACTCCAGCAGAAGGAAAATT AATGCAGTCCCACCCACTGTACCTGTGCAATGCCAGTGATGACGACAATCTGGAGCCTG GATTCATCAGCATCGTCAAGCTGGAGAGTCCTCGACGGGCCCCCCGCCCCTGCCTGTCA CTGGCTAGCAAGGCTCGGATGGCGGGTGAGCGAGGAGCCAGTGCTGTCCTCTTTGACAT CACTGAGGATCGAGCTGCTGCTGAGCAGCTGCAGCAGCCGCTGGGGCTGACCTGGCCAG TGGTGTTGATCTGGGGTAATGACGCTGAGAAGCTGATGGAGTTTGTGTACAAGAACCAA AAGGCCCATGTGAGGATTGAGCTGAAGGAGCCCCCGGCCTGGCCAGATTATGATGTGTG GATCCTAATGACAGTGGTGGGCACCATCTTTGTGATCATCCTGGCTTCGGTGCTGCGCA TCCGGTGCCGCCCCCGCCACAGCAGGCCGGATCCGCTTCAGCAGAGAACAGCCTGGGCC ATCAGCCAGCTGGCCACCAGGAGGTACCAGGCCAGCTGCAGGCAGGCCCGGGGTGAGTG GCCAGACTCAGGGAGCAGCTGCAGCTCAGCCCCTGTGTGTGCCATCTGTCTGGAGGAGT TCTCTGAGGGGCAGGAGCTACGGGTCATTTCCTGCCTCCATGAGTTCCATCGTAACTGT GTGGACCCCTGGTTACATCAGCATCGGACTTGCCCCCTCTGCGTGTTCAACATCACAGA GGGAGATTCATTTTCCCAGTCCCTGGGACCCTCTCGATCTTACCAAGAACCAGGTCGAA GACTCCACCTCATTCGCCAGCATCCCGGCCATGCCCACTACCACCTCCCTGCTGCCTAC CTGTTGGGCCCTTCCCGGAGTGCAGTGGCTCGGCCCCCACGACCTGGTCCCTTCCTGCC ATCCCAGGAGCCAGGCATGGGCCCTCGGCATCACCGCTTCCCCAGAGCTGCACATCCCC GGGCTCCAGGAGAGCAGCAGCGCCTGGCAGGAGCCCAGCACCCCTATGCACAAGGCTGG GGAATGAGCCACCTCCAATCCACCTCACAGCACCCTGCTGCTTGCCCAGTGCCCCTACG CCGGGCCAGGCCCCCTGACAGCAGTGGATCTGGAGAAAGCTATTGCACAGAACGCAGTG GGTACCTGGCAGATGGGCCAGCCAGTGACTCCAGCTCAGGGCCCTGTCATGGCTCTTCC AGTGACTCTGTGGTCAACTGCACGGACATCAGCCTACAGGGGGTCCATGGCAGCAGTTC TACTTTCTGCAGCTCCCTAAGCAGTGACTTTGACCCCCTAGTGTACTGCAGCCCTAAAG GGGATCCCCAGCGAGTGGACATGCAGCCTAGTGTGACCTCTCGGCCTCGTTCCTTGGAC TCGGTGGTGCCCACAGGGGAAACCCAGGTTTCCAGCCATGTCCACTACCACCGCCACCG GCACCACCACTACAAAAAGCGGTTCCAGTGGCATGGCAGGAAGCCTGGCCCAGAAACCG GAGTCCCCCAGTCCAGGCCTCCTATTCCTCGGACACAGCCCCAGCCAGAGCCACCTTCT CCTGATCAGCAAGTCACCGGATCCAACTCAGCAGCCCCTTCGGGGCGGCTCTCTAACCC ACAGTGCCCCAGGGCCCTCCCTGAGCCAGCCCCTGGCCCAGTTGACGCCTCCAGCATCT GCCCCAGTACCAGCAGTCTGTTCAACTTGCAAAAATCCAGCCTCTCTGCCCGACACCCA CAGAGGAAAAGGCGGGGGGGTCCCTCCGAGCCCACCCCTGGCTCTCGGCCCCAGGATGC AACTGTGCACCCAGCTTGCCAGATTTTTCCCCATTACACCCCCAGTGTGGCATATCCTT GGTCCCCAGAGGCACACCCCTTGATCTGTGGACCTCCAGGCCTGGACAAGAGGCTGCTA CCAGAAACCCCAGGCCCCTGTTACTCAAATTCACAGCCAGTGTGGTTGTGCCTGACTCC TCGCCAGCCCCTGGAACCACATCCACCTGGGGAGGGGCCTTCTGAATGGAGTTCTGACA CCGCAGAGGGCAGGCCATGCCCTTATCCGCACTGCCAGGTGCTGTCGGCCCAGCCTGGC TCAGAGGAGGAACTCGAGGAGCTGTGTGAACAGGCTGTGTGAGATGTTCAGGCCTAGCT CCAACCAAGAGTGTGCTCCAGATGTGTTTGGGCCCTACCTGGCACAGAGTCCTGCTCCT GGGAAAGGAAAGGACCACAGCAAACACCATTCTTTTTGCCGTACTTCCTAGAAGCACTG GAAGAGGACTGGTGATGGTGGAGGGTGAGAGGGTGCCGTTTCCTGCTCCAGCTCCAGAC CTTGTCTGCAGAAAACATCTGCAGTGCAGCAAATCCATGTCCAGCCAGGCAACCAGCTG CTGCCTGTGGCGTGTGTGGGCTGGATCCCTTGAAGGCTGAGTTTTTGAGGGCAGAAAGC TAGCTATGGGTAGCCAGGTGTTACAAAGGTGCTGCTCCTTCTCCAACCCCTACTTGGTT TCCCTCACCCCAAGCCTCATGTTCATACCAGCCAGTGGGTTCAGCAGAACGCATGACAC CTTATCACCTCCCTCCTTGGGTGAGCTCTGAACACCAGCTTTGGCCCCTCCACAGTAAG GCTGCTACATCAGGGGCAACCCTGGCTCTATCATTTTCCTTTTTTGCCAAAAGGACCAG TAGCATAGGTGAGCCCTGAGCACTAAAAGGAGGGGTCCCTGAAGCTTTCCCACTATAGT GTGGAGTTCTGTCCCTGAGGTGGGTACAGCAGCCTTGGTTCCTCTGGGGGTTGAGAATA AGAATAGTGGGGAGGGAAAAACTCCTCCTTGAAGATTTCCTGTCTCAGAGTCCCAGAGA GGTAGAAAGGAGGAATTTCTGCTGGACTTTATCTGGGCAGAGGAAGGATGGAATGAAGG TAGAAAAGGCAGAATTACAGCTGAGCGGGGACAACAAAGAGTTCTTCTCTGGGAAAAGT TTTGTCTTAGAGCAAGGATGGAAAATGGGGACAACAAAGGAAAAGCAAAGTGTGACCCT TGGGTTTGGACAGCCCAGAGGCCCAGCTCCCCAGTATAAGCCATACAGGCCAGGGACCC ACAGGAGAGTGGATTAGAGCACAAGTCTGGCCTCACTGAGTGGACAAGAGCTGATGGGC CTCATCAGGGTGACATTCACCCCAGGGCAGCCTGACCACTCTTGGCCCCTCAGGCATTA TCCCATTTGGAATGTGAATGTGGTGGCAAAGTGGGCAGAGGACCCCACCTGGGAACCT TTTTCCCTCAGTTAGTGGGGAGACTAGCACCTAGGTACCCACATGGGTATTTATATCT GAACCAGACAGACGCTTGAATCAGGCACTATGTTAAGAAATATATTTATTTGCTAATA TATTTAT ( SEQ ID NO : 24 )
The hypothetical protein encoded by this sequence is listed under GENBANK Accession No. BAA91085, provided below:
BAA91085 Amino Acid Sequence
MSGGHQ Q AALWPWLLMATLQAGFGRTGLVLAAAVESERSAEQKAVIRVIPLKMDPTG KLNLT EGVFAGVAEITPAEGKLMQSHPLY CNASDDDNLEPGFISIVKLESPRRAPRP CLSLASKARMAGERGASAVLFDITEDRAAAEQLQQPLGLTWPWLIWGNDAEKLMEFVY KNQKAHVRIELKEPPAWPDYDVWILMTWGTIFVIILASVLRIRCRPRHSRPDPLQQRT AWAISQLATRRYQASCRQARGEWPDSGSSCSSAPVCAICLEEFSEGQELRVISCLHEFH RNCVDP LHQHRTCPLCVFNITEGDSFSQSLGPSRSYQEPGRRLHLIRQHPGHAHYHLP AAYLLGPSRSAVARPPRPGPFLPSQEPGMGPRHHRFPRAAHPRAPGEQQRLAGAQHPYA QGWGMSHLQSTSQHPAACPVPLRRARPPDSSGSGESYCTERSGYLADGPASDSSSGPCH GSSSDSWNCTDISLQGVHGSSSTFCSSLSSDFDPLλ/YCSPKGDPQRVDMQPSVTSRPR SLDSWPTGETQVSSHVHYHRHRHHHYKKRFQWHGRKPGPETGVPQSRPPIPRTQPQPE PPSPDQQVTGSNSAAPSGRLSNPQCPRALPEPAPGPVDASSICPSTSSLFNLQKSSLSA RHPQRKRRGGPSEPTPGSRPQDATVHPACQIFPHYTPSVAYPWSPEAHPLICGPPGLDK RLLPETPGPCYSNSQPV LCLTPRQPLEPHPPGEGPSEWSSDTAEGRPCPYPHCQVLSA QPGSEEELEELCEQAV (SEQ ID NO: 25)
Candidate 4: GENBANK Accession No. AA813827 AA813827/IMAGE Clone 12717043' mRNA Sequence TTTTTTTTTAAACATTAAGATTTTATTACAAACCAGGCATTATATATTTCTTTACACTT AAGGAATAGATATGAAACAATCTTGGAGTAAAAATTAGAAGGCAACTTGCTTCAAGTTT GTACCAAGTCAATCAAGCAGAAACCTGAAGAACCTTGTTTTAAGATGAGAGTCATTTAT ACTTGGCAGGCATTTTCTTCCAATGAAAAAATAAAGTCAATGTGCCATTATCTTGACAC TTATAAAAATGTTTATAAAAAGCATTTAGGCCATTGATTCTCACAGTTGGCTGAATATT GGAATCACCTAGATTAAAAAAAATACTAATCCCTATACAACATCCCCAAAATTCAGATT TAATTAGTGTAAGTTAGGCCCTGGGCATATAGGCTGTTTTAAAATTCCTCGGGTGAGTC TAATGTGTA (SEQ ID NO: 26) IMAGE Clone 1341074 T7 Sequence
CCCNNCNNCCNNNNNNGNNNNNCTTANCTCGCAGNCANAATTCGGCCACGCAGGGTCGC CTTCGCCGCCATGGNACGCCACCGGGCGCTGACAGACCTATGGAGAGTCAGGGTGTGCC TCCCGGGCCTTATCGGGCCACCAAGCTGTGGAATGAAGTTACCACATCTTTTCGAGCAG GAATGCCTCTAAGAAAACACAGACAACACTTTAAAAAATATGGCAATTGTTTCACAGCA GGAGAAGCAGTGGATTGGCTTTATGACCTATTAAGAAATAATAGCAATTTTGGTCCTGA AGTTACAAGGCAACAGACTATCCAACTGTTGAGGAAATTTCTTAAGAATCATGTAATTG AAGATATCAAAGGGAGGTGGGGATCAGAAAATGTTGATGATAACAACCAGCTCTTCAGA TTTCCTGCAACTTCGCCACTTAAAACTCTACCACGAAGGTATCCAGAATTGAGAAAAAA CAACATAGAGAACTTTTCCAAAGATAAAGATAGCATTTTTAAATTACGAAACTTATCTC GTAGAACTCCTAAAAGGCATGGATTACATTTATCTCAGGAAAATGGCGAGAAAATAAAG CATGAAATAATCAATGAAAGATCAAGAAAATGCAATTGATAATAGAGAACTAAGCCAGG AAGATGTTGAAAGAAGNTTGGGAGATATGTTATTCTGATCCTACCTGCAAACCATTTTA AGGTGTGCCCATCCCCTAGAAGNAAGTTCTTAAATCCCAAACCAGGTAATTCCCCCAAN TANTTAATGNACAAACATGGNCCAATACAAGTTAANCCNGGGAGTAGTTNTTACTACAA AACCAATTCNGATGACCTTCCCCCACNGGNTNTTTNNCTNGCCATGGAAANGNCCCTAC CAAANTGGCCCAANAANNCANTGATTTGGAATAATCCNNCCTTTGGTTGGGATTNNANC AAATTGANTCCNAANNATCCCCAAATANTTTNCNAAANNCTCCCTGANCCCNACCTANC TTTGGAANTTNCCCAATTNTTTGGCAAACNTTTTGGGGANGGAAAGAATTCTCCGGATT TNAGCCCTTNTGGCAAAGGNTNCACCTNNNTTNAATTTNAAGANNNACACCCTNGGNAA ATNTAANGGGGCCCCCNNATTNTTTNAAATNCGCGGAANAAGNTCCCAGGNTCCCNTNT TTCCCCCCAAAATNNNATTGGGATTCCTNACCCCCCCAN (SEQ ID NO : 27)
IMAGE Clone 1341074 T3 Sequence CNNNNNANTGCGGCCGCTCATTTTTTTTTTTTTTTTTTCTCTATGNAAGCAGACTGNAG NAAGAAGGCACTCAGNTTGATTTGAAGGAATTCAAATTGTTTAAGTGAAGGAATTTTGA AGACTGTGGATCATCTTGAATTTTATGTATCCCACTGGATCTATCTGAAACTGTGATGT AGCCACAAACAACTACCAGGAAATGAAACAAAAATTAAGATGCAACTGTATGACAGTGG ACAAAAATAAAACAAAAACAATAGTAAAGTTAAAAAATAAAGCATTACTATAGTATATA TTGTTAGTATAGTATACACAGTAGTTGCTTAATTCAGAAGCCACTTAAATAGGACACAT GCAACATTCGGTTACAAACGTGCAAGACAGATGAGTGGTTTTCCCATTTGTAATATAAC TTTAAAAAATTATTTCAACAGCCTAATTAAATGGATTGAGCCAGAATACATTTAAAAAA TCTGTTCTCAGTCTGCAAGTACTAGAAACCTCATAAATATAAGATAATTGTGGTATAAT AAAATACATATATTTGATCTTTGTCCTTGGTACCTGGTATGGAGCTCCTAAAATCCTTG AAATTTCCTGAATGATAGAAGTCTTTAGTTACTCATAACAAGCCTATTTCAGCGNTATC CTGAGTTTCATGCCTAANGGTAACTGANGGCCNGGCCATGGGTTTGAATTTTCATCCAC CAACTACAACCCTTGTGGGGAGGAGAAAGGGNCTAGAAATTNAAGTTCNNTTGGNCCAC CAGTGACCCAATGAATTGGGTCCNGTCATGCCTTGGNTANTTAAACCTTCCAATTAAAA CNCNTAAAACATGCNAGGCTGANGGGAGTTTTNTAGGGTNNNGGAANCCTTGNATGGGG CTGGGNATCCCCGGATTGACCCAGAAANGGTAAAAAAAACNCTTNGGCCCCCCCCCCCC CCCTNACCCGGGGNCTTGGGAAACCCCTCCCTTTGGCCNTTTNCTGGAGGNCNACCCTT TTNAAATAAACTAAAAGCCATAGNTAAAGGGGCNTTTTNCTNNTTNCTGGGAANCTTGN ANGGAATTTTTNGACCCNGGNAAGGGGNTTTGAGGGAAANCCCAANTNGGTAATTGGCN GGGCGGGAATTTNNATACCCCCNGAACCCNATTNCNCGGAATTAAAAAAATTTNGGNNC GGNCCCCTTTNTNTNNNCCAGGGGTNAAANTTCTCNAAANNANAAA (SEQ ID NO: 28)
IMAGE Clone 1676529 T7 Sequence
AGCTCGNAGCCAGATTCGGCACGAGGGAGATTATATGTTTTATTTATCATTGTCTCTGC ATATCTGGAACAACGAAAGGCACATAGCAGTTGCTAAATAAATATCTTTTGAATGAATA TATGATTGCCTTATACTTCTTTTATATCCCCATCTTCTAATAGATTATGAAAACTAGAA TTCAAAATATATATACTGAACAAATGAATGACTGAAGCAATTGGGGATAATATTTAAGG CAAAACCAAATCTGATAAAATATACACATATTTTAAAAACACATACATATATATAAATA GATCAAAAGTGGAAAAAGAATATATAAAAGAGTGCAACATTTGGCAGCTGAGAATTATT TCATTGAGTTTTCAAATATTCTTCACATTCTTATACTTAGAAACAAAGAAGTAACCCCA AACAACTAATTCATTAGCTAATATCTCAGAACTTGCACATTTGCAGATAAATTTTCTTT TAAGAACAGAATTATAGTTTAATCCCTAACACAGCTCAGTTTTCAAAATTCAAGTAAAT AAAATTTTAGCACACATCATGATAGCCTTACTGGNATAGCTGTGTTAAAAACAAAAAGT ATTTGGTATCATCTATTGTTATGTGCTCTCAATTGAGATCTAGTTAGTTTCCTAAGAGT CTCACATTGATANCTATTTTGGGCACTTCCTTACATAATGNGNTTATTTAGAAATACCT TATTAATGACAGACTTCCTTTTGAGTAGCTACATTCTCAGATATGGCTNCATTTATCAA AGTTCCCCNAGGATTACCTAATTTTAATTCCAGTTAGNTATCTAAACTACGGAACTTTN GGNTTTCCTTAAANTCAACATTGGTTGCCTTGATTGGAAGGNTTGGCNCCCAAAAANGG CGGNCNTCCCNCNCCCGGGGGTGGNAANTCTTTTCNTGAANNTNCCAAGGNNAATTCCC TCCNGAAANCNGGNTTTAANTTTTTTNCCNTTTCCCCCTTNAANGGGAAACCCCCGGGT TTTNAAAAAAATTTTTCCCAAAANATTCNNCCNATGGGCCCCTTTGGAAAGGNAAAAAN TTTTTTGTCCCTTAAAAANCCCTGGNAACCNAATTTGGTTNANCAAATANAGGAAGG (SEQ ID NO:29)
IMAGE Clone 167529 T3 Sequence
GCGGCCGCTGGGCCTGNGTGTCGCCTTCGCCGCCATGGNCGCCACCGGGCGCTGACAGA CCTATGGAGAGTCAGGGTGTGCCTCCCGGGCCTTATCGGGCCACCAAGCTGTGGAATGA AGTTACCACATCTTTTCGAGCAGGAATGCCTCTAAGAAAACACAGACAACACTTTAAAA AATATGGCAATTGTTTCACAGCAGGAGAAGCAGTGGATTGGCTTTATGACCTATTAAGA AATAATAGCAATTTTGGTCCTGAAGTTACAAGGCAACAGACTATCCAACTGTTGAGGAA ATTTCTTAAGAATCATGTAATTGAAGATATCAAAGGGAGGTGGGGATCAGAAAATGTTG ATGATAACAACCAGCTCTTCAGATTTCCTGCAACTTCGCCACTTAAAACTCTACCACGA AGGTATCCAGAATTGAGAAAAAACAACATAGAGAACTTTTCCAAAGATAAAGATAGCAT TTTTAAATTACGAAACTTATCTCGTAGAACTCCTAAAAGGCATGGATTACATTTATCTC AGGAAAATGGCGAGAAAATAAAGCATGAAATAATCAATGAAGATCAAGAAAATGCAATT GATAATAGAGAACTAAGCCAGGAAGATGTTGAAGAAGTTTGGGAGATATGTTATTCTGA TCTACCTGCAAACCATTTTAGGTGTGCCATCCCTAGAAGAAGTCATAAATCCCAAACAA GTAATTCCCCAATATATAATGTACNACATGGCCAATACANGTAACGTGGGAGTAGTTAT ACTACAAACAAATCAGATGACCTCCCTCACTGGGTATTATCTGCCATGAAGNGCCTAGC AAATNGGCCAGAAGCATGATATGNAATAATCCACCTTTGNNGGATTTGACCGANATGTN TTNGAACATCCCGATTATTTCTAAACCCCTGACCNCTNNTACTTTGAAATNANAATTAT TGNAANCTTTGGGNTGCTNCNCCCTTTAAAGGGGTGCCNCCAAGCCTNNGTTNGTGNTG TTACTNCCCCCAANCGAAAAGNNCNCTTTATGGGTGNTNCCCAAGAACAATNTNN (SEQ ID NO:30)
