WO2001016291A2 - 07cg27 gene - Google Patents

07cg27 gene Download PDF

Info

Publication number
WO2001016291A2
WO2001016291A2 PCT/US2000/023291 US0023291W WO0116291A2 WO 2001016291 A2 WO2001016291 A2 WO 2001016291A2 US 0023291 W US0023291 W US 0023291W WO 0116291 A2 WO0116291 A2 WO 0116291A2
Authority
WO
WIPO (PCT)
Prior art keywords
gene
polypeptide
drug
sequence
nucleic acid
Prior art date
Application number
PCT/US2000/023291
Other languages
French (fr)
Other versions
WO2001016291A3 (en
Inventor
Sean V. Tavtigian
Brad Swedlund
Jacques Simard
Johanna M. Rommens
Original Assignee
Myriad Genetics, Inc.
The Hospital For Sick Children
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 Myriad Genetics, Inc., The Hospital For Sick Children filed Critical Myriad Genetics, Inc.
Priority to AU70709/00A priority Critical patent/AU7070900A/en
Publication of WO2001016291A2 publication Critical patent/WO2001016291A2/en
Publication of WO2001016291A3 publication Critical patent/WO2001016291A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • 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
    • 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

Definitions

  • the present invention relates generally to the field of human genetics. Specifically, the present invention relates to methods and materials used to isolate and detect a human prostate cancer marker gene (identified as 07CG27 herein). The invention further relates to the screening of drugs for therapy. Finally, the invention relates to the screening of the 07CG27 gene for mutations, which are useful for diagnosing the predisposition to disease.
  • a human prostate cancer marker gene identified as 07CG27 herein.
  • the invention further relates to the screening of drugs for therapy.
  • the invention relates to the screening of the 07CG27 gene for mutations, which are useful for diagnosing the predisposition to disease.
  • the genetics of cancer is complicated, involving gain or loss of function of three loosely defined classes of genes: (1) dominant, positive regulators of the transformed state (oncogenes); (2) recessive, negative regulators of the transformed state (tumor suppressor genes); (3) recessive genes involved in maintenance of genome integrity (caretaker genes) (Kinzler and Vogelstein, 1997). Over one hundred oncogenes have been characterized. About a dozen tumor suppressor genes and a similar number of caretaker genes have been identified; the number of genes falling into these last two classes is expected to increase beyond fifty (Knudson, 1993).
  • tumor predisposition genes Some of the tumor predisposition genes which have been cloned and characterized influence susceptibility to: 1) Retinoblastoma (RBI); 2) Wilms' tumor (WT1); 3) Li-Fraumeni (TP53); 4) Familial adenomatous polyposis (APC); 5) Neurofibromatosis type 1 (NFl); 6) Neurofibromatosis type 2 (NF2); 7) von Hippel-Lindau syndrome (VHL); 8) Multiple endocrine neoplasia type 2A (MEN2A); 9) Melanoma (CDKN2 and CDK4); 10) Breast and ovarian cancer (BRCA1 and BRCA2); 11) Cowden disease (MMACl); 12) Multiple endocrine neoplasia (MEN1)
  • Tumor predisposition loci that have been mapped genetically but not yet isolated include genes for: Lynch cancer family syndrome 2 (LCFS2); Neuroblastoma (NB); Beckwith- Wiedemann syndrome (BWS); Renal cell carcinoma (RCC); and Tuberous sclerosis 1 (TSC1).
  • LCFS2 Lynch cancer family syndrome 2
  • NB Neuroblastoma
  • BWS Beckwith- Wiedemann syndrome
  • RRC Renal cell carcinoma
  • TSC1 Tuberous sclerosis 1
  • Tumor predisposition genes that have been characterized to date encode products with similarities to a variety of protein types, including DNA binding proteins (WT1), ancillary transcription regulators (RBI), GTPase activating proteins or GAPs (NFl), cytoskeletal components (NF2), membrane bound receptor kinases (MEN2A), cell cycle regulators (CDKN2 and CDK4), tyrosine phosphatases (MMACl), as well as others with no obvious similarity to proteins of known function (BRCA2).
  • WT1 DNA binding proteins
  • RBI ancillary transcription regulators
  • NFl GTPase activating proteins or GAPs
  • NF2 cytoskeletal components
  • MEN2A membrane bound receptor kinases
  • CDKN2 and CDK4 cell cycle regulators
  • MMACl tyrosine phosphatases
  • the tumor predisposition gene originally identified through genetic studies has been shown to be lost or mutated in some sporadic tumors. This result suggests that regions of chromosomal aberration, whether germline, in tumors, or in tumor cell lines, may signify the position of important tumor predisposition genes involved both in genetic predisposition to cancer and in sporadic cancer.
  • Prostate cancer is the most common cancer in men in many western countries, and the second leading cause of cancer deaths in men. It accounts for more than 40,000 deaths in the US annually. The number of deaths is likely to continue rising over the next 10 to 15 years. In the US, prostate cancer is estimated to cost $1.5 billion per year in direct medical expenses. In addition to the burden of suffering, it is a major public-health issue. Numerous studies have provided evidence for familial clustering of prostate cancer, indicating that family history is a major risk factor for this disease (Cannon et al., 1982; Steinberg et al., 1990; Carter et al, 1993). Prostate cancer has long been recognized to be, in part, a familial disease.
  • Woolf (1960) described a relative risk of 3.0 of developing prostate cancer among first-degree relatives of prostate cancer cases in Utah using death certificate data. Relative risks ranging from 3 to 11 for first-degree relatives of prostate cancer cases have been reported (Cannon et al., 1982; Woolf, 1960; Fincham et al., 1990; Meikle et al., 1985; Krain, 1974; Morganti et al., 1956; Goldgar et al., 1994). Carter et al. (1992) performed segregation analysis on families ascertained through a single prostate cancer proband.
  • predisposing alleles of the gene are recessive, yet susceptibility to cancer is inherited in a dominant fashion: men who possess one predisposing allele (and one wild type allele) risk developing cancer, because their prostate cells may spontaneously lose the wild type allele.
  • This model applies to both tumor suppressor and caretaker genes described above.
  • prostate cancer predisposing alleles are truly dominant; that is, a wild type allele cannot overcome the tumor-forming role of the predisposing allele.
  • a cell that carries both wild type and mutant alleles would not necessarily lose the wild type copy before giving rise to malignant cells. Instead, prostate cells in predisposed individuals would undergo some other stochastic change(s) leading to cancer. If a prostate cancer predisposing alleles are recessive, the prostate cancer predisposing gene is expected to be expressed in normal prostate tissue but not functionally expressed in prostate tumors.
  • prostate cancer predisposing alleles are dominant, the wild type gene may or may not be expressed in normal prostate tissue. However, the predisposing allele will likely be expressed in prostate tumor cells.
  • Evidence for a prostate cancer susceptibility locus (identified in the literature as HPCl) on the long arm of chromosome 1, which is hypothesized to explain approximately 35% of families, was recently presented (Smith et al., 1996). Although several groups report evidence supporting this localization, it has not yet been confirmed statistically. Both the original Smith et al. report and a subsequent analysis of additional families (Cooney et al., 1997), suggest that the bulk of linkage evidence comes from African-American high-risk kindreds.
  • the present invention relates generally to the field of human genetics. Specifically, the present invention relates to methods and materials used to isolate and detect a human prostate cancer marker gene located within the HPCl region (identified herein as the 07CG27 gene). The invention further relates to the screening of drugs for therapy. Finally, the invention relates to the screening of the 07CG27 gene for mutations or for overexpression, which are useful for diagnosing the predisposition to disease.
  • the 07CG27 gene is a homo log of two known genes HLA-B associated transcript 1 (BAT1) and BAT2.
  • the 07CG27 gene is useful as a marker for the HPCl locus and as a marker for prostate cancer.
  • the present invention relates generally to the field of human genetics. Specifically, the present invention relates to methods and materials used to isolate and detect a human prostate cancer marker gene (identified herein as the 07CG27 gene). The invention further relates to the screening of drugs for therapy. Finally, the invention relates to the screening of the 07CG27 gene for mutations or overexpression, which are useful for diagnosing the predisposition to disease.
  • the 07CG27 gene is a homolog of two known genes HLA-B associated transcript 1 (BAT1) and BAT2.
  • the 07CG27 gene is useful as a marker for the HPCl locus and as a marker for prostate cancer.
  • the present invention provides an isolated polynucleotide comprising all, or a portion of the 07CG27 locus or of a mutated 07CG27 locus, preferably at least eight bases and not more than about 300 kb in length. Such polynucleotides may be antisense polynucleotides.
  • the present invention also provides a recombinant construct comprising such an isolated polynucleotide, for example, a recombinant construct suitable for expression in a transformed host cell.
  • Also provided by the present invention are methods of detecting a polynucleotide comprising a portion of the 07CG27 locus or its expression product in an analyte. Such methods may further comprise the step of amplifying the portion of the 07CG27 locus, and may further include a step of providing a set of polynucleotides which are primers for amplification of said portion of the 07CG27 locus. The method is useful for either diagnosis of the predisposition to disease or the prognosis of disease.
  • the present invention further provides an isolated polypeptide comprising all or part of the polypeptide encoded by the 07CG27 gene or a mutated form of the polypeptide.
  • the 07CG27 polypeptide is a GEF protein and functions as a ras activator.
  • the present invention also provides isolated antibodies, preferably monoclonal antibodies, which specifically bind to an isolated polypeptide comprised of at least five amino acid residues encoded by the 07CG27 locus.
  • the present invention also provides kits for detecting in an analyte a polynucleotide comprising a portion of the 07CG27 locus, the kits comprising a polynucleotide complementary to the portion of the 07CG27 locus packaged in a suitable container, and instructions for its use.
  • the present invention further provides methods of preparing a polynucleotide comprising polymerizing nucleotides to yield a sequence comprised of at least eight consecutive nucleotides of the 07CG27 locus; and methods of preparing a polypeptide comprising polymerizing amino acids to yield a sequence comprising at least five amino acids encoded within the 07CG27 locus.
  • the present invention further provides methods of screening the 07CG27 gene to identify mutations. Such methods may further comprise the step of amplifying a portion of the 07CG27 locus, and may further include a step of providing a set of polynucleotides which are primers for amplification of said portion of the 07CG27 locus.
  • Such methods may also include a step of providing the complete set of short polynucleotides defined by the sequence of 07CG27 or discrete subsets of that sequence, all single-base substitutions of that sequence or discrete subsets of that sequence, all 1-, 2-, 3-, or 4-base deletions of that sequence or discrete subsets of that sequence, and all 1-, 2-, 3-, or 4-base insertions in that sequence or discrete subsets of that sequence.