These sequences correspond to hypothetical gene FLJ20354/GENBANK Accession No. No. AK000361.
AK000361 Nucleotide Sequence GTGCCGAGACTCACCACTGCCGCGGCCGCTGGGCCTGAGTGTCGCCTTCGCCGCCATGG ACGCCACCGGGCGCTGACAGACCTATGGAGAGTCAGGGTGTGCCTCCCGGGCCTTATCG GGCCACCAAGCTGTGGAATGAAGTTACCACATCTTTTCGAGCAGGAATGCCTCTAAGAA AACACAGACAACACTTTAAAAAATATGGCAATTGTTTCACAGCAGGAGAAGCAGTGGAT TGGCTTTATGACCTATTAAGAAATAATAGCAATTTTGGTCCTGAAGTTACAAGGCAACA GACTATCCAACTGTTGAGGAAATTTCTTAAGAATCATGTAATTGAAGATATCAAAGGGA GGTGGGGATCAGAAAATGTTGATGATAACAACCAGCTCTTCAGATTTCCTGCAACTTCG CCACTTAAAACTCTACCACGAAGGTATCCAGAATTGAGAAAAAACAACATAGAGAACTT TTCCAAAGATAAAGATAGCATTTTTAAATTACGAAACTTATCTCGTAGAACTCCTAAAA GGCATGGATTACATTTATCTCAGGAAAATGGCGAGAAAATAAAGCATGAAATAATCAAT GAAGATCAAGAAAATGCAATTGATAATAGAGAACTAAGCCAGGAAGATGTTGAAGAAGT TTGGAGATATGTTATTCTGATCTACCTGCAAACCATTTTAGGTGTGCCATCCCTAGAAG AAGTCATAAATCCAAAACAAGTAATTCCCCAATATATAATGTACAACATGGCCAATACA AGTAAACGTGGAGTAGTTATACTACAAAACAAATCAGATGACCTCCCTCACTGGGTATT ATCTGCCATGAAGTGCCTAGCAAATTGGCCAAGAAGCAATGATATGAATGATCCAACTT ATGTTGGATTTGAACGAGATGTATTCAGAACAATCGCAGATTATTTTCTAGATCTCCCT GAACCTCTACTTACTTTTGAATATTACGAATTATTTGTAAACATTTTGGTTGTTTGTGG CTACATCACAGTTTCAGATAGATCCAGTGGGATACATAAAATTCAAGATGATCCACAGT CTTCAAAATTCCTTCACTTAAACAATTTGAATTCCTTCAAATCAACTGAGTGCCTTCTT CTCAGTCTGCTTCATAGAGAAAAAAACAAAGAAGAATCAGATTCTACTGAGAGACTACA GATAAGCAATCCAGGATTTCAAGAAAGATGTGCTAAGAAAATGCAGCTAGTTAATTTAA GAAACAGAAGAGTGAGTGCTAATGACATAATGGGAGGAAGTTGTCATAATTTAATAGGG TTAAGTAATATGCATGATCTATCCTCTAACAGCAAACCAAGGTGCTGTTCTTTGGAAGG AATTGTAGATGTGCCAGGGAATTCAAGTAAAGAGGCATCCAGTGTCTTTCATCAATCTT TTCCGAACATAGAAGGACAAAATAATAAACTGTTTTTAGAGTCTAAGCCCAAACAGGAA TTCCTGTTGAATCTTCATTCAGAGGAAAATATTCAAAAGCCATTCAGTGCTGGTTTTAA GAGAACCTCTACTTTGACTGTTCAAGACCAAGAGGAGTTGTGTAATGGGAAATGCAAGT CAAAACAGCTTTGTAGGTCTCAGAGTTTGCTTTTAAGAAGTAGTACAAGAAGGAATAGT TATATCAATACACCAGTGGCTGAAATTATCATGAAACCAAATGTTGGACAAGGCAGCAC AAGTGTGCAAACAGCTATGGAAAGTGAACTCGGAGAGTCTAGTGCCACAATCAATAAAA GACTCTGCAAAAGTACAATAGAACTTTCAGAAAATTCTTTACTTCCAGCTTCTTCTATG TTGACTGGCACACAAAGCTTGCTGCAACCTCATTTAGAGAGGGTTGCCATCGATGCTCT ACAGTTATGTTGTTTGTTACTTCCCCCACCAAATCGTAGAAAGCTTCAACTTTTAATGC GTATGATTTCCCGAATGAGTCAAAATGTTGATATGCCCAAACTTCATGATGCAATGGGT ACGAGGTCACTGATGATACATACCTTTTCTCGATGTGTGTTATGCTGTGCTGAAGAAGT GGATCTTGATGAGCTTCTTGCTGGAAGATTAGTTTCTTTCTTAATGGATCATCATCAGG AAATTCTTCAAGTACCCTCTTACTTACTAGACTGCTAGTGGATAATAACATCTTGACTA CTTAAAAAAGGGACATATTGAAAATCCTGGAGATGGACTATTTGCTCCTTTGCCTAACT TACTCATACTGTAAGCAGATTAGTGCTCAGGAGTTTGATGAGCAAAAAGTTTCTACCTC TCAAGCTGCAATTGCTAGAACTCTTTAGAAAATATTATTAAAATACAGGAGTTTACCTT AAAGGAAAAAAAAAAAACAAAAAAAAAAAAAAAAAA (SEQ ID NO: 31)
The hypothetical protein encoded by this sequence is contained under GENBANK Accession No. BAA91111, provided below:
BAA91111 Amino Acid Sequence
MESQGVPPGPYRATKLWNEVTTSFRAG PLRKHRQHFKKYGNCFTAGEAVDWLYDLLRNNSN FGPEVTRQQTIQLLRKFLKNHVIEDIKGRWGSENVDDNNQLFRFPATSPLKTLPRRYPELRK NNIENFSKDKDSIFKLRNLSRRTPKRHGLHLSQENGEKIKHEIINEDQENAIDNRELSQEDV EEVWRYVILIYLQTILGVPSLEEVINPKQVIPQYIMYNMANTSKRGWILQNKSDDLPH VL SAMKCLANWPRSNDMNDPTYVGFERDVFRTIADYFLDLPEPLLTFEYYELFVNILWCGYIT VSDRSSGIHKIQDDPQSSKFLHLNNLNSFKSTECLLLSLLHREKNKEESDSTERLQISNPGF QERCAKKMQLVNLRNRRVSANDIMGGSCHNLIGLSNMHDLSSNSKPRCCSLEGIλtDVPGNSS KEASSVFHQSFPNIEGQNNKLFLESKPKQEFLLNLHSEENIQKPFSAGFKRTSTLTVQDQEE LCNGKCKSKQLCRSQSLLLRSSTRRNSYINTPVAEIIMKPNVGQGSTSVQTAMESELGESSA TINKRLCKSTIELSENSLLPASSMLTGTQSLLQPHLERVAIDALQLCCLLLPPPNRRKLQLL MR ISR SQNVDMPKLHDAMGTRSLMIHTFSRCVLCCAEEλtDLDELLAGRLVSFLMDHHQEI LQVPSYLLDC (SEQ ID NO: 32)
'Electronic Northerns' (E-Northerns) depicting gene expression profiles ofthe above described sequences were determined using the Gene Logic (Gaithersburg, Maryland) datasuite. See Figures 2-5. The expression of candidate 3 in normal and malignant human tissues was further investigated by PCR experiments using commercially available human cDNA panels and cDNA samples prepared in-house from human tissues and cell lines. See Figures 6A-6B and 7A-7B.
Expression of Glyceraldehyde 3 -phosphate dehydrogenase (GAPDH) was measured in these experiments as a control for cDNA integrity. GAPDH is a housekeeping gene expressed abundantly in all human tissues. The following primers were used to amplify a 482 base pair product ofthe GAPDH gene:
5 " ACCACAGTCCATGCCATCAC 3' (SEQ ID NO : 56 ) 5 ' TCCACCACCCTGTTGCTGTA 3 ' (SEQ ID NO : 57 )
The following primers were used to amplify a 507 base pair product ofthe candidate 3 gene: 5 ' TCCCACCCGCTGTACCTGTGC 3 ' (SEQ ID NO : 58) 5 ' CCTGCAGCTGGCCTGGTACCT 3 ' (SEQ ID NO : 59)
Colon tumor samples were obtained from Grossmont Hospital in La Mesa, California. Colorectal cancer cell line HCTl 16 was obtained from the American Type Culture Collection (ATCC, Manassas, Virginia). RNA was prepared from frozen tissue sections using the RNEasy® Maxi kit (Qiagen, #75162) or from fresh HCTl 16 cells using the RNEasy® Mini kit (Qiagen, #74104). For each sample, 2.5μg RNA was first treated with DNAse I (Amplification Grade, Invifrogen #18068-015), then reverse transcribed using the SUPERSCRIPT® First Strand Synthesis System for RT-PCR (Invifrogen # 12371-019). For PCR, 1/25 ofthe reverse transcriptase (RT) reaction was used to screen for candidate 3, and 1/50 was used for GAPDH. The positive control for candidate 3 was IMAGE 2324560, obtained from the ATCC. The following primers were used to amplify a 415 base pair product ofthe candidate 3 gene:
5' GGAAGATCTGTTGAAGTGCATTGCTGCAGCTGGTAG 3' (SEQ ID NO: 60) 5' CGCCATCCGAGCCTTGCTAGCCAG 3' (SEQ ID NO: 61)
EXAMPLE 3
Using the same technology employed in Example 1 to identify the CICO genes, the following sequences were identified as differentially expressed in colon cancer:
bs421ms433-258
At the +2 PCR stage, bs421ms433-258 was found to be overexpressed in malignant colon compared to normal colon (Figure 1). This peak was purifed and amplified by PCR using the linkers with three additional nucleotides (+3 PCR). The +3 peaks were purified and sequenced.
bs421ms433-258 Nucleotide Sequence GATCTCACTCAGCAGACAGCAGCAGCCCGGGAGCCTGAGCTCAGGAGGAACTCTTACCTGGA AATTGGGAACTGTATGGAGACTCCAAACTGACTTCTTTCAAAAAACAAAAACAAAAAATTTT TTTAGCTTTGACAAACACACAAAAGTGGTAATAAAGAGAGCCCTCCTTGTCAACCCAAAATG TGAGCCCCCTGTGGCAAAACCACCCCCTACCCCATTA (SEQ ID NO: 33)
These bases correspond to the 3 'UTR and some of the final coding exon of the hypothetical protein bK175E3.C22.6, , the sequence of which is set forth below:
bK175E3.C22.6 Nucleotide Sequence cggccgcggggcccggcgcggcgcgggccaaggagacggcgttcgtggag gtggtgctgttcgagtcgagcccaagcggcgattacaccacctacaccac cggcctcacgggccgcttctcgcgggccggggccacgctcagcgccgagg gcgagatcgtgcagatgcacccactgggcctatgtaataacaatgacgaa gaggacttgtatgaatatggctgggtaggagtggtgaagctggaacagcc agaattggacccgaaaccatgcctcactgtcctaggcaaggccaagcgag cagtacagcggggagctactgcagtcatctttgatgtgtctgaaaaccca gaagctattgatcagctgaaccagggctctgaagacccgctcaagaggcc ggtggtgtatgtgaagggtgcagatgccattaagctgatgaacatcgtca acaagcagaaagtggctcgagcaaggatccagcaccgccctcctcgacaa cccactgaatactttgacatggggattttcctggctttcttcgtcgtggt ctccttggtctgcctcatcctccttgtcaaaatcaagctgaagcagcgac gcagtcagaattccatgaacaggctggctgtgcaggctctagagaagatg gaaaccagaaagttcaactccaagagcaaggggcgccgggaggggagctg tggggccctggacacactcagcagcagctccacgtccgactgtgccatct gtctggagaagtacattgatggagaggagctgcgggtcatcccctgtact caccggtttcacaggaagtgcgtggacccctggctgctgcagcaccacac ctgcccccactgtcggcacaacatcatagaacaaaagggaaacccaagcg cggtgtgtgtggagaccagcaacctctcacgtggtcggcagcagagggtg accctgccggtgcattaccccggccgcgtgcacaggaccaacgccatccc agcctaccctacgaggacaagcatggactcccacggcaaccccgtcacct tgctgaccatggaccggcacggggagcagagcctctattccccgcagacc cccgcctacatccgcagctacccacccctccacctggaccacagcctggc cgctcaccgctgcggcctggagcaccgggcctactccccagcccacccct tccgcaggcccaagttgagtggccgcagcttctccaaggcagcttgcttc tcccagtatgagaccatgtaccagcactactacttccagggcctcagcta cccggagcaggaggggcagtccccacctagcctcgcaccccggggcccgg cccgtgcctttcctccgagcggcagtggcagcctgctcttccccaccgtg gtgcacgtggccccgccctcccacctggagagcggcagcacgtccagctt cagctgctatcacggccaccgctcggtgtgcagtggctacctggccgact gcccaggcagcgacagcagcagcagcagcagctccggccagtgccactgt tcctccagtgactctgtggtagactgcactgaggtcagcaaccagggcgt gtacgggagctgctccaccttccgcagctccctcagcagcgactatgacc ccttcatctaccgcagccggagcccctgtcgtgccagtgaggcggggggc tcgggcagctcgggccggggacctgccctgtgcttcgagggctccccgcc tcccgaggagctcccggcggtgcacagtcatggtgctgggcggggcgagc cttggccgggccctgcctctccctcgggggatcaggtgtccacctgcagc ctggagatgaactacagcagcaactcctccctggagcacagggggcccaa tagctctacctcagaagtggggctcgaggcttctcctggggccgcccctg acctcaggaggacctggaaggggggccacgagttgccgtcgtgtgcctgc tgctgcgagccccagccctccccagccgggcctagcgccggagcagctgg cagcagcaccttgttcctggggccccacctctacgagggctctggcccgg cgggtggggagccccagtcaggaagctcccagggcttgtacggccttcac cccgaccatttgcccaggacagatggggtgaaatacgagggtctgccctg ctgcttctatgaagagaagcaggtggcccgcgggggcggagggggcagcg gctgctacactgaggactactcggtgagtgtgcagtacacgctcaccgag gaaccaccgcccggctgctaccccggggcccgggacctgagccagcgcat ccccatcattccagaggatgtggactgtgatctgggcctgccctcggact gccaagggacccacagcctcggctcctggggtgggacgcgaggcccggat accccacggccccacaggggcctgggagcaacccgggaagaggagcgggc tctgtgctgccaggctagggccctactgcggcctggctgccctccggagg aggcgggtgctgtcagggccaacttccctagtgccctccaggacactcag gagtccagcaccactgccactgaggctgcaggaccgagatctcactcagc agacagcagcagcccgggagcctgagctcaggaggaactcttacctggaa attgggaactgtatggagactccaaactgacttctttcaaaaaacaaaaa caaaaaatttttttagctttgacaaacacacaaaagtggtaataaagaga gccctccttgtcaacccaaaatgtgagccccctgtggcaaaaccaccccc taccccattaacaaatcaacagacaaaattctccgagtcctttgcctctt ttgataacatgttgttctgttttgtaaagtgtgtgtgcttggggttccga ggtgtgggattgagttctctgctttgtttttttttaagatattgtatgta aatgtaaaaagttatttaaatatatattttaaagaaccctaactgccaac ttttgctgaaaaagaaaaaaaaatcactgctgcattaaatgaaccacatc atgtgtagatactgttgtctccctgaagggagctcaggcctttgaaaagc tcagggcttcacctgccttagaaaatgaaccagaaacttgaagtaaagct