  • the method is useful for identifying mutations for use in either diagnosis of a predisposition to disease or the prognosis of disease.
  • the present invention further provides methods of screening suspected 07CG27 mutant alleles to identify mutations in the 07CG27 gene.
  • the present invention provides methods to screen drugs for inhibition of 07CG27 gene product function as an anticancer therapy. Since the 07CG27 gene is an oncogene which acts as an activator of ras, any small molecule which interrupts this interaction will have anti-cancer activity. Thus, such drugs are useful for therapy.
  • the 07CG27 locus which is a marker for prostate cancer, is a gene encoding an 07CG27 protein, which has been found to have no significant homology with publicly available protein or DNA sequences. This gene is termed 07CG27 herein.
  • the mutational events of the 07CG27 locus can involve deletions, insertions and point mutations within the coding sequence and the non-coding sequence.
  • the preliminary evidence is that 07CG27 is an oncogene.
  • the region containing the 07CG27 locus was identified using a variety of genetic techniques. Genetic mapping techniques initially defined the 07CG27 region in terms of recombination with genetic markers. Based upon studies of large extended families ("kindreds") with multiple cases of prostate cancer, a chromosomal region has been pinpointed that contains the 07CG27 gene, as well as putative susceptibility alleles in the 07CG27 locus. Population Resources Large, well-documented Utah kindreds are especially important in providing good resources for human genetic studies. Each large kindred independently gives evidence whether or not a 07CG27 predisposing allele is segregating in that family.
  • Recombinants informative for localization and isolation of the 07CG27 locus could be obtained only from kindreds large enough to confirm the presence of a susceptibility allele.
  • Large sibships are especially important for studying prostate cancer, since penetrance of the 07CG27 predisposing allele is reduced both by age and sex, making informative sibships difficult to find.
  • large sibships are essential for constructing haplotypes of deceased individuals by inference from the haplotypes of their close relatives.
  • markers are essential for linking a disease to a region of a chromosome.
  • markers include restriction fragment length polymorphisms (RFLPs) (Botstein et al, 1980), markers with a variable number of tandem repeats (VNTRs) (Jeffreys et al, 1985; Nakamura et al, 1987), and an abundant class of DNA polymorphisms based on short tandem repeats (STRs), especially repeats of CpA (Weber and May, 1989; Litt et al, 1989).
  • RFLPs restriction fragment length polymorphisms
  • VNTRs variable number of tandem repeats
  • STRs short tandem repeats
  • Genetic markers useful in searching for a genetic locus associated with a disease can be selected on an ad hoc basis, by densely covering a specific chromosome, or by detailed analysis of a specific region of a chromosome.
  • a preferred method for selecting genetic markers linked with a disease involves evaluating the degree of informativeness of kindreds to determine the ideal distance between genetic markers of a given degree of polymorphism, then selecting markers from known genetic maps which are ideally spaced for maximal efficiency. Informativeness of kindreds is measured by the probability that the markers will be heterozygous in unrelated individuals.
  • STR markers which are detected by amplification of the target nucleic acid sequence using PCR; such markers are highly informative, easy to assay (Weber and May, 1989), and can be assayed simultaneously using multiplexing strategies (Skolnick and Wallace, 1988), greatly reducing the number of experiments required.
  • markers that flank the disease locus i.e., one or more markers proximal to the disease locus, and one or more markers distal to the disease locus.
  • candidate markers can be selected from a known genetic map.
  • new markers can be identified by the STR technique.
  • Contig assembly Given a genetically defined interval flanked by meiotic recombinants, one needs to generate a contig of genomic clones that spans that interval. Publicly available resources, such as the Whitehead integrated maps of the human genome (e.g., the WICGR Chr 1 map of Nov.
  • YAC yeast artificial chromosome
  • STSs sequence tagged sites
  • YAC CEPH yeast artificial chromosome
  • Oligonucleotide primer pairs for the markers located in the interval can be synthesized and used to screen libraries of bacterial artificial chromosomes (BACs) and PI artificial chromosomes (PACs). Successive rounds of BAC/PAC library screening with BAC or PAC end markers enables the completion of a BAC/PAC clone contig that spans the genetically defined interval.
  • BACs bacterial artificial chromosomes
  • PACs PI artificial chromosomes
  • Genomic sequencing Given a tiling path of BAC and PAC clones across a defined interval, one useful gene finding strategy is to generate an almost complete genomic sequence of that interval. Random genomic clone sublibraries can be prepared from each BAC or PAC clone in the tiling path. Individual sublibrary clones sufficient in number to generate an, on average, 6x redundant sequence of each BAC or PAC can then be end-sequenced with vector primers. These sequences can be assembled into sequence contigs, and these contigs placed in a local genomic sequence database. One can search the genomic sequence contigs for sequence similarity with known genes and expressed sequence tags (ESTs), examine them for the presence of long open translational reading frames, and characterize them for CpG dinucleotide frequency.
  • ESTs expressed sequence tags
  • cDNA clones cognate to the tiling path BACs and PACs.
  • One preferred cDNA cloning strategy is hybrid selection.
  • cDNA can be prepared from a number of human tissues and human cell lines in such a manner that the cDNA molecules have PCR primer binding sites (anchors) at each end. This cDNA can be affinity captured with the tiling path BACs and PAGsr-Captured cDNA can then be PCR amplified using the anchor primers and then cloned. Individual clones can then be end-sequenced with vector primers.
  • sequences of these cDNA clones can_be analyzed for similarity to genomic sequence contigs generated from BACs and PACs on the tiling path. One can then identify individual exons of genes in the genetically defined interval by parsing the sequences of true-positive hybrid selected clones across these genomic sequence contigs.
  • RACE Rapid amplification of cDNA ends
  • cDNA can be prepared from a number of human tissues in a manner such that the cDNA molecules have PCR primer binding sites (anchors) at their 5' ends, 3' ends, or both.
  • anchors PCR primer binding sites
  • PCR amplification with 3' end anchor primers and gene specific forward primers can generate 3' RACE products.
  • cDNA cloning techniques can also miss exons that lie between already known exons of a gene; for instance, this can easily occur if a particular exon is only included in a relatively rare splice variant of a transcript.
  • Combinatorial inter-exon PCR is an effective strategy for detecting these exons.
  • One can design a forward primer based on sequences from the first known exon of the gene and a set of reverse primers, one based on the sequence of each of the downstream exons (or any subset thereof) of the gene.
  • PCR amplify from cDNA of tissues and cell lines thought to express the gene using all the combinations of the forward primer with each reverse primer. Combinations as complex as a forward primer from each exon paired with a reverse primer from each exon, subject only to the limitation that the forward primer should be from an exon upstream of the exon from which the reverse primer was designed, can be tried.
  • PCR products which differ in length from the expected product can be gel purified. In either RACE or combinatorial inter-exon PCRs, the PCR products can either be gel purified and then sequenced directly or first cloned and then sequenced.
  • cDNA library screening Another useful strategy for finding new 5', 3', or internal sequences is cDNA library screening.
  • bacteriophage 1 cDNA libraries prepared from RNA from tissues or cell lines thought to express the gene.
  • 07CG27 any particular gene located within the genetically defined interval is 07CG27 is obtained by finding sequences in DNA or RNA extracted from affected kindred members which create abnormal 07CG27 gene products or abnormal levels of 07CG27 gene product.
  • 07CG27 predisposing alleles will co-segregate with the disease in large kindreds. They will also be present at a much higher frequency in non-kindred individuals with prostate cancer than in individuals in the general population.
  • tumors often mutate somatically at loci which are in other instances mutated in the germline, we expect to see normal germline 07CG27 alleles mutated into sequences which are identical or similar to 07CG27 predisposing alleles in DNA extracted from tumor tissue.
  • Less severe disruptive mutations would include small in-frame deletions and nonconservative base pair substitutions which would have a significant effect on the protein produced, such as changes to or from a cysteine residue, from a basic to an acidic amino acid or vice versa, from a hydrophobic to hydrophilic amino acid or vice versa, or other mutations which would affect secondary, tertiary or quaternary protein structure.
  • Small deletions or base pair substitutions could also significantly alter protein expression by changing the level of transcription, splice pattern, mRNA stability, or translation efficiency of the 07CG27 transcript. Silent mutations or those resulting in conservative amino acid substitutions would not generally be expected to disrupt protein function.
  • alteration of the wild-type 07CG27 locus is detected.
  • the method can be performed by detecting the wild-type 07CG27 locus and confirming the lack of a predisposition to cancer at the 07CG27 locus.
  • "Alteration of a wild-type gene” encompasses all forms of mutations including deletions, insertions and point mutations in the coding and noncoding regions. Deletions may be of the entire gene or of only a portion of the gene. Point mutations may result in stop codons, frameshift mutations or amino acid substitutions. Somatic mutations are those which occur only in certain tissues, e.g., in the tumor tissue, and are not inherited in the germline.
  • Germline mutations can be found in any of a body's tissues and are inherited. The finding of 07CG27 mutations thus provides diagnostic information.
  • a 07CG27 allele which is not deleted e.g., found on the sister chromosome to a chromosome carrying an 07CG27 deletion
  • Useful diagnostic techniques include, but are not limited to fluorescent in situ hybridization (FISH), direct DNA sequencing, PFGE analysis, Southern blot analysis, single stranded conformation analysis (SSCA), RNase protection assay, allele-specific oligonucleotide (ASO), dot blot analysis and PCR-SSCP, as discussed in detail further below. Also useful is the recently developed technique of DNA microchip technology.
  • Predisposition to cancers can be ascertained by testing any tissue of a human for mutations of the 07CG27 gene.
  • a person who has inherited a germline 07CG27 mutation would be prone to develop cancers. This can be determined by testing DNA from any tissue of the person's body. Most simply, blood can be drawn and DNA extracted from the cells of the blood.
  • prenatal diagnosis can be accomplished by testing fetal cells, placental cells or amniotic cells for mutations of the 07CG27 gene. Alteration of a wild-type 07CG27 allele, whether, for example, by point mutation or deletion, can be detected by any of the means discussed herein.
  • Direct DNA sequencing either manual sequencing or automated fluorescent sequencing can detect sequence variation.
  • manual sequencing is very labor-intensive, but under optimal conditions, mutations in the coding sequence of a gene are rarely missed.
  • Another approach is the single-stranded conformation polymorphism assay (SSCA) (Orita et al, 1989). This method does not detect all sequence changes, especially if the DNA fragment size is greater than 200 bp, but can be optimized to detect most DNA sequence variation.
  • SSCA single-stranded conformation polymorphism assay
  • CDGE clamped denaturing gel electrophoresis
  • HA heteroduplex analysis
  • CMC chemical mismatch cleavage
  • tissue In order to detect the alteration of the wild-type 07CG27 gene in a tissue, it is helpful to isolate the tissue free from surrounding normal tissues.
  • Means for enriching tissue preparation for disease cells are known in the art.
  • the tissue may be isolated from paraffin or cryostat sections.
  • Disease cells may also be separated from normal cells by flow cytometry. These techniques, as well as other techniques for separating disease cells from normal cells, are well known in the art. If the disease tissue is highly contaminated with normal cells, detection of mutations is more difficult. Detection of point mutations may be accomplished by molecular cloning of the 07CG27 allele(s) and sequencing the allele(s) using techniques well known in the art.
  • the gene sequences can be amplified directly from a genomic DNA preparation from the tumor tissue, using known techniques. The DNA sequence of the amplified sequences can then be determined.
  • coli mutS protein (Modrich, 1991); and 6) allele-specific PCR (Rano and Kidd, 1989).
  • primers are used which hybridize at their 3' ends to a particular 07CG27 mutation. If the particular 07CG27 mutation is not present, an amplification product is not observed.
  • Amplification Refractory Mutation System (ARMS) can also be used, as disclosed in European Patent Application Publication No. 0332435 and in Newton et al, 1989. Insertions and deletions of genes can also be detected by cloning, sequencing and amplification.
  • RFLP restriction fragment length polymorphism
  • Such a method is particularly useful for screening relatives of an affected individual for the presence of the 07CG27 mutation found in that individual.
  • Other techniques for detecting insertions and deletions as known in the art can be used.
  • SSCA detects a band which migrates differentially because the sequence change causes a difference in single-strand, intramolecular base pairing.
  • RNase protection involves cleavage of the mutant polynucleotide into two or more smaller fragments.
  • DGGE detects differences in migration rates of mutant sequences compared to wild-type sequences, using a denaturing gradient gel.
  • an oligonucleotide is designed which detects a specific sequence, and the assay is performed by detecting the presence or absence of a hybridization signal.
  • the protein binds only to sequences that contain a nucleotide mismatch in a heteroduplex between mutant and wild-type sequences.
  • Mismatches are hybridized nucleic acid duplexes in which the two strands are not 100%) complementary. Lack of total homo logy may be due to deletions, insertions, inversions or substitutions. Mismatch detection can be used to detect point mutations in the gene or in its mRNA product. While these techniques are less sensitive than sequencing, they are simpler to perform on a large number of tumor samples.
  • An example of a mismatch cleavage technique is the RNase protection method.
  • the method involves the use of a labeled riboprobe which is complementary to the human wild-type 07CG27 gene coding sequence.
  • the riboprobe and either mRNA or DNA isolated from the tumor tissue are annealed (hybridized) together and subsequently digested with the enzyme RNase A which is able to detect some mismatches in a duplex RNA structure. If a mismatch is detected by RNase A, it cleaves at the site of the mismatch.
  • RNA product when the annealed RNA preparation is separated on an elecfrophoretic gel matrix, if a mismatch has been detected and cleaved by RNase A, an RNA product will be seen which is smaller than the full length duplex RNA for the riboprobe and the mRNA or DNA.
  • the riboprobe need not be the full length of the 07CG27 mRNA or gene but can be a segment of either. If the riboprobe comprises only a segment of the 07CG27 mRNA or gene, it will be desirable to use a number of these probes to screen the whole mRNA sequence for mismatches.
  • DNA probes can be used to detect mismatches, through enzymatic or chemical cleavage. See, e.g., Cotton et al, 1988; Shenk et al, 1975; Novack et al, 1986.
  • mismatches can be detected by shifts in the elecfrophoretic mobility of mismatched duplexes relative to matched duplexes. See, e.g., Cariello, 1988.
  • the cellular mRNA or DNA which might contain a mutation can be amplified using PCR (see below) before hybridization. Changes in DNA of the 07CG27 gene can also be detected using Southern hybridization, especially if the changes are gross rearrangements, such as deletions and insertions.
  • DNA sequences of the 07CG27 gene which have been amplified by use of PCR may also be screened using allele-specific probes.
  • These probes are nucleic acid oligomers, each of which contains a region of the 07CG27 gene sequence harboring a known mutation.
  • one oligomer may be about 30 nucleotides in length (although shorter and longer oligomers are also usable as well recognized by those of skill in the art), corresponding to a portion of the 07CG27 gene sequence.
  • PCR amplification products can be screened to identify the presence of a previously identified mutation in the 07CG27 gene.
  • Hybridization of allele-specific probes with amplified 07CG27 sequences can be performed, for example, on a nylon filter. Hybridization to a particular probe under high stringency hybridization conditions indicates the presence of the same mutation in the tumor tissue as in the allele-specific probe.
  • the newly developed technique of nucleic acid analysis via microchip technology is also applicable to the present invention. In this technique, literally thousands of distinct oligonucleotide probes are built up in an array on a silicon chip. Nucleic acid to be analyzed is fluorescently labeled and hybridized to the probes on the chip. It is also possible to study nucleic acid-protein interactions using these nucleic acid microchips.
  • the most definitive test for mutations in a candidate locus is to directly compare genomic 07CG27 sequences from diseased patients with those from a control population.
  • genomic 07CG27 sequences from diseased patients
  • those from a control population from which genomic 07CG27 sequences from diseased patients are compared.
  • Mutations from cancer patients falling outside the coding region of 07CG27 can be detected by examining the non-coding regions, such as introns and regulatory sequences near or within the 07CG27 gene.
  • An early indication that mutations in noncoding regions are important may come from Northern blot experiments that reveal messenger RNA molecules of abnormal size or abundance in cancer patients as compared to control individuals.
  • Alteration of 07CG27 mRNA expression can be detected by any techniques known in the art. These include Northern blot analysis, PCR amplification and RNase protection. Diminished mRNA expression indicates an alteration of the wild-type 07CG27 gene. Alteration of wild-type 07CG27 genes can also be detected by screening for alteration of wild-type 07CG27 protein. For example, monoclonal antibodies immunoreactive with 07CG27 can be used to screen a tissue. Lack of cognate antigen would indicate a 07CG27 mutation. Antibodies specific for products of mutant alleles could also be used to detect mutant 07CG27 gene product. Such immunological assays can be done in any convenient formats known in the art. These include Western blots, immunohistochemical assays and ELISA assays.
  • Any means for detecting an altered 07CG27 protein can be used to detect alteration of wild-type 07CG27 genes.
  • Functional assays such as protein binding determinations, can be used.
  • assays can be used which detect 07CG27 biochemical function. Finding a mutant 07CG27 gene product indicates alteration of a wild- type 07CG27 gene.
  • Mutant 07CG27 genes or gene products can also be detected in other human body samples, such as serum, stool, urine and sputum. The same techniques discussed above for detection of mutant 07CG27 genes or gene products in tissues can be applied to other body samples. In addition, the 07CG27 gene product itself may be secreted into the extracellular space and found in these body samples even in the absence of disease cells. By screening such body samples, a simple early diagnosis can be achieved for disease. In addition, the progress of therapy can be monitored more easily by testing such body samples for mutant 07CG27 genes or gene products.
  • the methods of diagnosis of the present invention are applicable to any disease in which 07CG27 has a role in its pathogenesis.
  • the diagnostic method of the present invention is useful for clinicians, so they can decide upon an appropriate course of treatment.
  • the primer pairs of the present invention are useful for determination of the nucleotide sequence of a particular 07CG27 allele using PCR.
  • the pairs of single-stranded DNA primers can be annealed to sequences within or surrounding the 07CG27 gene on chromosome 1 in order to prime amplifying DNA synthesis of the 07CG27 gene itself.
  • a complete set of these primers allows synthesis of all of the nucleotides of the 07CG27 gene coding sequences, i.e., the exons.
  • the set of primers preferably allows synthesis of both intron and exon sequences. Allele-specific primers can also be used. Such primers anneal only to particular 07CG27 mutant alleles, and thus will only amplify a product in the presence of the mutant allele as a template.
  • primers may have restriction enzyme site sequences appended to their 5' ends.
  • all nucleotides of the primers are derived from 07CG27 sequences or sequences adjacent to 07CG27, except for the few nucleotides necessary to form a restriction enzyme site.
  • the primers themselves can be synthesized using techniques which are well known in the art. Generally, the primers can be made using oligonucleotide synthesizing machines which are commercially available. Given the sequence of the 07CG27 open reading frame shown in SEQ ID NO:l, design of particular primers is well within the skill of the art.
  • the nucleic acid probes provided by the present invention are useful for a number of purposes. They can be used in Southern hybridization to genomic DNA and in the RNase protection method for detecting point mutations already discussed above. The probes can be used to detect PCR amplification products. They may also be used to detect mismatches with the
  • mutant 07CG27 alleles can be initially identified by identifying mutant (altered) 07CG27 proteins, using conventional techniques. The mutant alleles are then sequenced to identify the specific mutation for each allele. The mutations, especially those which lead to an altered function of the 07CG27 protein, are then used for the diagnostic methods of the present invention. It has also been discovered that the 07CG27 gene is a marker for prostate cancer.
  • Amplification of Polynucleotides utilizes methods such as the polymerase chain reaction (PCR), ligation amplification (or ligase chain reaction, LCR) and amplification methods based on the use of Q-beta replicase. Also useful are strand displacement amplification (SDA) and nucleic acid sequence based amplification (NASBA). These methods are well known and widely practiced in the art. See, e.g., U.S. Patents 4,683,195 and 4,683,202 and Innis et al, 1990 (for PCR); and Wu et al, 1989a (for LCR); U.S.
  • Patents 5,270,184 and 5,455,166 for SDA
  • Spargo et al. 1996
  • Primers useful to amplify sequences from the 07CG27 region are preferably complementary to, and hybridize specifically to sequences in the 07CG27 region or in regions that flank a target region therein.
  • 07CG27 sequences generated by amplification may be sequenced directly. Alternatively, but less desirably, the amplified sequence(s) may be cloned prior to sequence analysis.
  • a method for the direct cloning and sequence analysis of enzymatically amplified genomic segments has been described by Scharf, 1986.
  • analyte polynucleotide and “analyte strand” refer to a single- or double-stranded polynucleotide which is suspected of containing a target sequence, and which may be present in a variety of types of samples, including biological samples.