agttgataggggtacaggctctgaggagcagtgcaaaactgcctctttct ttctcgtggcaaatcccaatgtacacgatttcaggtctcagacgccatgc ctctccagcccacgcctttaggcaggtgatggcagcagctaggaataggg tgtacatgatccacagccctgcggagccaggtcaagccgctgctatgaaa gctccagggtgatggggacgattctgcccagtgtcctcagtctgtcccct caggtcatggtcccaagtgaaatgacagagttcacagccctggtcttggc tgaggtccaggtcatagtaagggcatgttcttggggccctcgacctgaac tctgaccctccgggcagggaagaggaggttgtcccctttggttgtcctgg ctttggagtcctttgcaaaaatattttgggccccctgccactggctgcag aaatggctcgacggggtgtgtggggacagacacccagaaggaatgtactt ttgtggccttggtgtccgatggggctgggggagagtgctctccactgacc cagcagcacacccatgtgcagtgcgcctgcatctgtgtgggggcagccac accccttggctgctgcttccttgggctgcctttctgggggcatgtgactg gacctacgaggtctgcactgagctccatttgaatgatacctttcctatcc catttcccccacggaagcaccgcttcagggttattcagtcctctgcctca tggctgaaattgctcatctcgtctgcagatgtctactatcctgtctacct aatgcactattatgtattgattctccatgagacagagagagagagagact atcagatagtttacacccaaagggtaggtttttgtatatttttccagcct tttttattaaggggaaggggagagtttaaaaacccaaaccgttgtggttt taaggtgtttcatttttaaaagggagagagaatctatttaaagctatttc agatcagggattgtcatcct ttttgtccaatgtattccttgttctttaa aaaaattttttttagaggaaactaatattagtctttgtgttcactaactc ttctggtcacttgtatttat tattcattcattcatcagatatttgttgc catctgaaagaactggcccagtgggtctgaaagctcgcttgagaatagga aactξgagacctggccccctgtgggtaggagaacaaggaccacctgggtt ctccagtcttgaacgagaatctcactcttatcagaatgtttttcttaacc tcagcgtatgatgaggaaatttacttatctctagctaggatttgacaaat tccaacatcaaatgatcaaaacatttgccactgaggcttcactggtgaga tccgttctccgtcctcgggtgcagtcccttgggggctgctcctcggactg cgccccgcacacctgttatcgagggtgtgagaagcgcctaagctggtgac atgtgatctgggacgcctteatttctcgggccaggagtagcagctgctaa ggacagcagcttgcattgcgtggttttagggaagcagggtctggctttta atatgaactgcaaaaagcagcttctcactgatatttttttgttgttgttt ctggggggtttttttgttttgtttttaatgcctttgagtgcatattttct tcctcgtctgaaaccgaactcccaaagtggctttctttagccctggctgg aaaaccacctctcaatagccttaagcaataaatagatgagtagagaatgt ggcttcaactgggcttattaaagtaagtgtgtctagttttcacttgaaca agtgatagctgcagatggcgaaagaaacccatttaatttttgtagcttac aggtggtagaaacaaaaatgcaattttaaaaccttaaataccaaatacca accattgccttttttttttttgagatggaattttgctcttgtcacccagg ctggagtgcaatggcgcgatctcacctcactgcaacctctgcctcccggg tccaagtgattctcctgcctcagcctcccaagtagctgggattacaggca tgcgccaccacacccagctaattttgtatttttggtagagacagggtatc tccatgttggtcaggctggtcttggattcccgacctcaggtgatccgccc acctcggcctcccaaagtgctgggattacaggcgtgagccaccatgcctg cccagcaataccaaccattgtcttttaaattcgtgttggcttctcagaca gggagatcactggaataaaataaccgatggtcttattttgtcacacgtaa atcaaaagaaatgtcctctttgaagttgtaagactccaccaatgacagac acccttttcggtggactctgagtggtgtgtagtggttttatagccatgga aactaggagtatctcactttccactgagaacccctgcccccaatccctct aagttggggtgtggcagttgggcagggtcaagtgacccagccctggctgt aggacagccatatacagtgaagagttctagaaccagctaaaaatggaagt ttgggtgtttaccaacaaggtacctctttatggatgcagccccagtaagc tggctttaactctcagctccttccctgtc cctcctaatccaagcccttt tataaaataaagccccttctgtcccactgctcacatacttatgtgctgct agtctctactcgaagttcgtgcaggactaatgcttttaaaatgaggtcta aaaaataattactagtcgagactattattctttaaacagaactgcctttt tctactctttatgtaaactctttctattgtgttggtctaacaaggcacta ttttaaaattttttaatttttcccatagcacttaaaagagattttgtaaa gaccttgctgtaaagattttgtaataaaatggtctaagggctctttttcc aacattaccatttttaaaaaatgttttaaaagctagaagacaacttatgt atattctgtatatgtatagcagcacatttcatttatggaaatatgttctc agaatatttatttactaatatatttatcttaagccatgtcttatgttgag agtgtgacattgttggaataatcattgaaaatgactaacacaagaccctg taaatacatgataattgcacacagattttacatatttgcagaccaaaaat gatttaaaacaagttgtagtcttctatggttttgtaacaaattgtacaca tgactgtaaaaaaaaaatacaattttatcaagtatgtgttata (SEQ ID NO: 34)
The above sequence encodes the following protein:
bKl75E3.C22.6 Amino Acid Sequence
MHPLGLCNNNDEEDLYEYGWVGλA/KLEQPELDPKPCLTVLGKAKRAVQRG ATAVIFDVSENPEAIDQLNQGSEDPLKRPVVYVKGADAIKLMNIVNKQKV ARARIQHRPPRQPTEYFDMGIFLAFFVWSLVCLILLVKIKLKQRRSQNS MNRLAVQALEKMETRKFNSKSKGRREGSCGALDTLSSSSTSDCAICLEKY IDGEELRVIPCTHRFHRKCVDPWLLQHHTCPHCRHNIIEQKGNPSAVCVE TSNLSRGRQQRVTLPVHYPGRVHRTNAIPAYPTRTS DSHGNPVTLLTMD RHGEQSLYSPQTPAYIRSYPPLHLDHSLAAHRCGLEHRAYSPAHPFRRPK LSGRSFSKAACFSQYETMYQHYYFQGLSYPEQEGQSPPSLAPRGPARAFP PSGSGSLLFPTWHVAPPSHLESGSTSSFSCYHGHRSVCSGYLADCPGSD SSSSSSSGQCHCSSSDSWDCTEVSNQGVYGSCSTFRSSLSSDYDPFIYR SRSPCRASEAGGSGSSGRGPALCFEGSPPPEELPAVHSHGAGRGEP PGP ASPSGDQVSTCSLEMNYSSNSSLEHRGPNSSTSEVGLEASPGAAPDLRRT WKGGHELPSCACCCEPQPSPAGPSAGAAGSSTLFLGPHLYEGSGPAGGEP QSGSSQGLYGLHPDHLPRTDGVKYEGLPCCFYEEKQVARGGGGGSGCYTE DYSVSVQYTLTEEPPPGCYPGARDLSQRIPIIPEDVDCDLGLPSDCQGTH SLGS GGTRGPDTPRPHRGLGATREEERALCCQARALLRPGCPPEEAGAV RANFPSALQDTQESSTTATEAAGPRSHSADSSSPGA (SEQ ID NO: 35)
This protein contains a transmembrane domain as determined by SMART (shown below), SOSUI, and TmPred. SMART also predicts that this protein contains a RING domain, which is a zinc finger domain involved in protein: protein interactions. The stracture ofthe protein is depicted schematically below:
Figure imgf000074_0001
EXAMPLE 4 Using the Gene Logic database and the methods described generally in Example 2, the following additional DNA sequences were identified as being overexpressed in colon tumor tissue:
AA781143/Hsl9 11415 28 1 1699a
Fragment AA781143 was upregulated 4.16-fold in the colon samples when compared to mixed normal tissue. E-Northem analysis of this fragment demonstrates that it is expressed in 69% ofthe colon tumors with greater than 50% malignant cells and shows little or no expression in normal tissues. See Figure 8.
AA781143 Nucleotide Sequence TTGTCTTCTACGACCAGCTGAAGCAAGTGATGAATGCGTACAGAGTCAAGCCGGCCGTCTTT GACCTGCTCCTGGCTGTTGGCATTGCTGCCTACCTCGGCATGGCCTACGTGGCTGTCCAGGT GAGCAGTGCCCAGGCTCAGCACTTCAGCCTCCTCTACAAGACCGTCCAGAGGCTGCTCGTGA AGGCCAAGACACAGTGACACAGCCACCCCCACAGCCGGAGCCCCCGCCGCTCCACAGTCCCT GGGGCCGAGCACGAGTTGGNAGGGGACCCTCTTCTCCCGTCNTGCCNTCGGGTTGCCCGCCT CCTCCAGAGACTTNNCAAGGGCCCATCACCACTGGCCTCTGGGCACTTGTGCTGAGACTCTG GGACCCAGGCAGCTGCCACCTTGTCACCATGAGAGAATTTGGGGAGTGCTTGCATGCTAGCC AGCAGGCTCCTGTCTGGGTGCCACGGGGCCAGCATTTTGGAGGGAGCTTCCTTCCTTCCTTC CTGGACAGGTCGTCATGATGGATGCACTGACTGACCGTCTGGGGCTCAGGCTGGTGTGGGAT GCAGCCGGCCG (SEQ ID NO: 36)
The GeneLogic database calls this protein "hypothetical protein from EUROIMAGE 2021883."
EUROIMAGE 2021883 Nucleotide Sequence CCAGAGTTTGTCTTCTACGACCAGCTGAAGCAAGTGATGAATGCGTACAGAGTCAAGCCGGC CGTCTTTGACCTGCTCCTGGCTGTTGGCATTGCTGCCTACCTCGGCATGGCCTACGTGGCTG TCCAGCACTTCAGCCTCCTCTACAAGACCGTCCAGAGGCTGCTCGTGAAGGCCAAGACACAG TGACACAGCCACCCCCACAGCCGGAGCCCCCGCCGCTCCACAGTCCCTGGGGCCGAGCACGA GTGAGTGGACACTGCCCCGCCGCGGGCGGCCCTGCAGGGACAGGGGCCCTCTCCCTCCCCGG CGGTGGTTGGAACACTGAATTACAGAGCTTTTTTCTGTTGCTCTCCGAGACTGGGGGGGGAT TGTTTCTTCTTTTCCTTGTCTTTGAACTTCCTTGGAGGAGAGCTTGGGAGACGTCCCGGGGC CAGGCTACGGACTTGCGGACGAGCCCCCCAGTCCTGGGAGCCGGCCGCCCTCGGTCTGGTGT AAGCACACATGCACGATTAAAGAGGAGACGCCGGGACCCCCTGCCCGATCGCGCGCGGCCTC CGCCCACCGCCTCCTGCCGCAAGGGGCCTGGACTGCAGGCCTGACCTGCTCCCTGCTCCGTG TCTGTCCTAGGACGTCCCCTCCCGCTCCCCGATGGTGGCGTGGACATGGTTATTTATCTCTG CTCCTTCTTGCCTGGAGGAGGGCAGTGCCAGCCCTGGGGTTCTGGGATTCCAGCCCTCCTGG AGCCTTTTGTTCCCCATGTGGTCTCAGTGACCCGTCCCCCTGACAGTGGGCTCGGGGAGCTG CATCACCCAGCCTTCCCCTTCTCCGACTGCAGGGTCTGATGTCATCATTGACAGCCTTTGCT TCGTGGGGGCCTGGCAGGGCCCCTGCCTCCCCGACCCCCGACCCACTGCAAATCCCCGTTCC CCTGCACTCCTCTTCTCCCAGCCCATCCCTCCGGCCCCTGTGCCTCTGCGGCCCCAGCCCAG CTCCCAGGGCCGTCACCTGCTTGGCCCTGGCCCAGCTCCCTGCCCTGAGTCCTGAGCCAGTG CCTGGTGTTTCCTGGGCTCGGTACTGGGCCCCCAGGCCATCCAGGCTTTGCCACGGCCAGTT GGTCCTCCCTGGGGAACTGGGTGCGGGTGGAGTACTGGGAGGCAGGAGGTGGCCCGGGGAGG CCTTGTGGCTCCTCCCCTCGCTCCTCGCCCTGGGCCTCAGCTTCCTCATCAATAGAAAGGAT GTGTTCGGGGTGGGGGCGTCAGGTGAGAACGTTTGCTGGGAAGGAGAGGACTTGGGGCATGG CCTCTGGGGCCACCCTTCCTGGAACTCAGAGAGGAAGGTCCGGGCCCTCGGGAAGCCTTGGA CAGAACCCTCCACCCCGCAGACCAGGCGTCGTGTGTGTGTGGGAGAGAAGGAGGCCCGTGTT GAGCTCAGGGAGACCCCGGTGTGTCCGTTCTTAGCAATATAACCTACCCAGTGCGTGCCGAG CAGGCTTGGTGGGGAAGGGACTTGAGCTGGGCAAGTCCTGGCCTGGCACCCGCAGCCGTCTC CCTTCCGTGGCCCAGGGAGGTGTTTGCTGTCCGAAGGACCTGGGCCGGCCCATGGGAGCCTG GGGTTCTGTCCAGATAGGACCAGGGGGTCTCACTTTGGCCACCAGTTCTTCGGCCAGCACCT CTGCCCTCCAGAACCTGCAGCCTGGAGGGGTGAGGGGACAACCACCCCTCTTTCCTCCAGGT TGGCAGGGGACCCTCTTCTCCCGTCTGCCCTGCGGGTTGCCCGCCTCCTCCAGAGACTTGCC CAAGGGCCCATCACCACTGGCCTCTGGGCACTTGTGCTGAGACTCTGGGACCCAGGCAGCTG CCACCTTGTCACCATGAGAGAATTTGGGGAGTGCTTGCATGCTAGCCAGCAGGCTCCTGTCT GGGTGCCACGGGGCCAGCATTTTGGAGGGAGCTTCCTTCCTTCCTTCCTGGACAGGTCGTCA TGATGGATGCACTGACTGACCGTCTGGGGCTCAGGCTGGTGTGGGATGCAGCCGGCCGATGA GAAAATAAAGCCATATTGAATGAT (SEQ ID NO: 37)
EUROIMAGE 2021883 Amino Acid Sequence
PEFVFYDQLKQVMNAYRVKPAVFDLLLAVGIAAYLGMAYVAVQHFSLLYKTVQRLLVKAKTQ (SEQ ID NO:38)
The protein set forth above contains one TM (transmembrane domain) by SMART, SOSUI, and TmPred prediction programs. However, the BLAST database and EST sequences suggest that the following alternative nucleotide and protein sequences correspond to AA781143:
Hsl9_l 1415_28_l_1699.a Nucleotide Sequence gcaaggtcacgtcctgtccccacctttcgcccctcaccctagctccccca acgccaaagacaaggttaagaaagtgatatcgcgaaatagttttttaaag cattttattgcattttatgacttggagtttatgtgaaacctcaacggtat tagccgaacagcctgccgcaccttccgggagttccagagtgggcctacaa ctcccacagggctccgcgagcgccggacggacggactacaattcccgaca ggcagcgcggctggcggggcggttcgccgcggtgcccacaggacctcagg gcgagtgcgggctgccccgcgcggcgcccgcaggaccccggcggctaccc atgccgaggtgagtccgcgggagccgccgccgccgccgtcccgtcccagc tgccgccccgcgcggccccgccgccggccaggATGCTGGAGGAAGCGGGC GAGGTGCTGGAGAACATGCTGAAGGCGTCTTGTCTGCCGCTCGGCTTCAT CGTCTTCCTGCCCGCTGTGCTGCTGCTGGTGGCGCCGCCGCTGCCTGCCG CCGACGCCGCGCACGAGTTCACCGTGTACCGCATGCAGCAGTACGACCTG CAGGGCCAGCCCTACGGCACACGGAATGCAGTGCTGAACACGGAGGCGCG CACGATGGCGGCGGAGGTGCTGAGCCGCCGCTGCGTGCTCATGCGGCTAC TGGACTTCTCCTACGAGCAGTACCAGAAGGCCCTGCGGCAGTCGGCGGGC GCCGTGGTCATCATCCTGCCCAGGGCCATGGCCGCCGTGCCCCAGGACGT CGTCCGGCAATTCATGGAGATCGAGCCGGAGATGCTGGCCATGGAGACCG CCGTCCCCGTGTACTTTGCCGTGGAGGACGAGGCCCTGCTGTCTATCTAC AAGCAGACCCAGGCTGCCTCCGCCTCCCAGGGCTCCGCCTCTGCTGCTGA AGTACTGCTGCGCACGGCCACTGCCAACGGCTTCCAGATGGTCACCAGCG GGGTACAGAGCAAGGCCGTGAGTGACTGGCTGATTGCCAGCGTGGAGGGG CGGCTGACGGGGCTGGGCGGAGAGGACCTTCCCACCATCGTCATCGTGGC CCACTACGACGCCTTTGGAGTGGCCCCCTGGCTGTCGCTGGGCGCGGACT CCAACGGGAGCGGCGTCTCTGTGCTGCTGGAGCTGGCACGCCTCTTCTCC CGGCTCTACACCTACAAGCGCACGCACGCCGCCTACAACCTCCTGTTCTT TGCGTCTGGAGGAGGCAAGTTTAACTACCAGGGAACCAAGCGCTGGCTGG AAGACAACCTGGACCACACAGACTCCAGCCTGCTTCAGGACAATGTGGCC TTCGTGCTGTGCCTGGACACCGTGGGCCGGGGCAGCAGCCTGCACCTGCA CGTGTCCAAGCCGCCTCGGGAGGGCACCCTGCAGCACGCCTTCCTGCGGG AGCTGGAGACGGTGGCCGCGCACCAGTTCCCTGAGGTACGGTTCTCCATG GTGCACAAGCGGATCAACCTGGCGGAGGACGTGCTGGCCTGGGAGCACGA GCGCTTCGCCATCCGCCGACTGCCCGCCTTCACGCTGTCCCACCTGGAGA GCCACCGTGACGGCCAGCGCAGCAGCATCATGGACGTGCGGTCCCGGGTG GATTCTAAGACCCTGACCCGTAACACGAGGATCATTGCAGAGGCCCTGAC TCGAGTCATCTACAACCTGACAGAGAAGGGGACACCCCCAGACATGCCGG TGTTCACAGAGCAGATGCAGATCCAGCAGGAGCAGCTGGACTCGGTGATG GACTGGCTCACCAACCAGCCGCGGGCCGCGCAGCTGGTGGACAAGGACAG CACCTTCCTCAGCACGCTGGAGCACCACCTGAGCCGCTACCTGAAGGACG TGAAGCAGCACCACGTCAAGGCTGACAAGCGGGACCCAGAGTTTGTCTTC TACGACCAGCTGAAGCAAGTGATGAATGCGTACAGAGTCAAGCCGGCCGT CTTTGACCTGCTCCTGGCTGTTGGCATTGCTGCCTACCTCGGCATGGCCT ACGTGGCTGTCCAGCACTTCAGCCTCCTCTACAAGACCGTCCAGAGGCTG CTCGTGAAGGCCAAGACACAGTGAcacagccacccccacagccggagccc ccgccgctccacagtccctggggccgagcacgagtgagtggacactgccc cgccgcgggcggccctgcagggacaggggccctctccctccccggcggtg gttggaacactgaattacagagcttttttctgttgctctccgagactggg gggggattgtttcttcttttccttgtctttgaacttccttggaggagagc ttgggagacgtcccggggccaggctacggacttgcggacgagccccccag tcctgggagccggccgccctcggtctggtgtaagcacacatgcacgatta aagaggagacgccgggaccccctgcccgatcgcgcgcggcctccgcccac cgcctcctgccgcaaggggcctggactgcaggcctgacctgctccctgct ccgtgtctgtcctaggacgtcccctcccgctccccgatggtggcgtggac atggttatttatctctgctccttcttgcctggaggagggcagtgccagcc ctggggttctgggattccagccctcctggagccttttgttccccatgtgg tctcagtgacccgtccccctgacagtgggctcggggagctgcatcaccca gccttccccttctccgactgcagggtctgatgtcatcattgacagccttt gcttcgtgggggcctggcagggcccctgcctccccgacccccgacccact gcaaatccccgttcccctgcactcctcttctcccagcccatccctccggc ccctgtgcctctgcggccccagcccagctcccagggccgtcacctgcttg gccctggcccagctccctgccctgagtcctgagccagtgcctggtgtttc ctgggctcggtactgggcccccaggccatccaggctttgccacggccagt tggtcctccctggggaactgggtgcgggtggagtactgggaggcaggagg tggcccggggaggccttgtggctcctcccctcgctcctcgccctgggcct cagcttcctcatcaatagaaaggatgtgttcggggtgggggcgtcaggtg agaacgtttgctgggaaggagaggacttggggcatggcctctggggccac ccttcctggaactcagagaggaaggtccgggccctcgggaagccttggac agaaccctccaccccgcagaccaggcgtcgtgtgtgtgtgggagagaagg aggcccgtgttgagctcagggagaccccggtgtgtccgttctttagcaat ataacctacccagtgcgtgccgagcaggcttggtggggaagggacttgag ctgggcaagtcctggcctggcacccgcagccgtctcccttccgtggccca gggaggtgtttgctgtccgaaggacctgggccggcccatgggagcctggg gttctgtccagataggaccagggggtctcactttggccaccagttcttcg gccagcacctctgccctccagaacctgcagcctggaggggtgaggggaca accacccctctttcctccaggttggcaggggaccctcttctcccgtctgc cctgcgggttgcccgcctcctccagagacttgcccaagggcccatcaeca ctggcctctgggcacttgtgctgagactctgggacccaggcagctgccac cttgtcaccatgagagaatttggggagtgcttgcatgctagccagcaggc tcctgtctgggtgccacggggccagcattttggagggagcttccttcctt ccttcctggacaggtcgtcatgatggatgcactgactgaccgtctggggc tcaggctggtgtgggatgcagccggccgatgagaaaataaagccatattg aatgatcg (SEQ ID NO: 39)
Hsl9_11415_28_l_1699.a Amino Acid Sequence MLEEAGEVLENMLKASCLPLGFIVFLPAVLLLVAPPLPAADAAHEFTVYR QQYDLQGQPYGTRNAVLNTEARTMAAEVLSRRCVLMRLLDFSYEQYQKA
LRQSAGAWII PRAMAAVPQD RQFMEIEPE LA ETAVPVYFAVEDE
ALLSIYKQTQAASASQGSASAAEVLLRTATANGFQMVTSGVQSKAVSDWL
IASVEGRLTGLGGEDLPTIVIVAHYDAFGVAP LSLGADSNGSGVSVLLE LARLFSRLYTYKRTHAAYNLLFFASGGGKFNYQGTKRWLEDNLDHTDSSL
LQDNVAFVLCLDTVGRGSSLHLHVSKPPREGTLQHAFLRELETVAAHQFP
EVRFSMVHKRINLAEDVLA EHERFAIRRLPAFTLSHLESHRDGQRSSIM
DVRSRVDSKTLTRNTRIIAEALTRVIYNLTEKGTPPDMPVFTEQMQIQQE
QLDSVMDWLTNQPRAAQLVDKDSTFLSTLEHHLSRYLKDVKQHHVKADKR DPEFVFYDQLKQVMNAYRVKPAVFDLLLAVGIAAYLGMAYVAVQHFSLLY
KTVQRLLVKAKTQ (SEQ ID NO: 40)
GENBANK also identifies RefSeq Loc56926 as corresponding to AA781143, which nucleotide and protein sequences are set forth below:
RefSeq Loq56926 Nucleotide Sequence
GGCGAGGTGCTGGAGAACATGCTGAAGGCGTCTTGTCTGCCGCTCGGCTTCATCGTCTTCCT GCCCGCTGTGCTGCTGCTGGTGGCGCCGCCGCTGCCTGCCGCCGACGCCGCGCACGAGTTCA CCGTGTACCGCATGCAGCAGTACGACCTGCAGGGCCAGCCCTACGGCACACGGAATGCAGTG CTGAACACGGAGGCGCGCACGATGGCGGCGGAGGTGCTGAGCCGCCGCTGCGTGCTCATGCG GCTACTGGACTTCTCCTACGAGCAGTACCAGAAGGCCCTGCGGCAGTCGGCGGGCGCCGTGG TCATCATCCTGCCCAGGGCCATGGCCGCCGTGCCCCAGGACGTCGTCCGGCAATTCATGGAG ATCGAGCCGGAGATGCTGGCCATGGAGACCGCCGTCCCCGTGTACTTTGCCGTGGAGGACGA GGCCCTGCTGTCTATCTACAAGCAGACCCAGGCTGCCTCCGCCTCCCAGGGCTCCGCCTCTG CTGCTGAAGTACTGCTGCGCACGGCCACTGCCAACGGCTTCCAGATGGTCACCAGCGGGGTA CAGAGCAAGGCCGTGAGTGACTGGCTGATTGCCAGCGTGGAGGGGCGGCTGACGGGGCTGGG CGGAGAGGACCTTCCCACCATCGTCATCGTGGCCCACTACGACGCCTTTGGAGTGGCCCCCT GGCTGTCGCTGGGCGCGGACTCCAACGGGAGCGGCGTCTCTGTGCTGCTGGAGCTGGCACGC CTCTTCTCCCGGCTCTACACCTACAAGCGCACGCACGCCGCCTACAACCTCCTGTTCTTTGC GTCTGGAGGAGGCAAGTTTAACTACCAGGGAACCAAGCGCTGGCTGGAAGACAACCTGGACC ACACAGACTCCAGCCTGCTTCAGGACAATGTGGCCTTCGTGCTGTGCCTGGACACCGTGGGC CGGGGCAGCAGCCTGCACCTGCACGTGTCCAAGCCGCCTCGGGAGGGCACCCTGCAGCACGC CTTCCTGCGGGAGCTGGAGACGGTGGCCGCGCACCAGTTCCCTGAGGTACGGTTCTCCATGG TGCACAAGCGGATCAACCTGGCGGAGGACGTGCTGGCCTGGGAGCACGAGCGCTTCGCCATC CGCCGACTGCCCGCCTTCACGCTGTCCCACCTGGAGAGCCACCGTGACGGCCAGCGCAGCAG CATCATGGACGTGCGGTCCCGGGTGGATTCTAAGACCCTGACCCGTAACACGAGGATCATTG CAGAGGCCCTGACTCGAGTCATCTACAACCTGACAGAGAAGGGGACACCCCCAGACATGCCG GTGTTCACAGAGCAGATGCAGATCCAGCAGGAGCAGCTGGACTCGGTGATGGACTGGCTCAC CAACCAGCCGCGGGCCGCGCAGCTGGTGGACAAGGACAGCACCTTCCTCAGCACGCTGGAGC ACCACCTGAGCCGCTACCTGAAGGACGTGAAGCAGCACCACGTCAAGGCTGACAAGCGGGAC CCAGAGTTTGTCTTCTACGACCAGCTGAAGCAAGTGATGAATGCGTACAGAGTCAAGCCGGC CGTCTTTGACCTGCTCCTGGCCGTTGGCATTGCTGCCTACCTCGGCATGGCCTACGTGGCTG TCCAGCACTTCAGCCTCCTCTACAGGACCGTCCAGAGGCTGCTCGTGAAGGCCAAGACACAG TGACACAGCCACCCCCACAGCCGGAGCCCCCGCCGCTCCACAGTCCCTGGGGCCGAGCACGA GTGAGTGGACACTGCCCCGCCGCGGGCGGCCCTGCAGGGACAGGGGCCCTCTCCCTCCCCGG CGGTGGTTGGAACACTGAATTACAGAGCTTTTTTCTGTTGCTCTCCGAGACTGGGGGGGGAT TGTTTCTTCTTTTCCTTGTCTTTGAACTTCCTTGGAGGAGAGCTTGGGAGACGTCCCGGGGC CAGGCTACGGACTTGCGGACGAGCCCCCCAGTCCTGGGAGCCGGCCGCCCTCGGTCTGGTGT AAGCACACATGCACGATTAAAGAGGAGACGCCGGGACCCCCTGCCCGATCGCGCGCGGCCTC CGCCCACCGCCTCCTGCCGCAAGGGGCCTGGACTGCAGGCCTGACCTGCTCCCTGCTCCGTG TCTGTCCTAGGACGTCCCCTCCCGCTCCCCGATGGTGGCGTGGACATGGTTATTTATCTCTG CTCCTTCTTGCCTGGAGGAGGGCAGTGCCAGCCCTGGGGTTCTGGGATTCCAGCCCTCCTGG AGCCTTTTGTTCCCCATGTGGTCTCAGTGACCCGTCCCCCTGACAGTGGGCTCGGGGAGCTG CATCACCCAGCCTTCCCCTTCTCCGACTGCAGGGTCTGATGTCATCGTTGACAGCCTTTGCT TCGTGGGGGCCTGGCAGGGCCCCTGCCTCCCCGACCCCCGACCCACTGCAAACCCCCGTTCC CCTGCACTCCTCTTCTCCCAGCCCATCCCTCCGGCCCCTGTGCCTCTGCGGCCCCAGCCCAG CTCCCAGGGCCGTCACCTGCTTGGCCCTGGCCCAGCTCCCTGCCCTGAGTCCTGAGCCAGTG CCTGGTGTTTCCTGGGCTCGGTACTGGGCCCCCAGGCCATCCAGGCTTTGCCACGGCCAGTT GGTCCTCCCTGGGGAACTGGGTGCGGGTGGAGTACTGGGAGGCAGGAGGTGGCCCGGGGAGG CCTTGTGGCTCCTCCCCTCGCTCCTCGCCCTGGGCCTCAGCTTCCTCATCAATAGAAAGGAT GTGTTCGGGGTGGGGGCGTCAGGTGAGAACGTTTGCTGGGAAGGAGAGGACTTGGGGCATGG CCTCTGGGGCCACCCTTCCTGGAACTCAGAGAGGAAGGTCCGGGCCCTCGGGAAGCCTTGGA CAGAACCCTCCACCCCGCAGACCAGGCGTCGTGTGTGTGTGGGAGAGAAGGAGGCCCGTGTT GAGCTCAGGGAGACCCCGGTGTGTCCGTTCTTTAGCAATATAACCTACCCAGTGCGTGCCGA GCAGGCTTGGTGGGGAAGGGACTTGAGCTGGGCAAGTCCTGGCCTGGCACCCGCAGCCGTCT CCCTTCCGTGGCCCAGGGAGGTGTTTGCTGTCCGAAGGACCTGGGCCGGCCCATGGGAGCCT GGGGTTCTGTCCAGATAGGACCAGGGGGTCTCACTTTGGCCACCAGTTCTTCGGCCAGCACC TCTGCCCTCCAGAACCTGCAGCCTGGAGGGGTGAGGGGACAACCACCCCTCTTTCCTCCAGG TTGGCAGGGGACCCTCTTCTCCCGTCTGCCCTGTGGGTTGCCCGCCTCCTCCAGAGACTTGC CCAAGGGCCCATCACCACTGGCCTCTGGGCACTTGTGCTGAGACTCTGGGACCCAGGCAGCT GCCACCTTGTCACCATGAGAGAATTTGGGGAGTGCTTGCATGCTAGCCAGCAGGCTCCTGTC TGGGTGCCACGGGGCCAGCATTTTGGAGGGAGCTTCCTTCCTTCCTTCCTGGACAGGTCGTC AGGATGGATGCACTGACTGACCGTCTGGGGCTCAGGCTGGTGTGGGATGCAGCCGGCCGATG AGAAAATAAAGCCATATTGAATGATAAAAAAAAAAAAAAAAAAA (SEQ ID NO: 49)
RefSeq Loq56926 Amino Acid Sequence
MLKASCLPLGFIVFLPAVLLLVAPPLPAADAAHEFTVYRMQQYDLQGQPYGTRNAVLNTEAR TIYIAAEVLSRRCVLMRLLDFSYEQYQKALRQSAGAWIILPRAMAAVPQDVWQFMEIEPEML AMETAVPVYFAVEDEALLSIYKQTQAASASQGSASAAEVLLRTATANGFQMVTSGVQSKAVS D LIASVEGRLTGLGGEDLPTIVIVAHYDAFGVAPWLSLGADSNGSGVSVLLELARLFSRLY TYKRTHAAYNLLFFASGGGKFNYQGTKR LEDNLDHTDSSLLQDNVAFVLCLDTVGRGSSLH LHVSKPPREGTLQHAFLRELETVAAHQFPEVRFSMVHKRINLAEDVLAWEHERFAIRRLPAF TLSHLESHRDGQRSSIMDVRSRVDSKTLTRNTRIIAEALTRVIYNLTEKGTPPDMPVFTEQM QIQQEQLDSVMDWLTNQPRAAQLVDKDSTFLSTLEHHLSRYLKDVKQHHVKADKRDPEFVFY DQLKQVMNAYRVKPAVFDLLLAVGIAAYLGMAYVAVQHFSLLYRTVQRLLVKAKTQ (SEQ ID NO: 50) The RefSeq Loq56926 protein has a transmembrane domain as predicted by SOSUI and TmPred. It also has both a signal peptide and a transmembrane domain predicted by SMART, suggesting that this is a type I membrane protein with the majority ofthe protein being extracellular.
The expression of Loc56926 in normal and malignant human tissues was further investigated by PCR experiments using commercially available human cDNA panels and cDNA samples prepared in-house from human tissues and cell lines. See Figures 9A-9B, 10A-10B, 11 A- 1 IB, and 12A-12B. Expression of Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was measured in these experiments as a control for cDNA integrity. GAPDH is a housekeeping gene expressed abundantly in all human tissues. The following primers were used to amplify a 482 base pair product ofthe GAPDH gene:
5 ' ACCACAGTCCATGCCATCAC 3 ' (SEQ ID NO:62) 5 ' TCCACCACCCTGTTGCTGTA 3 ' (SEQ ID NO:63)
For expression studies, malignant colon samples were obtained from Analytical Pathology Medical Group and frozen within thirty minutes of surgery. The HCTl 16 colon cancer cell line was obtained from American Type Culture Collection (ATCC of Manassas, Virginia.). RNA was extracted from the samples using RNEASY® Maxi Kit (Qiagen #75162) or from fresh HCT 116 cells using the RNEASY® Mini kit (Qiagen, #74104) according to the manufacture's instractions and reverse transcribed into cDNA using SUPERSCRIPT® II Kit (Invifrogen # 12371-019). The positive confrol for Loc56926 IMAGE clone 4428206 was obtained from the ATCC. Primers used to amplify a 283 base pair product of Loc56926 were: 5 AATGCAGTGCTGAACACGGAG 3 ' (SEQ ID NO:64)
5 ' TCTGCTTGTAGATAGACAGCAGG 3 ' (SEQ ID NO:65)
AW779536