  • Antibodies The present invention also provides polyclonal and/or monoclonal antibodies and fragments thereof, and immunologic binding equivalents thereof, which are capable of specifically binding to the 07CG27 polypeptides and fragments thereof or to polynucleotide sequences from the 07CG27 region, particularly from the 07CG27 locus or a portion thereof.
  • antibody is used both to refer to a homogeneous molecular entity, or a mixture such as a serum product made up of a plurality of different molecular entities.
  • Polypeptides may be prepared synthetically in a peptide synthesizer and coupled to a carrier molecule (e.g., keyhole limpet hemocyanin) and injected over several months into rabbits. Rabbit sera is tested for immunoreactivity to the 07CG27 polypeptide or fragment.
  • Monoclonal antibodies may be made by injecting mice with the protein polypeptides, fusion proteins or fragments thereof. Monoclonal antibodies will be screened by ELISA and tested for specific immunoreactivity with 07CG27 polypeptide or fragments thereof. See, Harlow and Lane, 1988. These antibodies will be useful in assays as well as pharmaceuticals.
  • antibodies specific for binding may be either polyclonal or monoclonal, and may be produced by in vitro or in vivo techniques well known in the art.
  • an appropriate target immune system typically mouse or rabbit
  • Substantially purified antigen is presented to the immune system in a fashion determined by methods appropriate for the animal and by other parameters well known to immunologists. Typical sites for injection are in footpads, intramuscularly, intraperitoneally, or intradermally. Of course, other species may be substituted for mouse or rabbit.
  • Polyclonal antibodies are then purified using techniques known in the art, adjusted for the desired specificity.
  • An immunological response is usually assayed with an immunoassay.
  • immunoassays involve some purification of a source of antigen, for example, that produced by the same cells and in the same fashion as the antigen.
  • a variety of immunoassay methods are well known in the art. See, e.g., Harlow and Lane, 1988, or Goding, 1986. Monoclonal antibodies with affinities of 10 " M "1 or preferably 10 "9 to 10 "10 M “1 or stronger will typically be made by standard procedures as described, e.g., in Harlow and Lane, 1988 or Goding, 1986. Briefly, appropriate animals will be selected and the desired immunization protocol followed.
  • the spleens of such animals are excised and individual spleen cells fused, typically, to immortalized myeloma cells under appropriate selection conditions. Thereafter, the cells are clonally separated and the supernatants of each clone tested for their production of an appropriate antibody specific for the desired region of the antigen.
  • Suitable techniques involve in vitro exposure of lymphocytes to the antigenic polypeptides, or alternatively, to selection of libraries of antibodies in phage or similar vectors. See Huse et al, 1989.
  • the polypeptides and antibodies of the present invention may be used with or without modification. Frequently, polypeptides and antibodies will be labeled by joining, either covalently or non-covalently, a substance which provides for a detectable signal.
  • labels and conjugation techniques are known and are reported extensively in both the scientific and patent literature. Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent agents, chemiluminescent agents, magnetic particles and the like. Patents teaching the use of such labels include U.S.
  • Binding partner refers to a molecule capable of binding a ligand molecule with high specificity, as for example, an antigen and an antigen-specific antibody or an enzyme and its inhibitor.
  • the specific binding partners must bind with sufficient affinity to immobilize the analyte copy/complementary strand duplex (in the case of polynucleotide hybridization) under the isolation conditions.
  • Specific binding partners are known in the art and include, for example, biotin and avidin or streptavidin, IgG and protein A, the numerous, known receptor- ligand couples, and complementary polynucleotide strands.
  • the partners are normally at least about 15 bases in length, and may be at least 40 bases in length. It is well recognized by those of skill in the art that lengths shorter than 15, between 15 and 40, and greater than 40 bases may also be used.
  • the polynucleotides may be composed of DNA, RNA, or synthetic nucleotide analogs.
  • a “biological sample” refers to a sample of tissue or fluid suspected of containing an analyte polynucleotide or polypeptide from an individual including, but not limited to, e.g., plasma, serum, spinal fluid, lymph fluid, the external sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, blood cells, tumors, organs, tissue and samples of in vitro cell culture constituents.
  • diagnosis or “prognosing,” as used in the context of neoplasia, are used to indicate 1) the classification of lesions as neoplasia, 2) the determination of the severity of the neoplasia, or 3) the monitoring of the disease progression, prior to, during and after treatment.
  • Encode A polynucleotide is said to "encode” a polypeptide if, in its native state or when manipulated by methods well known to those skilled in the art, it can be transcribed and/or translated to produce the mRNA for and/or the polypeptide or a fragment thereof.
  • the anti- sense strand is the complement of such a nucleic acid, and the encoding sequence can be deduced therefrom.
  • nucleic acid e.g., an RNA, DNA or a mixed polymer
  • An “isolated” or “substantially pure” nucleic acid is one which is substantially separated from other cellular components which naturally accompany a native human sequence or protein, e.g., ribosomes, polymerases, many other human genome sequences and proteins.
  • the term embraces a nucleic acid sequence or protein which has been removed from its naturally occurring environment, and includes recombinant or cloned DNA isolates and chemically synthesized analogs or analogs biologically synthesized by heterologous systems.
  • 07CG27 Allele refers to normal alleles of the 07CG27 locus as well as alleles carrying variations that predispose individuals to develop disease. Such predisposing alleles are also called “07CG27 predisposing alleles”.
  • Polynucleotide each refer to polynucleotides, all of which are in the 07CG27 region, that are likely to be expressed in normal tissue, certain alleles of which predispose an individual to develop disease. Mutations at the 07CG27 locus may be involved in the initiation and/or progression of disease.
  • the 07CG27 locus is intended to include coding sequences, intervening sequences and regulatory elements controlling transcription and/or translation.
  • the 07CG27 locus is intended to include all allelic variations of the DNA sequence.
  • nucleic acid when applied to a nucleic acid, refer to a nucleic acid which encodes an
  • 07CG27 polypeptide, fragment, homolog or variant including, e.g., protein fusions or deletions.
  • the nucleic acids of the present invention will possess a sequence which is either derived from, or substantially similar to a natural 07CG27-encoding gene or one having substantial homology with a natural 07CG27-encoding gene or a portion thereof.
  • the polynucleotide compositions of this invention include RNA, cDNA, genomic DNA, synthetic forms, and mixed polymers, both sense and antisense strands, and may be chemically or biochemically modified or may contain non-natural or derivatized nucleotide bases, as will be readily appreciated by those skilled in the art.
  • Such modifications include, for example, labels, methylation, substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoramidates, carbamates, etc.), charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), pendent moieties (e.g., polypeptides), intercalators (e.g., acridine, psoralen, etc.), chelators, alkylators, and modified linkages (e.g., alpha anomeric nucleic acids, etc.).
  • uncharged linkages e.g., methyl phosphonates, phosphotriesters, phosphoramidates, carbamates, etc.
  • charged linkages e.g., phosphorothioates, phosphorodithioates, etc.
  • pendent moieties
  • synthetic molecules that mimic polynucleotides in their ability to bind to a designated sequence via hydrogen bonding and other chemical interactions.
  • Such molecules are known in the art and include, for example, those in which peptide linkages substitute for phosphate linkages in the backbone of the molecule.
  • the present invention provides recombinant nucleic acids comprising all or part of the 07CG27 region.
  • the recombinant construct may be capable of replicating autonomously in a host cell. Alternatively, the recombinant construct may become integrated into the chromosomal DNA of the host cell.
  • a recombinant polynucleotide comprises a polynucleotide of genomic, cDNA, semi-synthetic, or synthetic origin which, by virtue of its origin or manipulation, 1) is not associated with all or a portion of a polynucleotide with which it is associated in nature; 2) is linked to a polynucleotide other than that to which it is linked in nature; or 3) does not occur in nature.
  • nucleic acids comprising sequences otherwise not naturally occurring are provided by this invention.
  • wild-type sequence may be employed, it will often be altered, e.g., by deletion, substitution or insertion.
  • cDNA or genomic libraries of various types may be screened as natural sources of the nucleic acids of the present invention, or such nucleic acids may be provided by amplification of sequences resident in genomic DNA or other natural sources, e.g., by PCR.
  • the choice of cDNA libraries normally corresponds to a tissue source which is abundant in mRNA for the desired proteins. Phage libraries are normally preferred, but other types of libraries may be used. Clones of a library are spread onto plates, transferred to a substrate for screening, denatured and probed for the presence of desired sequences.
  • the DNA sequences used in this invention will usually comprise at least about five codons (15 nucleotides), more usually at least about 7-15 codons, and most preferably, at least about 35 codons. One or more introns may also be present. This number of nucleotides is usually about the minimal length required for a successful probe that would hybridize specifically with an 07CG27-encoding sequence.
  • nucleic acid manipulation is described generally, for example, in Sambrook et al, 1989 or Ausubel et al, 1992.
  • Reagents useful in applying such techniques such as restriction enzymes and the like, are widely known in the art and commercially available from such vendors as New England BioLabs, Boehringer Mannheim, Amersham, Promega Biotec, U. S. Biochemicals, New England Nuclear, and a number of other sources.
  • the recombinant nucleic acid sequences used to produce fusion proteins of the present invention may be derived from natural or synthetic sequences. Many natural gene sequences are obtainable from various cDNA or from genomic libraries using appropriate probes. See, GenBank, National Institutes of Health.
  • 07CG27 Region refers to a portion of human chromosome 1 containing the 07CG27 gene.
  • 07CG27 locus As used herein, the terms “07CG27 locus”, “07CG27 allele” and “07CG27 region” all refer to the double-stranded DNA comprising the locus, allele, or region, as well as either of the single-stranded DNAs comprising the locus, allele or region.
  • a "portion" of the 07CG27 locus or region or allele is defined as having a minimal size of at least about eight nucleotides, or preferably about 15 nucleotides, or more preferably at least about 25 nucleotides, and may have a minimal size of at least about 40 nucleotides. This definition includes all sizes in the range of 8-40 nucleotides as well as greater than 40 nucleotides.
  • this definition includes nucleic acids of 8, 12, 15, 20, 25, 40, 60, 80, 100, 200, 300, 400, 500 nucleotides, or nucleic acids having any number of nucleotides within these ranges of values (e.g., 9, 10, 11, 16, 23, 30, 38, 50, 72, 121, etc., nucleotides), or nucleic acids having more than 500 nucleotides.
  • the present invention includes all novel nucleic acids having at least 8 nucleotides derived from any of SEQ ID NOs:l and 3-21 and any combination of these sequences as described in further detail below, its complement or functionally equivalent nucleic acid sequences.
  • the present invention .does not include nucleic acids which exist in the prior art.