In a comparison of malignant colon samples containing greater than 50% malignant cells in the sample against mixed normal tissues, fragment AW779536 was upregulated 3.7 fold. E- Northern analysis shown in Figure 13 demonstrates that the fragment is expressed in 77% of the tumors and poorly expressed in normal tissue. AW779536 Nucleotide Sequence
TTCTTCCTGTGTTACAATTACCCTGTTTCTGATTACTACAGCCCAACCCGGGCGGACACCAC
CACCATTCTGGCTGCCGGGGCTGGAGTGACCATAGGATTCTGGATCAACCATTTCTTCCAGC
TTGTATCCAAGCCCGCTGAATCTCTCCCTGTTATTCAGAACATCCCACCGNTCACCACCTAC ATGTTAGNTTTGGGTCTGACCAAATTTGCAGTGGGAATTGTGTTGATCCTCTTGGTTCGTCA
GCTTGTACAAAATCTCTCACTGCAAGTATTATACTCATGGTTCNAGGTNGGTCNCCAGGAAC
AAGGAGGCCAGGCGGAGACTGGAGATTGAAGTGCCTTACAAGTTTGTTACCTACACATCTGT
TGGCATCTGCGCTACAACCTTTGTGCCGATGCTTCACAGGTTTCTGGGATTACCCTGAGTCT
CAAACAGTTGGAAACTAGCCCACTGGACATGAAAGCCAAGACATAGGAAAGTTATTGGTAGG CAAATCTTGACAACTTATTTTTCTTTAACAACAACAAAAAGTCATACGGCTGTCTTGCTACT
(SEQ ID NO: 41)
BLAST searching with this sequence revealed a hypothetical protein predicted by Acembly, Ensembl and Fgenesh++, Hs2_5283_28_l_l 143.b with the following nucleotide sequence:
Hs2_5283_28_l_l 143.b Nucleotide Sequence
GCTTATGTACAGAAGTACGTCGTGAAGAATTATTTCTACTATTACCTATT CCAATTTTCAGCTGCTTTGGGCCAAGAAGTGTTCTACATCACGTTTCTTC Cattcactcactggaatattgacccttatttatccagaagattgatcatcatatgggttttg gtgatgtatattggccaagtggccaaggatgtcttgaagtggccccgtccctcctcccctcc agttgtaaaactggaaaagagactgatcgctgaatatggaatgccatccacccacgccatgg cggccactgccattgccttcaccctccttatctctactatggacagataccagtatccattt gtgttgggactggtgatggccgtggtgttttccaccttggtgtgtctcagcaggctctacac tgggatgcatacggtcctggatgtgctgggtggcgtcctgatcaccgcactcctcatcgtcc tcacctaccctgcctggaccttcatcgactgcctggactcggccagccccctcttccccgtg tgtgtcatagttgtgccattcttcctgtgttacaattaccctgtttctgattactacagccc aacccgggcggacaccaccaccattctggctgccggggctggagtgaccataggattctgga tcaaccatttcttccagcttgtatccaagcccgctgaatctctccctgttattcagaacatc ccaccactcaccacctacatgttagttttgggtctgaccaaatttgcagtgggaattgtgtt gatcctcttggttcgtcagcttgtacaaaatctctcactgcaagtattatactcatggt ca aggtggtcaccaggaacaaggaggccaggcggagactggagattgaagtgccttacaagttt gttacctacacatctgttggcatctgcgctacaacctttgtgccgatgcttcacaggtttct gggattaccctgagtctcaaacagttggaaactagcccactggacatgaaagccaagacata ggaaagttattggtaggcaaatcttgacaacttatttttctttaacaacaacaaaaagtcat acggctgtcttgctactaccagataaatgatgctgctgtgtgaaaggaagaactgtctcata gcggtcattggtcgtccgtggtggttggttgtgctacagttgaacccaggctaaagaccata atccggatctttaaaggcacacaccgcgccccccccccccccgcccggcccctgctcctctc gctgttgcacgggctttggatctagtcatgggctggcaggaattgtggcctggcttaggaat agctatgagccccactgggttctggagagccagtagagatggggtgatctgggaggctggag gtagagcctttcttttccgttacaaccttgcctagcatggagttaactgtgcctggttgggt ggtaagatcactctgaaagaaagctcactgtgaagagatgaaaggtggaggcagagctgtga ggtcatggggaaaagcctgctttccttataagtcctgctgttcatgttggaataaggatctg ctcttccttgtttccatgcattttgcaggattccaggtaccattaccacactcttctgaccc atgaaaccaactggctgctcacacatcaccaaacaggttgggggttagccttcagcacaggt ggatacatctgggattcactgagattcctgccctctcctgcttcctagtggtttgggacagg ccctctgcccatcgtcagcagttttttgctttcatacaaacctggaaggcactggcatctgc ctaggaaagtggatctgtgaagaacagatgaactcaatcctttctggagtctgacaaagaag ggataggcttccttgacattgcctgtcctgacaaggcctccctgacattactcctccaattt cacagttaccttctgtaaatctattttctcatctactgaatagaatcaggcgccctttttgt cttcccacctcttatctcttggcaattttaaggggaattaatgcaagaacaactttagtgtc tcttgggaaaacaagccaaccaaatacaaaacccattaagcctactagggtgagtcctctta acatgggaaggcgatgattatgcaaacaccggagttccctcctcttcagttcctaagaataa agaacaggtatcaagaactttctttaaagttagtgtaactatagttaacaaagtatccattg aagtttagtgcctgtaggactgagccagtgctttatcaacccaacacatcatcaccatgtgc atactctagaaaaaaaaatagcttccttaaaagttacagaggctcttaacgtgttaaaaccg aaaaatcacatttttcttgatttcaaatatgttctacggccttactgttgggatgatattta gtatgtaacttagcattccaatttctcaagaatttttaggccgggtgcggtggctcatgcct gtaatcccagcactttgggaggccgaggtgggcggaccacgaggtcaggagatcgagaccat cctggctaacacggtaccccgtctctactgaaaatacaaaaaaattagccggacgtggtgga gggcgcctgtagtcccagctactcaggaggctgaggcaggagaatggcgtgaacccggtgag cggagcttgcagtgagccgagattgcgccactgcactccagcctgggcgacagagcgagact ctctcaaaaaaaaaaaaaaagaatttttagcaaaacatcctgtttttacttaaaattcttct catatttattatagttagaaggcaaagatcaagatgacctgccgtttgactgcttttacatc aaactctgcccagtatttgcagcacaactcaggggaagggccttagcttacaggtactccca gccttcatctgcccctgcagagcagtggctgtcagccggatgcggcacttttctgtattttc atccacacagctgcccagccagagttcgcaacactggatatttacaccaaataattgtggtt gacttgtctgaagccagctgacaaaaggatcagcttttcccacttgtattttttaaaaagag ggattgtgatcattgtcacagagtgggtgctggcctctcatatatatgatatatatatatca ttttatatatatatatatatcatatacataatttttactgctgtctctagttttaagtccca acaataggaaggccgatcagctatattgatatatttaaggctgtacttaactaatttgggct gaggatgaatatatcagccacagcacattaaagaatgagccaaggatttgtcatggttggtc actttttaaagtatttgattactgcaactggagaatgaaaagtgtatattggtgacgccaac ctcagtttctgagcactcctgctctgtggtgagaatcagacaaaaattcatcggggtgaaaa aggcattacctgattcacacccttgtcttgctagccctcttccattcatttctcacacagca ctttgctctgttaaatcctctctctgtctcagaccattgcttgccccttcaaagggtatggt tcaggctcctttcaagacatttggagtttctctctggggaaagagagccccctactggtttg gcttcagtctaggtccaccatccctctcgatctggcatcttggagattaatttaaaaggcaa gctcaccacaatgtaagcctatggtctggccaaccttgcttttgggaactgtgacaccaaag cccccaggactatctgcctctccaggagccagatagaatgacatgcctttttcctaattgtc cacattccacccccaacccactgccactgtgggccaagccatccatcttgcaatcttcatct aaaacagctctcatttcatgccagttttgctcaaacctgcaccgtcacaagatattcagaag atgaaaacgtagaagacacccctgaattaaaaacacttacatagcagtggctggaattactc caaaacgtgcccagtgatcgcactgtaacatgggattttctcacccaaataggcaactcatg cttcctgagtgtaatcaaagcatgtggtgttttggggccatatgcaccaggtttctatttta gaaaccttcagctgtcttgcttatgtactgtatgtaaatttattctttttaaaaatcacttt tatttgattttgacttattaaatgctttaaaagccag(SEQ ID NO: 42)
The amino acid sequence of Hs2_5283_28_l_l 143.b is set forth below:
Hs2_5283_28_l_1143.b Amino Acid Sequence
AYVQKYWKNYFYYYLFQFSAALGQEVFYITFLPFTHWNIDPYLSRRLII I VLVMYIGQVAKDVLK PRPSSPPWKLEKRLIAEYGMPSTHAMAATAI AFTLLISTMDRYQYPFVLGLVMAWFSTLVCLSRLYTGMHTVLDVLGGVL ITALLIVLTYPA TFIDCLDSASPLFPVCVIWPFFLCYNYPVSDYYSPT RADTTTILAAGAGVTIGFWINHFFQLVSKPAESLPVIQNIPPLTTYMLVL GLTKFAVGIVLILLVRQLVQNLSLQVLYS FKWTRNKEARRRLEIEVPY KFVTYTSVGICATTFVPMLHRFLGLP ( SEQ ID NO : 43 )
This amino acid sequence is predicted to contain 9 transmembrane domains by SMART and TmPred and 8 transmembrane domains by SOSUI. By contrast, when analyzed by use ofthe GENEID™ program, the following gene is identified as being overexpressed in colon tissue:
chr2_2054 Nucleotide Sequence
ATGGCGGCCACTGCCATTGCCTTCACCCTCCTTATCTCTACTATGGACAG ATACCAGTATCCATTTGTGTTGGGACTGGTGATGGCCGTGGTGTTTTCCA CCTTGGTGTGTCTCAGCAGGCTCTACACTGGGATGCATACGGTCCTGGAT GTGCTGGGTGGCGTCCTGATCACCGCACTCCTCATCGTCCTCACCTACCC TGCCTGGACCTTCATCGACTGCCTGGACTCGGCCAGCCCCCTCTTCCCCG TGTGTGTCATAGTTGTGCCATTCTTCCTGTGTTACAATTACCCTGTTTCT GATTACTACAGCCCAACCCGGGCGGACACCACCACCATTCTGGCTGCCGG GGCTGGAGTGACCATAGGATTCTGGATCAACCATTTCTTCCAGCTTGTAT CCAAGCCCGCTGAATCTCTCCCTGTTATTCAGAACATCCCACCACTCACC ACCTACATGTTAGTTTTGGGTCTGACCAAATTTGCAGTGGGAATTGTGTT GATCCTCTTGGTTCGTCAGCTTGTACAAAATCTCTCACTGCAAGTATTAT ACTCATGGTTCAAGGTGGTCACCAGGAACAAGGAGGCCAGGCGGAGACTG GAGATTGAAGTGCCTTACAAGTTTGTTACCTACACATCTGTTGGCATCTG CGCTACAACCTTTGTGCCGATGCTTCACAGGTTTCTGGGATTACCCTGA (SEQ ID NO: 44)
This gene encodes a protein having the following predicted stracture:
chr2_2054 Amino Acid Sequence
MAATAIAFTLLISTMDRYQYPFVLGLVMAWFSTLVCLSRLYTGMHTVLD VLGGVLITALLIVLTYPAWTFIDCLDSASPLFPVCVIWPFFLCYNYPVS DYYSPTRADTTTILAAGAGVTIGFWINHFFQLVSKPAESLPVIQNIPPLT TYMLVLGLTKFAVGIVLILLVRQLVQNLSLQVLYSWFKWTRNKEARRRL EIEVPYKFVTYTSVGICATTFVPMLHRFLGLP* (SEQ ID NO: 45) When this sequence is analyzed by SOSUI and TmPred it is predicted to possess 7 transmembrane domains. By contrast, analyses by SMART suggests that the protein has 5 transmembrane domains and a signal sequence. These analyses also indicate that the protein contains a PFAM domain indicating that the protein contains an acid phosphatase domain.
AL531683
In a comparison of malignant colon samples with greater than 50% malignant cells in the sample against mixed normal tissues, fragment AL531683 was found to be upregulated 3.76- fold. The E-Northem analysis shown in Figure 14 demonstrates that the fragment is expressed in 100% ofthe tumors analyzed and poorly expressed in normal tissue.
AL53168 Nucleotide Sequence
CGCCGGCGGTGCGTGTGGGAAGGCGTGGGGTGCGGACCCCGGCCCGACCTCNCCGTCCCGCC
CGCCGCCTTCTGCGTCGCGGGNGCGGGCCGGCGGGGTCCTCTGACGCGGCAGACAGNCCCTC GCTGTCGCCTCCAGTGGTTGTCGACTTGCGGGCGGCCCCCCTCCGCGGCGGTGGGGGTGCCG TCCCGCCGGCCCGTCGTGCTGCCCTCTCNNGGGGGGTTTGCGCGAGCGTCGGCTCCGCCTGG GCCCTTGCGGTGCTCCTGGAGCGCTCCGGGTTGTCCCTCAGGTGCCCGAGGCCGAACGGTGG TGTGTCGTTCCCGCCCCCGGCGCCCCCTCCTCCGGTCGCCGCCGCGGTGTCCGCGCGTGGGT CCTGAGGGAGCTCGTCGGTGTGGGGTTCGAGGCGGTTTGAGTGAGACGAGACGAGAC (SEQ ID NO : 46 )
AI202201
In a comparison of malignant colon samples with greater than 50% malignant cells in the sample against mixed normal tissues, fragment AI202201 was upregulated 3.18-fold. E- Northern analysis shown in Figure 15 demonstrates that the fragment is expressed in 77% of the tumors and poorly expressed in normal tissue.
AI202201 Nucleotide Sequence ACCCTATAGCTCCTTACGCTGGGAAAGCTGGTTTTTTAAAAAAATAATAATAAAA TATTTAATCTTATTAAGTGTTCATTTAAAATGCGTAATGCTTTGGAAATAATGGGTAACAGA TAGCGAGAGGATATGTTTATAAAGTGAGCATGTTGGTCCCATTTATAAATATATGTATGATT TATAAGCTTTTTTAAAACAAAGCTCAAATTGTTGGTATTTTTCTAAAATGTGCACAGCTGTA TTTTACATGAAGGCTCTTTCTAATGGGTTGTTATACTGTACTCAACATTTTGGACAGCACAT GAAGTCTGCCAATGTACTTAATAAAACATGACTTTGTTTATTTAAAGTTTCTTGCTGTGAAA AAGAACTCCCTACCTGTGAGTTCCTTTATTTATAATTCTTGAAACCAAAATGTATAATGTAC AGTTTTCACAACTGTATCTGCTCTAATA (SEQ ID NO: 47)
AL389942
In a comparison of malignant colon samples with greater than 50% malignant cells in the sample against mixed normal tissues, fragment AL389942 was upregulated 3.83-fold. E- Northern analysis shown in Figure 16 demonstrates that the fragment is expressed in 55% of the tumors and poorly expressed in normal tissue.
AL389942 Nucleotide Sequence
GAAGCTCCAAATGCTCTGGGTTTCAGCTCCTCTGTGCTGTGGACNCTGACTTTGGCTCAGAA
CTCCGATTTAGTACAAAAGGCTCATTTTTATTTCAGGGGCACTCTTCCTAAAGCAAACCTAA TAAATGAAATATGGAATTCACAGATACACACACACATTAAAAAATTAACCTAGTGTATCTGT GAGGAGTAGGCAGAAATTCNCTGTATAAAAGAATGCTTCATTTCATAGAGAATTTGTGTTAA GATTCCATTAGATAGTACATTTCTCAAAGATTTTTGAGGTTGTATTTGCTTTACCAAAACTT GGTTTATGTAAGTGGAAAAAGCATGTTGCAAAATAACTTGGTGTCTATGATTCAGTTTATGT AAAATAATAAATGTATGTAGGAATACGTGTGTTGAAAGATGTACATCAATTTGCTAACAATG GTTATCTCTGACGTGGTGGGATTTGAGATGTGTTTTTCTTTTTGGTTGTATTTTTCTCTATT GTTTGACTTA (SEQ ID NO: 48)
EXAMPLE 5 Identification Of Gene Unregulated In Colon Cancer
Using the GeneLogic database and the methods described generally in Example 2, the following additional DNA sequences were identified as being overexpressed in colon tumor tissue:
DNA fragment NM_021246 is 5 -fold upregulated as shown by hybridization in the malignant colon when compared with mixed normal samples, greater than 3 -fold upregulated compared with normal kidney, liver and lung, and greater than 2-fold upregulated in all other tissues. NM_021246 Nucleotide Sequence
AACCGAATGCGGTGCTACAACTGTGGTGGAAGCCCCAGCAGTTCTTGCAAAGAGGCCGTGAC CACCTGTGGCGAGGGCAGACCCCAGCCAGGCCTGGAACAGATCAAGCTACCTGGAAACCCCC CAGTGACCTTGATTCACCAACATCCAGCCTGCGTCGCAGCCCATCATTGCAATCAAGTGGAG ACAGAGTCGGTGGGAGACGTGACTTATCCAGCCCACAGGGACTGCTACCTGGGAGACCTGTG CAACAGCGCCGTGGCAAGCCATGTGGCCCCTGCAGGCATTTTGGCTGCAGCAGCTACCGCCC TGACCTGTCTCTTGCCAGGACTGTGGAGCGGATAGGGGGAGTAGGAGTAGAGAAGGGAACAA GGGAGCAAGGGAACAAGGGACATCTGAACATCT (SEQ ID NO: 56)
The E-nothem results in Figure 17 indicate that this fragment is upregulated in colon and rectal malignancies. Accordingly, this gene can be targeted for the treatment of colon or rectal cancer. A search of commercial databases reveals that NM_021246 is apparently part the Ly6G6D gene set forth below:
Ly6G6D mRNA Sequence cccatggcagtcttattcctcctcctgttcctatgtggaactccccaggc tgcagacaacatgcaggccatctatgtggccttgggggaggcagtagagc tgccatgtccctcaccacctactctacatggggacgaacacctgtcatgg ttctgcagccctgcagcaggctccttcaccaccctggtagcccaagtcca agtgggcaggccagccccagaccctggaaaaccaggaagggaatccaggc tcagactgctggggaactattctttgtggttggagggatccaaagaggaa gatgccgggcggtactggtgcgctgtgctaggtcagcaccacaactacca gaactggagggtgtacgacgtcttggtgctcaaaggatcccagttatctg caagggctgcagatggatccccctgcaatgtcctcctgtgctctgtggtc cccagcagacgcatggactctgtgacctggcaggaagggaagggtcccgt gaggggccgtgttcagtccttctggggcagtgaggctgccctgctcttgg tgtgtcctggggaggggctttctgagcccaggagccgaagaccaagaatc atccgctgcctcatgactcacaacaaaggggtcagctttagcctggcagc ctccatcgatgcttctcctgccctctgtgccccttccacgggctgggaca tgccttggattctgatgctgctgctcacaatgggccagggagttgtcatc ctggccctcagcatcgtgctctggaggcagagggtccgtggggctccagg cagaggaaaccgaatgcggtgctacaactgtggtggaagccccagcagtt cttgcaaagaggccgtgaccacctgtggcgagggcagaccccagccaggc ctggaacagatcaagctacctggaaaccccccagtgaccttgattcacca acatccagcctgcgtcgcagcccatcattgcaatcaagtggagacagagt cggtgggagacgtgacttatccagcccacagggactgctacctgggagac ctgtgcaacagcgccgtggcaagccatgtggcccctgcaggcattttggc tgcagcagctaccgccctgacctgtctcttgccaggactgtggagcggat agggggagtaggagtagagaagggaacaagggagcaagggaacaagggac atctgaacatctaatgtgagaagagaaacatccttctgtgagtcattaaa atctatgaaccactct (SEQ ID NO: 57)
The amino acid sequence for Ly6G6D is set forth below:
Ly6G6D Amino Acid Sequence
MAVLFLLLFLCGTPQAADNMQAIYVALGEAVELPCPSPPTLHGDEHLSWF CSPAAGSFTTLVAQVQVGRPAPDPGKPGRESRLRLLGNYSLWLEGSKEED AGRY CAVLGQHHNYQNWRVYDVLVLKGSQLSARAADGSPCNVLLCSWP SRRMDSVT QEGKGPVRGRVQSFWGSEAALLLVCPGEGLSEPRSRRPRII RCLMTHNKGVSFSLAASIDASPALCAPSTGWDMP ILMLLLTMGQGWIL ALSIVL RQRVRGAPGRGNRMRCYNCGGSPSSSCKEAVTTCGEGRPQPGL EQIKLPGNPPVTLIHQHPACVAAHHCNQVETESVGDVTYPAHRDCYLGDL CNSAVASPΪVAPAGILAAAATALTCLLPGLWSG(SEQ ID NO: 58)
Analysis ofthe Ly6G6D protein sequence using the SMART program identified two potential transmembrane domains and an Ig domain, suggesting that this protein is a cell surface protein.
EXAMPLE 6 Identification of Colon-Cancer Associated Gene AI821606 FLJ32334 Fragment AI821606 set forth below, also was shown to be upregulated in colon, pancreas and rectal malignancies. This is supported by the E-Northem results in Figure 18. AI821606 Nucleotide Sequence
TTCCTCGGAGGGGCCGTGGTGAGTCTCCAGTATGTTCGGCCCAGCGCTCTTCGCACCCTTCT GGACCAAAGCGCCAAGGACTGCAGCCAGGAGAGAGGGGGCTCACCTCTTATCCTCGGCGACC CACTGCACAAGCAGGCCGCTCTCCCAGACTTAAAATGTATCACCACTAACCTGTGAGGGGGA CCCAATCTGGACTCCTTCCCCGCCTTGGGACATCGCAGGCCGGGAAGCAGTGCCCGCCAGGC CTGGGCCAGGAGAGCTCCAGGAAGGGCACTGAGCGCTGCTGGCGCGAGGCCTCGGACATCCG CAGGCACCAGGGAAAGTCTCCTGGGGCGATCTGTAAAT (SEQ ID NO: 51)
A database search revealed that AI821606 is in the 3 'UTR of predicted genes corresponding to both strands of a chromosome. Based thereon, this fragment could be part ofthe following genes:
ENST00000267803 Nucleotide Sequence gcttccagcggacggcagcgcgcgagcattgccccccctgcaccacctca ccaagATGGCTACTTTGGGACACACATTCCCCTTCTATGCTGGCCCCAAG CCAACCTTCCCGATGGACACCACTTTGGCCAGCATCATCATGATCTTTCT GACTGCACTGGCCACGTTCATCGTCATCCTGCCTGGCATTCGGGGAAAGA CGAGGCTGTTCTGGCTGCTTCGGGTGGTGACCAGCTTATTCATCGGGGCT GCAATCCTGGGGACCCCCGTGCAGCAGCTGAATGAGACCATCAATTACAA CGAGGAGTTCACCTGGCGCCTGGGTGAGAACTATGCTGAGGAGTATGCAA AGGCTCTGGAGAAGGGGCTGCCAGACCCTGTGTTGTACCTAGCTGAGAAG TTCACTCCAAGAAGCCCATGTGGCCTATACCGCCAGTACCGCCTGGCGGG ACACTACACCTCAGCCATGCTATGGGTGGCATTCCTCTGCTGGCTGCTGG CCAATGTGATGCTCTCCATGCCTGTGCTGGTATATGGTGGCTACATGCTA TTGGCCACGGGCATCTTCCAGCTGTTGGCTCTGCTCTTCTTCTCCATGGC CACATCACTCACCTCACCCTGTCCCCTGCACCTGGGCGCTTCTGTGCTGC ATACTCACCATGGGCCTGCCTTCTGGATCACATTGACCACAGGACTGCTG TGTGTGCTGCTGGGCCTGGCTATGGCGGTGGCCCACAGGATGCAGCCTCA CAGGCTGAAGGCTTTCTTCAACCAGAGTGTGGATGAAGACCCCATGCTGG AGTGGAGTCCTGAGGAAGGTGGACTCCTGAGCCCCCGCTACCGGTCCATG GCTGACAGTCCCAAGTCCCAGGACATTCCCCTGTCAGAGGCTTCCTCCAC CAAGGCATACTGTAAGGAGGCACACCCCAAAGATCCTGATTGTGCTTTAt aacattcctccccgtggaggccacctggacttccagtctggctccaaacc tcattggcgccccataaaaccagcagaactgccctcagggtggctgttac cagacacccagcaccaatctacagacggagtagaaaaaggaggctctata tactgatgttaaaaaacaaaacaaaacaaaaagccctaagggactgaaga gatgctgggcctgtccataaagcctgttgccatgataaggccaagcaggg gctagcttatctgcacagcaacccagcctttccgtgctgccttgcctctt caagatgctattcactgaaacctaacttcacccccataacaccagcaggg tgggggttacatatgattctcctatggtttcctctcatccctcggcacct cttgttttcctttttcctgggttccttttgttcttcctttacttctccag cttgtgtggccttttggtacaatgaaagacagcactggaaaggaggggaa accaaacttctcatcctaggtctaacattaaccaactatgccacattctc tttgagcttcagttcccaaatttgctacataagattgcaagacttgccaa gaatcttgggatttatctttctatgccttgctgacacctaccttggccct caaacaccacctcacaagaagccaggtgggaagttagggaatcaactcca aaacgctattccttcccaccccactcagctgggctagctgagtggcatcc aggacgggggagtgggtgacctgcctcatcactgccacctaacgtccccc tggggtggttcagaaagatgctagctctggtagggtccctccggcctcac tagagggcgcccctattactctggagtcgacgcagagaatcaggtttcac agcactgcggagagtgtactaggctgtctccagcccagcgaagctcatga ggacgtgcgaccccggcgcggagaagccatgaaaattaatgggaaaaaca gtttttaaaaaacaaaagaaaaaaaggtttatttacagatcgccccagga gactttccctggtgcctgcggatgtccgaggcctcgcgccagcagcgctc agtgcccttcctggagctctcctggcccaggcctggcgggcactgcttcc cggcctgcgatgtcccaaggcggggaaggagtccagattgggtccccctc acaggttagtggtgatacattttaagtctgggagagcggcctgcttgtgc agtgggtcgccgaggataagaggtgagccccctctctcctggctgcagtc cttggcgctttggtccagaagggtgcgaagagcgctgggccgaacatact ggagactcaccacggcccctccgaggaagaggcacaggacgcctgtggcg gtggggatcgaaagaaaggagggcatgtggagtcagggctatgttgccca ggctggtctcgaactctggcctcaaacgaccttcctgcctcgacctccca aagtgctgggattacaggcgtgatgcccgggccttcttccatcttttgga gcctaccccttgtgttacctcccgccacacacctctaatctgaattacat gaaacacggcaagacaccaaacccttctgagccccccacttttcatctgt aaaatggtcataacagtgcctgtttctgcgaactattgagaggggcaaat agggtaatagatgtgaattcattctgtaaactgg (SEQ ID NO: 52)
The predicted coding sequence for ENST00000267803 is set forth below:
ENST00000267803 Amino Acid Sequence
MATLGHTFPFYAGPKPTFPMDTTLASIIMIFLTALATFIVILPGIRGKTR LF LLRWTSLFIGAAILGTPVQQLNETINYNEEFT RLGENYAEEYAKA LEKGLPDPVLYLAEKFTPRSPCGLYRQYRLAGHYTSAML VAFLCWLLAN VMLSMPVLVYGGYMLLATGIFQLLALLFFSMATSLTSPCPLHLGASVLHT HHGPAFWITLTTGLLCVLLGLAMAVAHRMQPHRLKAFFNQSVDEDPMLE SPEEGGLLSPRYRS ADSPKSQDIPLSEASSTKAYCKEAHPKDPDCAL (SEQ ID NO: 53)
SMART analysis predicted that the protein contains several transmembrane domains (rectangles) and a signal sequence, as depicted schematically below:
100 200
Figure imgf000093_0001
Based on a sequence contained on the opposite strand ofthe chromosome, the following gene sequence is predicted:
chrl5.41.013.a Nucleotide Sequence
ATGACCCTGTGGAACGGCGTACTGCCTTTTTACCCCCAGCCCCGGCATGC CGCAGGCTTCAGCGTTCCACTGCTCATCGTTATTCTAGTGTTTTTGGCTC TAGCAGCAAGCTTCCTGCTCATCTTGCCGGGGATCCGTGGCCACTCGCGC TGGTTTTGGTTGGTGAGAGTTCTTCTCAGTCTGTTCATAGGCGCAGAAAT TGTGGCTGTGCACTTCAGTGCAGAATGGTTCGTGGGTACAGTGAACACCA ACACATCCTACAAAGCCTTCAGCGCAGCGCGCGTTACAGCCCGTGTCCGT CTGCTCGTGGGCCTGGAGGGCATTAATATTACACTCACAGGGACCCCAGT GCATCAGCTGAACGAGACCATTGACTACAACGAGCAGTTCACCTGGCGTC TGAAAGAGAATTACGCCGCGGAGTACGCGAACGCACTGGAGAAGGGGCTG CCGGACCCAGTGCTCTACCTGGCGGAGAAGTTCACACCGAGTAGCCCTTG CGGCCTGTACCACCAGTACCACCTGGCGGGACACTACGCCTCGGCCACGC TATGGGTGGCGTTCTGCTTCTGGCTCCTCTCCAACGTGCTGCTCTCCACG CCGGCCCCGCTCTACGGAGGCCTGGCACTGCTGACCACCGGAGCCTTCGC GCTCTTCGGGGTCTTCGCCTTGGCCTCCATCTCTAGCGTGCCGCTCTGCC CGCTCCGCCTAGGCTCCTCCGCGCTCACCACTCAGTACGGCGCCGCCTTC TGGGTCACGCTGGCAACCGGTGAGGACCGAGAGAATGGGCCCCGGGGGCT AAGGGTGGAGACAGGATTCACACCGGGCGTCCTGTGCCTCTTCCTCGGAG GGGCCGTGGCCGGGAAGCAGTGCCCGCCAGGCCTGGGCCAGGAGAGCTCC AGGAAGGGCACTGAGCGCTGCTGGCGCGAGGCCTCGGACATCCGCAGGCA CCAGGGAAAGTCTCCTGGGGCGATCTGTAAA (SEQ ID NO: 54)
This sequence is predicted to encode the following protein:
chrl5.41.013.a Amino Acid Sequence MTLNGVLPFYPQPRHAAGFSVPLLIVILVFLALAASFLLILPGIRGHSR WFWLVRVLLSLFIGAEIVAVHFSAE FVGTVNTNTSYKAFSAARVTARVR LLVGLEGINITLTGTPVHQLNETIDYNEQFTWRLKENYAAEYANALEKGL PDPVLYLAEKFTPSSPCGLYHQYHLAGHYASATLWVAFCFWLLSNVLLST PAPLYGGLALLTTGAFALFGVFALASISSVPLCPLRLGSSALTTQYGAAF VTLATGEDRENGPRGLRVETGFTPGVLCLFLGGAVAGKQCPPGLGQESS RKGTERC REASDIRRHQGKSPGAICK (SEQ ID NO: 55)
SMART analysis identified three transmembrane domains (rectangles) and a signal sequence. The predicted stracture ofthe protein is depicted schematically below:
100 200
Figure imgf000094_0001