  • the present invention includes all nucleic acids having at least 8 nucleotides derived from any of SEQ ID NOs:l and 3-21 and any combination of these sequences as described in further detail below with the proviso that it does not include nucleic acids existing in the prior art.
  • "07CG27 protein” or “07CG27 polypeptide” refers to a protein or polypeptide encoded by the 07CG27 locus, variants or fragments thereof.
  • the term “polypeptide” refers to a polymer of amino acids and its equivalent and does not refer to a specific length of the product; thus, peptides, oligopeptides and proteins are included within the definition of a polypeptide.
  • polypeptides for example, glycosylations, acetylations, phosphorylations, and the like. Included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), polypeptides with substituted linkages as well as other modifications known in the art, both naturally and non-naturally occurring. Ordinarily, such polypeptides will be at least about 50% homologous to the native 07CG27 sequence, preferably in excess of about 90%, and more preferably at least about 95% or 98% or more homologous. Also included are proteins encoded by DNA which hybridize under high or low stringency conditions, to 07CG27-encoding nucleic acids and closely related polypeptides or proteins retrieved by antisera to the 07CG27 protein(s).
  • An 07CG27 polypeptide may be that derived from any of the exons described herein which may be in isolated and/or purified form, free or substantially free of material with which it is naturally associated.
  • the polypeptide may, if produced by expression in a prokaryotic cell or produced synthetically, lack native post-translational processing, such as glycosylation.
  • the present invention is also directed to polypeptides which are sequence variants, alleles or derivatives of an 07CG27 polypeptide.
  • Such polypeptides may have an amino acid sequence which differs from that derived form any of the exons described herein by one or more of addition, substitution, deletion or insertion of one or more amino acids.
  • Preferred such polypeptides have 07CG27 function.
  • Substitutional variants typically contain the exchange of one amino acid for another at one or more sites within the protein, and may be designed to modulate one or more properties of the polypeptide, such as stability against proteolytic cleavage, without the loss of other functions or properties.
  • Amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophihcity, and/or the amphipathic nature of the residues involved.
  • Preferred substitutions are ones which are conservative, that is, one amino acid is replaced with one of similar shape and charge.
  • Conservative substitutions are well known in the art and typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and tyrosine, phenylalanine.
  • Certain amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies or binding sites on substrate molecules or binding sites on proteins interacting with an 07CG27 polypeptide. Since it is the interactive capacity and nature of a protein which defines that protein's biological functional activity, certain amino acid substitutions can be made in a protein sequence, and its underlying DNA coding sequence, and nevertheless obtain a protein with like properties. In making such changes, the hydropathic index of amino acids may be considered. The importance of the hydrophobic amino acid index in conferring interactive biological function on a protein is generally understood in the art (Kyte and Doolittle, 1982).
  • the substitution of like amino acids can be made effectively on the basis of hydrophihcity.
  • the importance of hydrophihcity in conferring interactive biological function of a protein is generally understood in the art (U.S. Patent 4,554,101).
  • the use of the hydrophobic index or hydrophihcity in designing polypeptides is further discussed in U.S. Patent 5,691,198.
  • the length of polypeptide sequences compared for homology will generally be at least about 16 amino acids, usually at least about 20 residues, more usually at least about 24 residues, typically at least about 28 residues, and preferably more than about 35 residues.
  • “Operably linked” refers to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner.
  • a promoter is operably linked to a coding sequence if the promoter affects its transcription or expression.
  • peptide mimetic or mimetic is intended to refer to a substance which has the essential biological activity of an 07CG27 polypeptide.
  • a peptide mimetic may be a peptide- containing molecule that mimics elements of protein secondary structure (Johnson et al., 1993).
  • the underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions, such as those of antibody and antigen, enzyme and substrate or scaffolding proteins.
  • a peptide mimetic is designed to permit molecular interactions similar to the natural molecule.
  • a mimetic may not be a peptide at all, but it will retain the essential biological activity of a natural 07CG27 polypeptide.
  • Probes Polynucleotide polymorphisms associated with 07CG27 alleles which predispose to certain cancers or are associated with most cancers are detected by hybridization with a polynucleotide probe which forms a stable hybrid with that of the target sequence, under highly stringent to moderately stringent hybridization and wash conditions. If it is expected that the probes will be perfectly complementary to the target sequence, high stringency conditions will be used. Hybridization stringency may be lessened if some mismatching is expected, for example, if variants are expected with the result that the probe will not be completely complementary. Conditions are chosen which rule out nonspecific/adventitious bindings, that is, which minimize noise.
  • Probes for 07CG27 alleles may be derived from the sequences of the 07CG27 region or its cDNAs. The probes may be of any suitable length, which span all or a portion of the 07CG27 region, and which allow specific hybridization to the 07CG27 region.
  • the probes may be short, e.g., in the range of about 8-30 base pairs, since the hybrid will be relatively stable under even highly stringent conditions. If some degree of mismatch is expected with the probe, i.e., if it is suspected that the probe will hybridize to a variant region, a longer probe may be employed which hybridizes to the target sequence with the requisite specificity.
  • the probes will include an isolated polynucleotide attached to a label or reporter molecule and may be used to isolate other polynucleotide sequences, having sequence similarity by standard methods. For techniques for preparing and labeling probes see, e.g., Sambrook et al, 1989 or Ausubel et al, 1992. Other similar polynucleotides may be selected by using homologous polynucleotides. Alternatively, polynucleotides encoding these or similar polypeptides may be synthesized or selected by use of the redundancy in the genetic code. Various codon substitutions may be introduced, e.g., by silent changes (thereby producing various restriction sites) or to optimize expression for a particular system. Mutations may be introduced to modify the properties of the polypeptide, perhaps to change ligand-binding affinities, interchain affinities, or the polypeptide degradation or turnover rate.
  • Probes comprising synthetic oligonucleotides or other polynucleotides of the present invention may be derived from naturally occurring or recombinant single- or double-stranded polynucleotides, or be chemically synthesized. Probes may also be labeled by nick translation, Klenow fill-in reaction, or other methods known in the art. Portions of the polynucleotide sequence having at least about eight nucleotides, usually at least about 15 nucleotides, and fewer than about 6 kb, usually fewer than about 1.0 kb, from a polynucleotide sequence encoding 07CG27 are preferred as probes.
  • this definition includes probes of 8, 12, 15, 20, 25, 40, 60, 80, 100, 200, 300, 400 or 500 nucleotides or probes having any number of nucleotides within these ranges of values (e.g., 9, 10, 11, 16, 23, 30, 38, 50, 72, 121, etc., nucleotides), or probes having more than 500 nucleotides.
  • the probes may also be used to determine whether mRNA encoding 07CG27 is present in a cell or tissue.
  • the present invention includes all novel probes having at least 8 nucleotides derived from SEQ ID NO:l, its complement or functionally equivalent nucleic acid sequences.
  • the present invention does not include probes which exist in the prior art. That is, the present invention includes all probes having at least 8 nucleotides derived from SEQ ID NO: 1 with the proviso that they do not include probes existing in the prior art.
  • primers which may be used for the amplification of all or part of the 07CG27 gene.
  • a definition for primers includes primers of 8, 12, 15, 20, 25, 40, 60, 80, 100, 200, 300, 400, 500 nucleotides, or primers having any number of nucleotides within these ranges of values (e.g., 9, 10, 11, 16, 23, 30, 38, 50, 72, 121, etc. nucleotides), or primers having more than 500 nucleotides, or any number of nucleotides between 500 and 9000.
  • the primers may also be used to determine whether mRNA encoding 07CG27 is present in a cell or tissue.
  • the present invention includes all novel primers having at least 8 nucleotides derived from the 07CG27 locus for amplifying the 07CG27 gene, its complement or functionally equivalent nucleic acid sequences.
  • the present invention does not include primers which exist in the prior art. That is, the present invention includes all primers having at least 8 nucleotides with the proviso that it does not include primers existing in the prior art.
  • Protein modifications or fragments are provided by the present invention for 07CG27 polypeptides or fragments thereof which are substantially homologous to primary structural sequence but which include, e.g., in vivo or in vitro chemical and biochemical modifications or which incorporate unusual amino acids. Such modifications include, for example, acetylation, carboxylation, phosphorylation, glycosylation, ubiquitination, labeling, e.g., with radionuclides, and various enzymatic modifications, as will be readily appreciated by those well skilled in the art.
  • a variety of methods for labeling polypeptides and of substituents or labels useful for such purposes are well known in the art, and include radioactive isotopes such as P, ligands which bind to labeled antiligands (e.g., antibodies), fluorophores, chemiluminescent agents, enzymes, and antiligands which can serve as specific binding pair members for a labeled ligand.
  • radioactive isotopes such as P
  • ligands which bind to labeled antiligands e.g., antibodies
  • fluorophores e.g., fluorophores
  • chemiluminescent agents e.g., enzymes
  • antiligands which can serve as specific binding pair members for a labeled ligand.
  • the choice of label depends on the sensitivity required, ease of conjugation with the primer, stability requirements, and available instrumentation.
  • Methods of labeling polypeptides are well known in the art. See Sambrook et al, 1989 or
  • the present invention provides for biologically active fragments of the polypeptides.
  • Significant biological activities include ligand-binding, immunological activity and other biological activities characteristic of 07CG27 polypeptides.
  • Immunological activities include both immunogenic function in a target immune system, as well as sharing of immunological epitopes for binding, serving as either a competitor or substitute antigen for an epitope of the 07CG27 protein.
  • epitope refers to an antigenic determinant of a polypeptide.
  • An epitope could comprise three amino acids in a spatial conformation which is unique to the epitope. Generally, an epitope consists of at least five such amino acids, and more usually consists of at least 8-10 such amino acids. Methods of determining the spatial conformation of such amino acids are known in the art.
  • tandem-repeat polypeptide segments may be used as immunogens, thereby producing highly antigenic proteins.
  • polypeptides will serve as highly efficient competitors for specific binding. Production of antibodies specific for 07CG27 polypeptides or fragments thereof is described below.
  • the present invention also provides for fusion polypeptides, comprising 07CG27 polypeptides and fragments.
  • Homologous polypeptides may be fusions between two or more 07CG27 polypeptide sequences or between the sequences of 07CG27 and a related protein.
  • heterologous fusions may be constructed which would exhibit a combination of properties or activities of the derivative proteins. For example, ligand-binding or other domains may be "swapped" between different new fusion polypeptides or fragments.
  • Such homologous or heterologous fusion polypeptides may display, for example, altered strength or specificity of binding.
  • Fusion partners include immunoglobulins, bacterial b-galactosidase, trpE, protein A, b- lactamase, alpha amylase, alcohol dehydrogenase and yeast alpha mating factor. See Godowski et al, 1988.
  • Fusion proteins will typically be made by either recombinant nucleic acid methods, as described below, or may be chemically synthesized. Techniques for the synthesis of polypeptides are described, for example, in Merrifield, 1963.
  • Protein purification refers to various methods for the isolation of the 07CG27 polypeptides from other biological material, such as from cells transformed with recombinant nucleic acids encoding 07CG27, and are well known in the art.
  • polypeptides may be purified by immunoaffinity chromatography employing, e.g., the antibodies provided by the present invention.
  • immunoaffinity chromatography employing, e.g., the antibodies provided by the present invention.
  • Various methods of protein purification are well known in the art, and include those described in Deutscher, 1990 and Scopes, 1982.
  • isolated is substantially pure when at least about 60 to 75% of a sample exhibits a single polypeptide sequence.
  • a substantially pure protein will typically comprise about 60 to 90% W/W of a protein sample, more usually about 95%, and preferably will be over about 99% pure.
  • Protein purity or homogeneity may be indicated by a number of means well known in the art, such as polyacrylamide gel electrophoresis of a protein sample, followed by visualizing a single polypeptide band upon staining the gel. For certain purposes, higher resolution may be provided by using HPLC or other means well known in the art which are utilized for purification.
  • a 07CG27 protein is substantially free of naturally associated components when it is separated from the native contaminants which accompany it in its natural state.
  • a polypeptide which is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be substantially free from its naturally associated components.
  • a protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art.
  • a polypeptide produced as an expression product of an isolated and manipulated genetic sequence is an "isolated polypeptide," as used herein, even if expressed in a homologous cell type. Synthetically made forms or molecules expressed by heterologous cells are inherently isolated molecules.
  • Recombinant nucleic acid is a nucleic acid which is not naturally occurring, or which is made by the artificial combination of two otherwise separated segments of sequence. This artificial combination is often accomplished by either chemical synthesis means, or by the artificial manipulation of isolated segments of nucleic acids, e.g., by genetic engineering techniques. Such is usually done to replace a codon with a redundant codon encoding the same or a conservative amino acid, while typically introducing or removing a sequence recognition site. Alternatively, it is performed to join together nucleic acid segments of desired functions to generate a desired combination of functions.
  • regulatory sequences refers to those sequences normally within 100 kb of the coding region of a locus, but they may also be more distant from the coding region, which affect the expression of the gene (including transcription of the gene, and translation, splicing, stability or the like of the messenger RNA).
  • nucleic acid or fragment thereof is “substantially homologous” ("or substantially similar") to another if, when optimally aligned (with appropriate nucleotide insertions or deletions) with the other nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 60% of the nucleotide bases, usually at least about 70%, more usually at least about 80%, preferably at least about 90%), and more preferably at least about 95-98% of the nucleotide bases. Identity means the degree of sequence relatedness between two polypeptide or two polynucleotides sequences as determined by the identity of the match between two strings of such sequences. Identity can be readily calculated.
  • identity is well known to skilled artisans (Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M.
  • nucleic acid or fragment thereof will hybridize to another nucleic acid (or a complementary strand thereof) under selective hybridization conditions, to a strand, or to its complement.
  • Selectivity of hybridization exists when hybridization which is substantially more selective than total lack of specificity occurs.
  • selective hybridization will occur when there is at least about 55% homology over a stretch of at least about 14 nucleotides, preferably at least about 65%, more preferably at least about 75%, and most preferably at least about 90%. See, Kanehisa, 1984.
  • the length of homology comparison, as described, may be over longer stretches, and in certain embodiments will often be over a stretch of at least about nine nucleotides, usually at least about 20 nucleotides, more usually at least about 24 nucleotides, typically at least about 28 nucleotides, more typically at least about 32 nucleotides, and preferably at least about 36 or more nucleotides.
  • Nucleic acid hybridization will be affected by such conditions as salt concentration, temperature, or organic solvents, in addition to the base composition, length of the complementary strands, and the number of nucleotide base mismatches between the hybridizing nucleic acids, as will be readily appreciated by those skilled in the art.
  • Stringent temperature conditions will generally include temperatures in excess of 30°C, typically in excess of 37°C, and preferably in excess of 45°C.
  • Stringent salt conditions will ordinarily be less than 1000 mM, typically less than 500 mM, and preferably less than 200 mM. However, the combination of parameters is much more important than the measure of any single parameter. See, e.g., Wetmur and Davidson, 1968. See also Ausubel et al, 1992.
  • Probe sequences may also hybridize specifically to duplex DNA under certain conditions to form triplex or other higher order DNA complexes. The preparation of such probes and suitable hybridization conditions are well known in the art.
  • substantially homology when referring to polypeptides, indicate that the polypeptide or protein in question exhibits at least about 30% identity with an entire naturally-occurring protein or a portion thereof, usually at least about 70% identity, and preferably at least about 95% identity.
  • homology for polypeptides, is typically measured using sequence analysis software. See, e.g., the Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 910 University Avenue, Madison, Wisconsin 53705 and the references cited above with respect to nucleic acid homology. Protein analysis software matches similar sequences using measures of homology assigned to various substitutions, deletions and other modifications. Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
  • Methods commonly employed to determine identity between two sequences include, but are not limited to those disclosed in Guide to Huge Computers, Martin J. Bishop, ed., Academic Press, San Diego, 1994, and Carillo, H., and Lipman, D. (1988). Preferred methods to determine identity are designed to give the largest match between the two sequences tested. Such methods are codified in computer programs. Preferred computer program methods to determine identity between two sequences include, but are not limited to, GCG program package (Devereux et al. (1984), BLASTP, BLASTN, FASTA (Atschul et al. (1990)).
  • Substantially similar function refers to the function of a modified nucleic acid or a modified protein, with reference to the wild-type 07CG27 nucleic acid or wild-type 07CG27 polypeptide.
  • the modified polypeptide will be substantially homologous to the wild-type 07CG27 polypeptide and will have substantially the same function.
  • the modified polypeptide may have an altered amino acid sequence and/or may contain modified amino acids.
  • the modified polypeptide may have other useful properties, such as a longer half-life.
  • the similarity of function (activity) of the modified polypeptide may be substantially the same as the activity of the wild-type 07CG27 polypeptide.
  • the similarity of function (activity) of the modified polypeptide may be higher than the activity of the wild-type 07CG27 polypeptide.
  • the modified polypeptide is synthesized using conventional techniques, or is encoded by a modified nucleic acid and produced using conventional techniques.
  • the modified nucleic acid is prepared by conventional techniques.
  • a nucleic acid with a function substantially similar to the wild-type 07CG27 gene function produces the modified protein described above.
  • a polypeptide "fragment,” “portion” or “segment” is a stretch of amino acid residues of at least about five to seven contiguous amino acids, often at least about seven to nine contiguous amino acids, typically at least about nine to 13 contiguous amino acids and, most preferably, at least about 20 to 30 or more contiguous amino acids.
  • polypeptides of the present invention may be coupled to a solid-phase support, e.g., nitrocellulose, nylon, column packing materials (e.g., Sepharose beads), magnetic beads, glass wool, plastic, metal, polymer gels, cells, or other substrates.
  • a solid-phase support e.g., nitrocellulose, nylon, column packing materials (e.g., Sepharose beads), magnetic beads, glass wool, plastic, metal, polymer gels, cells, or other substrates.
  • Such supports may take the form, for example, of beads, wells, dipsticks, or membranes.
  • Target region refers to a region of the nucleic acid which is amplified and/or detected.
  • target sequence refers to a sequence with which a probe or primer will form a stable hybrid under desired conditions.
  • polynucleotides of the present invention may be produced by replication in a suitable host cell.
  • Natural or synthetic polynucleotide fragments coding for a desired fragment will be incorporated into recombinant polynucleotide constructs, usually DNA constructs, capable of introduction into and replication in a prokaryotic or eukaryotic cell.
  • polynucleotide constructs will be suitable for replication in a unicellular host, such as yeast or bacteria, but may also be intended for introduction to (with and without integration within the genome) cultured mammalian or plant or other eukaryotic cell lines.
  • a unicellular host such as yeast or bacteria
  • the purification of nucleic acids produced by the methods of the present invention is described, e.g., in
  • the polynucleotides of the present invention may also be produced by chemical synthesis, e.g., by the phosphoramidite method described by Beaucage and Carruthers, 1981 or the triester method according to Matteucci and Caruthers, 1981, and may be performed on commercial, automated oligonucleotide synthesizers.
  • a double-stranded fragment may be obtained from the single-stranded product of chemical synthesis either by synthesizing the complementary strand and annealing the strands together under appropriate conditions or by adding the complementary strand using DNA polymerase with an appropriate primer sequence.
  • Polynucleotide constructs prepared for introduction into a prokaryotic or eukaryotic host may comprise a replication system recognized by the host, including the intended polynucleotide fragment encoding the desired polypeptide, and will preferably also include transcription and translational initiation regulatory sequences operably linked to the polypeptide encoding segment.
  • Expression vectors may include, for example, an origin of replication or autonomously replicating sequence (ARS) and expression control sequences, a promoter, an enhancer and necessary processing information sites, such as ribosome-binding sites, RNA splice sites, polyadenylation sites, transcriptional terminator sequences, and mRNA stabilizing sequences.
  • ARS origin of replication or autonomously replicating sequence
  • Secretion signals may also be included where appropriate, whether from a native 07CG27 protein or from other receptors or from secreted polypeptides of the same or related species, which allow the protein to cross and/or lodge in cell membranes, and thus attain its functional topology, or be secreted from the cell.
  • Such vectors may be prepared by means of standard recombinant techniques well known in the art and discussed, for example, in Sambrook et al, 1989 or Ausubel et al. 1992.
  • An appropriate promoter and other necessary vector sequences will be selected so as to be functional in the host, and may include, when appropriate, those naturally associated with 07CG27 genes. Examples of workable combinations of cell lines and expression vectors are described in Sambrook et al, 1989 or Ausubel et al, 1992; see also, e.g., Metzger et al, 1988. Many useful vectors are known in the art and may be obtained from such vendors as Stratagene, New England BioLabs, Promega Biotech, and others. Promoters such as the tip, lac and phage promoters, tRNA promoters and glycolytic enzyme promoters may be used in prokaryotic hosts.