Claims

What is claimed is:
1. An isolated nucleic acid that is expressed by human colon cancer cells comprising: (i) the nucleotide sequence of any one of SEQ ID NOs: 2, 4, 6, 8, 10, 13 and 15; (ii) a variant of (i), wherein said variant has a nucleotide sequence that is at least 70% identical to the sequence of (i) when aligned without allowing for gaps; or (iii) a fragment of (i) or (ii) having a size of at least 20 nucleotides in length.
2. The isolated nucleic acid of claim 1 which comprises the nucleotide sequence of any one of SEQ ID NOs: 2, 4, 6, 8, 10, 13 and 15, or a fragment thereof.
3. A primer mixture comprising primers that result in the specific amplification of any one ofthe nucleic acids identified in claim 1.
4. A method of detecting colon cancer comprising (i) obtaining a human colon cell sample; and (ii) determining whether such cell sample expresses a colon cancer gene having a nucleotide sequence of any one of SEQ ID NOs: 2, 4, 6, 8, 10, 13, 15, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 34, 36, 37, 39, 41, 42, 44, 46, 47, 48, 49, 51, 52, 54, 56, or 57.
5. The method of claim 4, wherein said method comprises detecting the expression of said colon cancer gene using a nucleic acid that specifically hybridizes thereto.
6. The method of claim 4, wherein said method comprises detecting the expression of said colon cancer gene using primers that result in the amplification thereof.
7. The method of claim 4, wherein the expression of said colon cancer gene is detected by performing an assay to detect the presence or level ofthe antigen encoded by said gene.
8. The method of claim 7, wherein said assay involves the use of a monoclonal antibody or fragment that specifically binds to said antigen.
9. The method of claim 8, wherein said assay comprises an ELISA or competitive binding assay.
10. An antigen expressed by human colon cancer cells comprising:
(i) an antigen encoded by the nucleic acid of any one of SEQ ID NOs: 2,
4, 6, 8, 10, 13, 15, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 34, 36, 37, 39, 41, 42, 44, 46, 47, 48, 49, 51, 52, 54, 56, or 57; (ii) an antigen having the amino acid sequence of any one of SEQ ID NO.
5, 7, 9, 11, 14, 16, 25, 32, 35, 38, 40, 43, 45, 50, 53, 55, or 58; or (iii) a fragment or variant of (i) or (ii), wherein said fragment or variant specifically binds the antigen of (i) or (ii).
11. A colon antigen encoded by the nucleic acid of any one of SEQ ID NOs: 2, 4, 6, 8, 10,
13, 15, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 34, 36, 37, 39, 41, 42, 44, 46, 47, 48, 49, 51, 52, 54, 56, or 57.
12. A monoclonal antibody or antigen-binding fragment thereof that specifically binds to a colon antigen of claim 11.
13. The antigen of claim 11 , further comprising a detectable label, wherein the detectable label is attached directly or indirectly to the antigen.
14. A colon antigen comprising the amino acid sequence of any one of SEQ ID NO. 5, 7,
9, 11, 14, 16, 25, 32, 35, 38, 40, 43, 45, 50, 53, 55, or 58.
15. A monoclonal antibody or antigen-binding fragment thereof that specifically binds to a colon antigen of claim 14.
16. The antigen of claim 14, further comprising a detectable label, wherein the detectable label is attached directly or indirectly to the antigen.
17. A diagnostic kit for detecting colon cancer which comprises a DNA according to claim 1 and a detectable label.
18. A diagnostic kit for detection of colon cancer which comprises primers according to claim 3 and a diagnostically acceptable carrier.
19. A diagnostic kit for detection of colon cancer which comprises a monoclonal antibody according to claim 12.
20. A diagnostic kit for detection of colon cancer which comprises a monoclonal antibody according to claim 15.
21. A method for treating colon cancer which comprises administering a therapeutically effective amount of a ribozyme or antisense oligonucleotide that inhibits the expression of a gene having a DNA sequence of any one of any one of SEQ ID NOs: 2, 4, 6, 8, 10, 13, 15, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 34, 36, 37, 39, 41, 42, 44, 46, 47, 48, 49, 51, 52, 54, 56, or 57, or fragment or variant thereof.
22. A method for treating colon cancer in a subject, which comprises administering to said subject a ligand that specifically binds to a nucleic acid molecule comprising a nucleotide sequence of any one of any one of SEQ ID NOs: 2, 4, 6, 8, 10, 13, 15, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 34, 36, 37, 39, 41, 42, 44, 46, 47, 48, 49, 51, 52, 54, 56, or 57, or fragment or variant thereof.
23. The method of claim 22, wherein said ligand further comprises and effector moiety.
24. The method of claim 23, wherein said effector moiety is a therapeutic radiolabel, enzyme, cytotoxin, growth factor, or drag.
25. A method for treating colon cancer comprising administering a therapeutically effective amount of:
(a) a colon antigen encoded by the nucleic acid of any one of SEQ ID NOs: 2, 4, 6, 8, 10, 13, 15, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 34, 36, 37, 39, 41, 42, 44, 46, 47, 48, 49, 51, 52, 54, 56, or 57; and
(b) an adjuvant, to thereby elicit a humoral or cytotoxic T-lymphocyte response to said antigen.
26. A method for treating colon cancer comprising administering a therapeutically effective amount of:
(a) a colon antigen comprising the amino acid sequence of any one of SEQ ID NO. 5, 7, 9, 11, 14, 16, 25, 32, 35, 38, 40, 43, 45, 50, 53, 55, or 58; and
(b) an adjuvant, to thereby elicit a humoral or cytotoxic T-lymphocyte response to said antigen.
27. A method for treating colon cancer comprising administering a therapeutically effective amount of a ligand which specifically binds to a protein encoded by a gene which includes the nucleic acid of any one of SEQ ID NOs: 2, 4, 6, 8, 10, 13, 15, 17, 18, 19, 20, 21, 22, 23, 24, 26, 27, 28, 29, 30, 31, 34, 36, 37, 39, 41, 42, 44, 46, 47, 48, 49, 51, 52, 54, 56, or 57.
28. The method of claim 27, wherein said ligand further comprises and effector moiety.
29. The method of claim 28, wherein said effector moiety is a therapeutic radiolabel, enzyme, cytotoxin, growth factor, or drag.
30. The method of claim 28, wherein said effector moiety is a radiolabel, enzyme, cytotoxin, growth factor, or drag.
31. The method of claim 30 wherein the radiolabel is 90yttrium.
32. The method of claim 31 wherein the radiolabel is mindium.
33. The method of claim 27 wherein said ligand is a monoclonal antibody or fragment thereof.
34. The method of claim 27 wherein said ligand is a small molecule.
35. The method of claim 27 wherein said ligand is a peptide.
36. The method of claim 1, wherein the isolated nucleic acid comprises the nucleotide sequence of SEQ ID NO:2.
37. The method of claim 1 , wherein the isolated nucleic acid comprises the nucleotide sequence of SEQ ID NO:4.
38. The method of claim 1, wherein the isolated nucleic acid comprises the nucleotide sequence of SEQ ID NO:6.
39. The method of claim 1 , wherein the isolated nucleic acid comprises the nucleotide sequence of SEQ ID NO:8.
40. The method of claim 1, wherein the isolated nucleic acid comprises the nucleotide sequence of SEQ ID NO: 10.
41. The method of claim 1 , wherein the isolated nucleic acid comprises the nucleotide sequence of SEQ ID NO: 13.
42. The method of claim 1, wherein the isolated nucleic acid comprises the nucleotide sequence of SEQ ID NO: 15.
PCT/US2003/009534 2002-03-28 2003-03-28 Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas WO2003083074A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU2003222103A AU2003222103A1 (en) 2002-03-28 2003-03-28 Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas
US10/509,131 US20060089493A1 (en) 2002-04-30 2003-03-28 Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
US36772702P 2002-03-28 2002-03-28
US60/367,727 2002-03-28
US38132802P 2002-05-20 2002-05-20
US60/381,328 2002-05-20
US38674702P 2002-06-10 2002-06-10
US60/386,747 2002-06-10
US42756402P 2002-11-20 2002-11-20
US60/427,564 2002-11-20