  • Useful yeast promoters include promoter regions for metallothionein, 3-phosphoglycerate kinase or other glycolytic enzymes such as enolase or glyceraldehyde-3-phosphate dehydrogenase, enzymes responsible for maltose and galactose utilization, and others. Vectors and promoters suitable for use in yeast expression are further described in Hitzeman et al, EP 73, 675 A.
  • Non-native mammalian promoters might include the early and late promoters from SV40 (Fiers et al, 1978) or promoters derived from murine Moloney leukemia virus, mouse tumor virus, avian sarcoma viruses, adenovirus II, bovine papilloma virus or polyoma.
  • the construct may be joined to an amplifiable gene (e.g., DHFR) so that multiple copies of the gene may be made.
  • DHFR e.g., DHFR
  • Enhancers and Eukaryotic Gene Expression Cold Spring Harbor Press, Cold Spring Harbor, New York (1983). See also, e.g., U.S. Patent Nos. 5,691,198; 5,735,500; 5,747,469 and 5,436,146.
  • expression vectors may replicate autonomously, they may also replicate by being inserted into the genome of the host cell, by methods well known in the art.
  • Expression and cloning vectors will likely contain a selectable marker, a gene encoding a protein necessary for survival or growth of a host cell transformed with the vector. The presence of this gene ensures growth of only those host cells which express the inserts.
  • Typical selection genes encode proteins that a) confer resistance to antibiotics or other toxic substances, e.g. ampicillin, neomycin, methotrexate, etc.; b) complement auxotrophic deficiencies, or c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
  • the choice of the proper selectable marker will depend on the host cell, and appropriate markers for different hosts are well known in the art.
  • the vectors containing the nucleic acids of interest can be transcribed in vitro, and the resulting RNA introduced into the host cell by well-known methods, e.g., by injection (see, Kubo et al, 1988), or the vectors can be introduced directly into host cells by methods well known in the art, which vary depending on the type of cellular host, including electroporation; transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; infection (where the vector is an infectious agent, such as a retroviral genome); and other methods. See generally, Sambrook et al, 1989 and Ausubel et al, 1992.
  • the introduction of the polynucleotides into the host cell by any method known in the art, including, inter alia, those described above, will be referred to herein as "transformation.”
  • the cells into which have been introduced nucleic acids described above are meant to also include the progeny of such cells.
  • Large quantities of the nucleic acids and polypeptides of the present invention may be prepared by expressing the 07CG27 nucleic acids or portions thereof in vectors or other expression vehicles in compatible prokaryotic or eukaryotic host cells.
  • the most commonly used prokaryotic hosts are strains of Escherichia coli, although other prokaryotes, such as Bacillus subtilis or Pseudomonas may also be used.
  • Mammalian or other eukaryotic host cells such as those of yeast, filamentous fungi, plant, insect, or amphibian or avian species, may also be useful for production of the proteins of the present invention. Propagation of mammalian cells in culture is per se well known. See, Jakoby and Pastan, 1979. Examples of commonly used mammalian host cell lines are VERO and HeLa cells, Chinese hamster ovary (CHO) cells, and WI38, BHK, and COS cell lines. An example of a commonly used insect cell line is SF9. However, it will be appreciated by the skilled practitioner that other cell lines may be appropriate, e.g., to provide higher expression, desirable glycosylation patterns, or other features.
  • Clones are selected by using markers depending on the mode of the vector construction.
  • the marker may be on the same or a different DNA molecule, preferably the same DNA molecule.
  • the transformant may be selected, e.g., by resistance to ampicillin, tetracycline or other antibiotics. Production of a particular product based on temperature sensitivity may also serve as an appropriate marker.
  • Prokaryotic or eukaryotic cells transformed with the polynucleotides of the present invention will be useful not only for the production of the nucleic acids and polypeptides of the present invention, but also, for example, in studying the characteristics of 07CG27 polypeptides.
  • Antisense polynucleotide sequences are useful in preventing or diminishing the expression of the 07CG27 locus, as will be appreciated by those skilled in the art.
  • polynucleotide vectors containing all or a portion of the 07CG27 locus or other sequences from the 07CG27 region (particularly those flanking the 07CG27 locus) may be placed under the control of a promoter in an antisense orientation and introduced into a cell. Expression of such an antisense construct within a cell will interfere with 07CG27 transcription and/or translation and/or replication.
  • the probes and primers based on the 07CG27 gene sequences disclosed herein are used to identify homologous 07CG27 gene sequences and proteins in other species. These 07CG27 gene sequences and proteins are used in the diagnostic/prognostic, therapeutic and drug screening methods described herein for the species from which they have been isolated. Methods of Use: Nucleic Acid Diagnosis and Diagnostic Kits
  • a biological sample such as blood is prepared and analyzed for the presence or absence of predisposing alleles of 07CG27. Results of these tests and interpretive information are returned to the health care provider for communication to the tested individual. Such diagnoses may be performed by diagnostic laboratories, or, alternatively, diagnostic kits are manufactured and sold to health care providers or to private individuals for self-diagnosis. Initially, the screening method involves amplification of the relevant 07CG27 sequences.
  • the screening method involves a non-PCR based strategy.
  • Such screening methods include two-step label amplification methodologies that are well known in the art. Both PCR and non-PCR based screening strategies can detect target sequences with a high level of sensitivity.
  • the most popular method used today is target amplification.
  • the target nucleic acid sequence is amplified with polymerases.
  • One particularly preferred method using polymerase- driven amplification is the polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • the polymerase chain reaction and other polymerase-driven amplification assays can achieve over a million-fold increase in copy number through the use of polymerase-driven amplification cycles.
  • the resulting nucleic acid can be sequenced or used as a substrate for DNA probes.
  • the biological sample to be analyzed such as blood or serum
  • the biological sample to be analyzed may be treated, if desired, to extract the nucleic acids.
  • the sample nucleic acid may be prepared in various ways to facilitate detection of the target sequence; e.g. denaturation, restriction digestion, electrophoresis or dot blotting.
  • the targeted region of the analyte nucleic acid usually must be at least partially single-stranded to form hybrids with the targeting sequence of the probe. If the sequence is naturally single-stranded, denaturation will not be required. However, if the sequence is double-stranded, the sequence will probably need to be denatured.
  • Denaturation can be carried out by various techniques known in the art. Analyte nucleic acid and probe are incubated under conditions which promote stable hybrid formation of the target sequence in the probe with the putative targeted sequence in the analyte.
  • the region of the probes which is used to bind to the analyte can be made completely complementary to the targeted region of human chromosome 1. Therefore, high stringency conditions are desirable in order to prevent false positives. However, conditions of high stringency are used only if the probes are complementary to regions of the chromosome which are unique in the genome.
  • the stringency of hybridization is determined by a number of factors during hybridization and during the washing procedure, including temperature, ionic strength, base composition, probe length, and concentration of formamide.
  • the formation of higher order hybrids such as triplexes, quadraplexes, etc., may be desired to provide the means of detecting target sequences. Detection, if any, of the resulting hybrid is usually accomplished by the use of labeled probes. Alternatively, the probe may be unlabeled, but may be detectable by specific binding with a ligand which is labeled, either directly or indirectly.
  • Suitable labels, and methods for labeling probes and ligands are known in the art, and include, for example, radioactive labels which may be incorporated by known methods (e.g., nick translation, random priming or kinasing), biotin, fluorescent groups, chemilummescent groups (e.g., dioxetanes, particularly triggered dioxetanes), enzymes, antibodies and the like. Variations of this basic scheme are known in the art, and include those variations that facilitate separation of the hybrids to be detected from extraneous materials and/or that amplify the signal from the labeled moiety.
  • non-PCR based screening assays are also contemplated in this invention.
  • This procedure hybridizes a nucleic acid probe (or an analog such as a methyl phosphonate backbone replacing the normal phosphodiester), to the low level DNA target.
  • This probe may have an enzyme covalently linked to the probe, such that the covalent linkage does not interfere with the specificity of the hybridization.
  • This enzyme-probe-conjugate-target nucleic acid complex can then be isolated away from the free probe enzyme conjugate and a substrate is added for enzyme detection. Enzymatic activity is observed as a change in color development or luminescent output resulting in a 10 -10 increase in sensitivity.
  • Jablonski et al, 1986 For an example relating to the preparation of oligodeoxynucleotide-alkaline phosphatase conjugates and their use as hybridization probes see Jablonski et al, 1986.
  • Two-step label amplification methodologies are known in the art. These assays work on the principle that a small ligand (such as digoxigenin, biotin, or the like) is attached to a nucleic acid probe capable of specifically binding 07CG27. Allele specific probes are also contemplated within the scope of this example and exemplary allele specific probes include probes encompassing the predisposing or potentially predisposing mutations summarized in herein.
  • the small ligand attached to the nucleic acid probe is specifically recognized by an antibody-enzyme conjugate.
  • digoxigenin is attached to the nucleic acid probe. Hybridization is detected by an antibody-alkaline phosphatase conjugate which turns over a chemilummescent substrate.
  • the small ligand is recognized by a second ligand-enzyme conjugate that is capable of specifically complexing to the first ligand.
  • a well known embodiment of this example is the biotin-avidin type of interactions. For methods for labeling nucleic acid probes and their use in biotin-avidin based assays see Rigby et al, 1977 and Nguyen et al, 1992.
  • nucleic acid probe assays of this invention will employ a cocktail of nucleic acid probes capable of detecting 07CG27.
  • a cocktail of nucleic acid probes capable of detecting 07CG27 in one example to detect the presence of 07CG27 in a cell sample, more than one probe complementary to 07CG27 is employed and in particular the number of different probes is alternatively 2, 3, or 5 different nucleic acid probe sequences.
  • the cocktail includes probes capable of binding to the allele-specific mutations identified in populations of patients with alterations in 07CG27.
  • any number of probes can be used, and will preferably include probes corresponding to the major gene mutations identified as predisposing an individual to prostate cancer.
  • Some candidate probes contemplated within the scope of the invention include probes that include the allele-specific mutations identified herein and those that have the 07CG27 regions corresponding to SEQ ID NO:l both 5' and 3' to a mutation site.
  • the neoplastic condition of lesions can also be detected on the basis of the alteration of wild-type 07CG27 polypeptide. Such alterations can be determined by sequence analysis in accordance with conventional techniques. More preferably, antibodies (polyclonal or monoclonal) are used to detect differences in, or the absence of, 07CG27 peptides.
  • the antibodies may be prepared as discussed above under the heading "Antibodies” and as further shown in the Examples infra. Other techniques for raising and purifying antibodies are well known in the art and any such techniques may be chosen to achieve the preparations claimed in this invention.