Publications (2)

Publication Number Publication Date
WO2003083074A2 true WO2003083074A2 (en) 2003-10-09
WO2003083074A9 WO2003083074A9 (en) 2009-10-08

Family

ID=28679084

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2003/009534 WO2003083074A2 (en) 2002-03-28 2003-03-28 Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas

Country Status (2)

Country Link
AU (1) AU2003222103A1 (en)
WO (1) WO2003083074A2 (en)

Cited By (75)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1487877A2 (en) * 2001-09-18 2004-12-22 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
EP1923072A1 (en) * 2005-08-09 2008-05-21 Takeda Pharmaceutical Company Limited Prophylactic/therapeutic agent for cancer
JP2008532477A (en) * 2005-02-10 2008-08-21 オンコセラピー・サイエンス株式会社 Method for diagnosing bladder cancer
US7557204B2 (en) * 2004-02-04 2009-07-07 The Chinese University Of Hong Kong CUDR as biomarker for cancer progression and therapeutic response
EP2260858A2 (en) 2003-11-06 2010-12-15 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
EP2286844A2 (en) 2004-06-01 2011-02-23 Genentech, Inc. Antibody-drug conjugates and methods
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
WO2011130598A1 (en) 2010-04-15 2011-10-20 Spirogen Limited Pyrrolobenzodiazepines and conjugates thereof
US8057996B2 (en) 2002-08-16 2011-11-15 Agensys, Inc. Nucleic acids and corresponding proteins entitled 202P5A5 useful in treatment and detection of cancer
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
WO2012140627A1 (en) 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
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
US8552146B2 (en) 2006-10-17 2013-10-08 Oncotherapy Science, Inc. Peptide vaccines for cancers expressing MPHOSPH1 or DEPDC1 polypeptides
WO2014057074A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014140862A2 (en) 2013-03-13 2014-09-18 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
WO2014159981A2 (en) 2013-03-13 2014-10-02 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
US8975086B2 (en) 2008-08-28 2015-03-10 Oncotherapy Science, Inc. Method for treating or preventing bladder cancer using the DEPDC1 polypeptide
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
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
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
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
WO2017165734A1 (en) 2016-03-25 2017-09-28 Genentech, Inc. Multiplexed total antibody and antibody-conjugated drug quantification assay
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
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
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
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
US10676508B2 (en) 2015-08-12 2020-06-09 Oncotherapy Science, Inc. DEPDC1-derived peptide and vaccine containing same
WO2020123275A1 (en) 2018-12-10 2020-06-18 Genentech, Inc. Photocrosslinking peptides for site specific conjugation to fc-containing proteins
US10695439B2 (en) 2016-02-10 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine conjugates
US10695433B2 (en) 2012-10-12 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine-antibody 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

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2935273A1 (en) 2012-12-21 2015-10-28 MedImmune Limited Unsymmetrical pyrrolobenzodiazepines-dimers for use in the treatment of proliferative and autoimmune diseases
CN110452242A (en) 2012-12-21 2019-11-15 麦迪穆有限责任公司 Pyrrolobenzodiazepines Zhuo and its conjugate

Cited By (111)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7939268B2 (en) 2001-09-18 2011-05-10 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
EP1487877A4 (en) * 2001-09-18 2005-08-24 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
EP1487877A2 (en) * 2001-09-18 2004-12-22 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7951546B2 (en) 2001-09-18 2011-05-31 Genetech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US8426571B2 (en) 2002-08-16 2013-04-23 Agensys, Inc. Nucleic acids and corresponding proteins entitled 202P5A5 useful in treatment and detection of cancer
US8057996B2 (en) 2002-08-16 2011-11-15 Agensys, Inc. Nucleic acids and corresponding proteins entitled 202P5A5 useful in treatment and detection of cancer
EP2489364A1 (en) 2003-11-06 2012-08-22 Seattle Genetics, Inc. Monomethylvaline compounds onjugated to antibodies
EP2260858A2 (en) 2003-11-06 2010-12-15 Seattle Genetics, Inc. Monomethylvaline compounds capable of conjugation to ligands
EP3858387A1 (en) 2003-11-06 2021-08-04 Seagen Inc. Monomethylvaline compounds capable of conjugation to ligands
EP2486933A1 (en) 2003-11-06 2012-08-15 Seattle Genetics, Inc. Monomethylvaline compounds conjugated with antibodies
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
EP3434275A1 (en) 2003-11-06 2019-01-30 Seattle Genetics, Inc. Assay for cancer cells based on the use of auristatin conjugates with antibodies
US8163476B2 (en) 2004-02-04 2012-04-24 The Chinese University Of Hong Kong CUDR as biomarker for cancer progression and therapeutic response
US7557204B2 (en) * 2004-02-04 2009-07-07 The Chinese University Of Hong Kong CUDR as biomarker for cancer progression and therapeutic response
EP2286844A2 (en) 2004-06-01 2011-02-23 Genentech, Inc. Antibody-drug conjugates and methods
EP3088004A1 (en) 2004-09-23 2016-11-02 Genentech, Inc. Cysteine engineered antibodies and conjugates
JP2008532477A (en) * 2005-02-10 2008-08-21 オンコセラピー・サイエンス株式会社 Method for diagnosing bladder cancer
EP2011885A3 (en) * 2005-02-10 2009-06-24 Oncotherapy Science, Inc. Method of diagnosing bladder cancer
US8685641B2 (en) 2005-02-10 2014-04-01 Oncotherapy Science, Inc. Method of screening compounds for treating bladder cancer
US7998695B2 (en) 2005-02-10 2011-08-16 Oncotherapy Science, Inc. Method of diagnosing bladder cancer
EP1923072A4 (en) * 2005-08-09 2010-03-03 Takeda Pharmaceutical Prophylactic/therapeutic agent for cancer
EP1923072A1 (en) * 2005-08-09 2008-05-21 Takeda Pharmaceutical Company Limited Prophylactic/therapeutic agent for cancer
US8653234B2 (en) 2006-10-17 2014-02-18 Oncotherapy Science, Inc. Peptide vaccines for cancers expressing MPHOSPH1 or DEPDC1 polypeptides
US9017688B2 (en) 2006-10-17 2015-04-28 Oncotherapy Science, Inc. Peptide vaccines for cancers expressing MPHOSPH1 or DEPDC1 polypeptides
US9545437B2 (en) 2006-10-17 2017-01-17 Oncotherapy Science, Inc. Peptide vaccines for cancers expressing MPHOSPH1 or DEPDC1 polypeptides
US8552146B2 (en) 2006-10-17 2013-10-08 Oncotherapy Science, Inc. Peptide vaccines for cancers expressing MPHOSPH1 or DEPDC1 polypeptides
US8557955B2 (en) 2006-10-17 2013-10-15 Oncotherapy Science, Inc. Peptide vaccines for cancers expressing MPHOSPH1 or DEPDC1 polypeptides
US8975086B2 (en) 2008-08-28 2015-03-10 Oncotherapy Science, Inc. Method for treating or preventing bladder cancer using the DEPDC1 polypeptide
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
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
WO2012140627A1 (en) 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
US9409987B2 (en) 2011-04-15 2016-08-09 Compugen Ltd Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
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
WO2013130093A1 (en) 2012-03-02 2013-09-06 Genentech, Inc. Biomarkers for treatment with anti-tubulin chemotherapeutic compounds
US10722594B2 (en) 2012-10-12 2020-07-28 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US9931414B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11779650B2 (en) 2012-10-12 2023-10-10 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US11771775B2 (en) 2012-10-12 2023-10-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
WO2014057074A1 (en) 2012-10-12 2014-04-17 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
US10335497B2 (en) 2012-10-12 2019-07-02 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10994023B2 (en) 2012-10-12 2021-05-04 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9931415B2 (en) 2012-10-12 2018-04-03 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
EP2839860A1 (en) 2012-10-12 2015-02-25 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
US9919056B2 (en) 2012-10-12 2018-03-20 Adc Therapeutics S.A. Pyrrolobenzodiazepine-anti-CD22 antibody conjugates
US10799596B2 (en) 2012-10-12 2020-10-13 Adc Therapeutics S.A. Pyrrolobenzodiazepine-antibody conjugates
US10780181B2 (en) 2012-10-12 2020-09-22 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10751346B2 (en) 2012-10-12 2020-08-25 Medimmune Limited Pyrrolobenzodiazepine—anti-PSMA antibody conjugates
US10736903B2 (en) 2012-10-12 2020-08-11 Medimmune Limited Pyrrolobenzodiazepine-anti-PSMA 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
US10695433B2 (en) 2012-10-12 2020-06-30 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
US10646584B2 (en) 2012-10-12 2020-05-12 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US9889207B2 (en) 2012-10-12 2018-02-13 Medimmune Limited 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
WO2014159981A2 (en) 2013-03-13 2014-10-02 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
US9956299B2 (en) 2013-10-11 2018-05-01 Medimmune Limited Pyrrolobenzodiazepine—antibody conjugates
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
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
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
US10188746B2 (en) 2014-09-10 2019-01-29 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2016037644A1 (en) 2014-09-10 2016-03-17 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
WO2016040825A1 (en) 2014-09-12 2016-03-17 Genentech, Inc. Anthracycline disulfide intermediates, antibody-drug conjugates and methods
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
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
US10676508B2 (en) 2015-08-12 2020-06-09 Oncotherapy Science, Inc. DEPDC1-derived peptide and vaccine containing same
US11673915B2 (en) 2015-08-12 2023-06-13 Oncotherapy Science, Inc. DEPDC1-derived peptide and vaccine containing same
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
WO2017165734A1 (en) 2016-03-25 2017-09-28 Genentech, Inc. Multiplexed total antibody and antibody-conjugated drug quantification assay
EP4273551A2 (en) 2016-03-25 2023-11-08 F. Hoffmann-La Roche AG 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
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
US11938192B2 (en) 2017-06-14 2024-03-26 Medimmune Limited 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
WO2003083074A9 (en) 2009-10-08
AU2003222103A1 (en) 2003-10-13
AU2003222103A8 (en) 2009-11-19

Similar Documents

Publication Publication Date Title
US20080213166A1 (en) Novel Gene Targets and Ligands that Bind Thereto for Treatment and Diagnosis of Colon Carcinomas
WO2003083074A2 (en) Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas
US20020051990A1 (en) Novel gene targets and ligands that bind thereto for treatment and diagnosis of ovarian carcinomas
US20070104718A1 (en) Gene BRCC-1 and diagnostic and therapeutic uses thereof
US20110104252A1 (en) Anti-apoptotic gene scc-s2 and diagnostic and therapeutic uses thereof
EP1657256B1 (en) Compositions and methods for the treatment of tumor
CA2367125A1 (en) Reagents and methods useful for detecting diseases of the prostate
US7442520B2 (en) Gene BRCC-3 and diagnostic and therapeutic uses thereof
KR20090111307A (en) Prostate specific transcripts and the use thereof for prostate cancer therapeutics and diagnostics
US7253272B2 (en) Gene BRCC-2 and diagnostic and therapeutic uses thereof
US7351811B2 (en) Gene SCC-112 and diagnostic and therapeutic uses thereof
EP1642907B1 (en) Composition and methods for the diagnosis of tumours
US7244565B2 (en) Gene shinc-3 and diagnostic and therapeutic uses thereof
US7138512B2 (en) Gene SHINC-2 and diagnostic and therapeutic uses thereof
WO2003070889A2 (en) Prostate specific genes and the use thereof in design or therapeutics
US20060089493A1 (en) Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas
US20020061552A1 (en) Mammalian dishevelled-associated proteins
US20040161775A1 (en) Gene SHINC-1 and diagnostic and therapeutic uses thereof
WO2003059256A2 (en) Genes overexpresses by ovarian cancer and their use in developing novel therapeutics especially antibodies
US20030186232A1 (en) Human and non-human primate homologues of Nkd protein, nucleic acid sequences encoding, and uses thereof

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 SC SD SE SG SK SL TJ TM TN TR TT TZ UA UG US UZ VC 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 BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

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

Ref country code: DE

122 Ep: pct application non-entry in european phase
ENP Entry into the national phase in:

Ref document number: 2006089493

Country of ref document: US

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 10509131

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 10509131

Country of ref document: US

NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)