  • antibodies will immunoprecipitate 07CG27 proteins from solution as well as react with 07CG27 protein on Western or immunoblots of polyacrylamide gels.
  • antibodies will detect 07CG27 proteins in paraffin or frozen tissue sections, using immunocytochemical techniques.
  • Preferred embodiments relating to methods for detecting 07CG27 or its mutations include enzyme linked immunosorbent assays (ELISA), radioimmunoassays (RIA), immunoradiometric assays (IRMA) and immunoenzymatic assays (IEMA), including sandwich assays using monoclonal and/or polyclonal antibodies. Exemplary sandwich assays are described by David et al. in U.S. Patent Nos. 4,376,110 and 4,486,530, hereby incorporated by reference, and exemplified in Example 15.
  • Drug Screening This invention is particularly useful for screening compounds by using a wild-type or mutant 07CG27 polypeptide or binding fragment thereof in any of a variety of drug screening techniques.
  • the 07CG27 polypeptide or fragment employed in such a test may either be free in solution, affixed to a solid support, or borne on a cell surface.
  • One method of drug screening utilizes eucaryotic or procaryotic host cells which are stably transformed with recombinant polynucleotides expressing the polypeptide or fragment, preferably in competitive binding assays. Such cells, either in viable or fixed form, can be used for standard binding assays.
  • One may measure, for example, for the formation of complexes between an 07CG27 polypeptide or fragment and the agent being tested, or examine the degree to which the formation of a complex between an 07CG27 polypeptide or fragment and a known ligand, e.g.
  • the present invention provides methods of screening for drugs comprising contacting such an agent with an 07CG27 polypeptide or fragment thereof and assaying (i) for the presence of a complex between the agent and the 07CG27 polypeptide or fragment, or (ii) for the presence of a complex between the 07CG27 polypeptide or fragment and a ligand, by methods well known in the art. In such competitive binding assays the 07CG27 polypeptide or fragment is typically labeled.
  • Free 07CG27 polypeptide or fragment is separated from that present in a proteimprotein complex, and the amount of free (i.e., uncomplexed) label is a measure of the binding of the agent being tested to 07CG27 or its interference with 07CG27:ligand binding, respectively. Since 07CG27 is a GEF protein and activates ras, the effect of a drug candidate can be determined by measuring ras activation by a wild-type or mutant 07CG27 polypeptide.
  • Another technique for drug screening provides high throughput screening for compounds having suitable binding affinity to the 07CG27 polypeptides and is described in detail in Geysen, PCT published application WO 84/03564, published on September 13, 1984. Briefly stated, large numbers of different small peptide test compounds are synthesized on a solid substrate, such as plastic pins or some other surface. The peptide test compounds are reacted with 07CG27 polypeptide and washed. Bound 07CG27 polypeptide is then detected by methods well known in the art.
  • Purified 07CG27 can be coated directly onto plates for use in the aforementioned drug screening techniques. However, non-neutralizing antibodies to the polypeptide can be used to capture antibodies to immobilize the 07CG27 polypeptide on the solid phase.
  • This invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of specifically binding the 07CG27 polypeptide compete with a test compound for binding to the 07CG27 polypeptide or fragments thereof. In this manner, the antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants of the 07CG27 polypeptide.
  • a further technique for drug screening involves the use of host eukaryotic cell lines or cells (such as described above) which have a wild-type or mutant 07CG27 gene.
  • the host cell lines or cells are grown in the presence of drug compound.
  • the ras activation occurring in the host cells is measured to determine if the compound is capable of regulating the interaction of 07CG27 and ras.
  • a method of screening for a substance which modulates activity of a polypeptide may include contacting one or more test substances with the polypeptide in a suitable reaction medium, testing the activity of the treated polypeptide and comparing that activity with the activity of the polypeptide in comparable reaction medium untreated with the test substance or substances. A difference in activity between the treated and untreated polypeptides is indicative of a modulating effect of the relevant test substance or substances.
  • test substances Prior to or as well as being screened for modulation of activity, test substances may be screened for ability to interact with the polypeptide, e.g., in a yeast two-hybrid system (e.g., Bartel et al., 1993; Fields and Song, 1989; Chevray and Nathans, 1992; Lee et al, 1995).
  • This system may be used as a coarse screen prior to testing a substance for actual ability to modulate activity of the polypeptide.
  • the screen could be used to screen test substances for binding to an 07CG27 specific binding partner, or to find mimetics of an 07CG27 polypeptide.
  • Rational Drug Design The goal of rational drug design is to produce structural analogs of biologically active polypeptides of interest or of small molecules with which they interact (e.g., agonists, antagonists, inhibitors) in order to fashion drugs which are, for example, more active or stable forms of the polypeptide, or which, e.g., enhance or interfere with the function of a polypeptide in vivo. See, e.g., Hodgson, 1991.
  • one first determines the three-dimensional structure of a protein of interest (e.g., 07CG27 polypeptide) or, for example, of the 07CG27- receptor or ligand complex, by x-ray crystallography, by computer modeling or most typically, by a combination of approaches.
  • peptides e.g., 07CG27 polypeptide
  • alanine scan Wells, 1991
  • an amino acid residue is replaced by Ala, and its effect on the peptide's activity is determined.
  • Each of the amino acid residues of the peptide is analyzed in this manner to determine the important regions of the peptide.
  • improved 07CG27 polypeptide activity or stability By virtue of the availability of cloned 07CG27 sequences, sufficient amounts of the 07CG27 polypeptide may be made available to perform such analytical studies as x-ray crystallography.
  • the knowledge of the 07CG27 protein sequence provided herein will guide those employing computer modeling techniques in place of, or in addition to x-ray crystallography.
  • the substance may be investigated further. Furthermore, it may be manufactured and/or used in preparation, i.e., manufacture or formulation, or a composition such as a medicament, pharmaceutical composition or drug. These may be administered to individuals.
  • the present invention extends in various aspects not only to a substance identified using a nucleic acid molecule as a modulator of polypeptide activity, in accordance with what is disclosed herein, but also a pharmaceutical composition, medicament, drug or other composition comprising such a substance, a method comprising administration of such a composition comprising such a substance, a method comprising administration of such a composition to a patient, e.g., for treatment of prostate cancer, use of such a substance in the manufacture of a composition for administration, e.g., for treatment of prostate cancer, and a method of making a pharmaceutical composition comprising admixing such a substance with a pharmaceutically acceptable excipient, vehicle or carrier, and optionally other ingredients.
  • a substance identified as a modulator of polypeptide function may be peptide or non- peptide in nature.
  • Non-peptide "small molecules" are often preferred for many in vivo pharmaceutical uses. Accordingly, a mimetic or mimic of the substance (particularly if a peptide) may be designed for pharmaceutical use.
  • the designing of mimetics to a known pharmaceutically active compound is a known approach to the development of pharmaceuticals based on a " lead" compound. This might be desirable where the active compound is difficult or expensive to synthesize or where it is unsuitable for a particular method of administration, e.g., pure peptides are unsuitable active agents for oral compositions as they tend to be quickly degraded by proteases in the alimentary canal.
  • Mimetic design, synthesis and testing is generally used to avoid randomly screening large numbers of molecules for a target property.
  • the pharmacophore Once the pharmacophore has been found, its structure is modeled according to its physical properties, e.g., stereochemistry, bonding, size and/or charge, using data from a range of sources, e.g., spectroscopic techniques, x-ray diffraction data and NMR. Computational analysis, similarity mapping (which models the charge and/or volume of a pharmacophore, rather than the bonding between atoms) and other techniques can be used in this modeling process.
  • a range of sources e.g., spectroscopic techniques, x-ray diffraction data and NMR.
  • Computational analysis, similarity mapping which models the charge and/or volume of a pharmacophore, rather than the bonding between atoms
  • other techniques can be used in this modeling process.
  • the three-dimensional structure of the ligand and its binding partner are modeled. This can be especially useful where the ligand and/or binding partner change conformation on binding, allowing the model to take account of this in the design of the mimetic.
  • a template molecule is then selected onto which chemical groups which mimic the pharmacophore can be grafted.
  • the template molecule and the chemical groups grafted onto it can conveniently be selected so that the mimetic is easy to synthesize, is likely to be pharmacologically acceptable, and does not degrade in vivo, while retaining the biological activity of the lead compound.
  • the mimetic is peptide-based
  • further stability can be achieved by cyclizing the peptide, increasing its rigidity.
  • the mimetic or mimetics found by this approach can then be screened to see whether they have the target property, or to what extent they exhibit it. Further optimization or modification can then be carried out to arrive at one or more final mimetics for in vivo or clinical testing.
  • the present invention contemplates an antisense polynucleotide up to about 50 nucleotides in length that hybridizes with mRNA molecules that encode a 07CG27 polypeptide, and the use of one or more of those polynucleotides in treating disease. See U.S. Patent Nos. 5,891,858 and 5,885,970, incorporated herein by reference, for further details.
  • the antisense polynucleotide is useful for treating disease caused by a mutant 07CG27 as well as overexpression of a wild-type 07CG27.
  • an antisense polynucleotide is contacted with a cancer cells.
  • the contact is carried out in vivo in a host mammal, and contact is effected by administration to the mammal of a pharmaceutical composition containing the polynucleotide dissolved or dispersed in a physiologically tolerable diluent so that a body fluid such as blood or lymph provides at least a portion of the aqueous medium.
  • a body fluid such as blood or lymph provides at least a portion of the aqueous medium.
  • In vivo contact is maintained until the polynucleotide is eliminated from the mammal's body by a normal bodily function such as excretion in the urine or feces or enzymatic breakdown.
  • a polynucleotide can also be administered in the form of liposomes.
  • liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by monoor multi-lamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used.
  • the present compositions in liposome form can contain stabilizers, preservatives, excipients, and the like in addition to the agent.
  • the preferred lipids are phospholipids and phosphatidyl cholines (lecithins), both natural and synthetic.
  • An antisense polynucleotide can also be administered by gene therapy.
  • the polynucleotide may be introduced into the cell in a vector such that the polynucleotide remains extrachromosomal. In such a situation, the polynucleotide will be expressed by the cell from the extrachromosomal location.
  • Vectors for introduction of polyucleotides for extrachromosomal maintenance are known in the art, and any suitable vector may be used.
  • Methods for introducing DNA into cells such as electroporation, calcium phosphate coprecipitation and viral transduction are known in the art, and the choice of method is within the competence of the routineer.
  • the antisense polynucleotide may be employed in gene therapy methods in order to decrease the amount of the expression products of 07CG27 in disease cells, especially in those cases where 07CG27 is overexpressed. Such gene therapy is particularly appropriate for use in both disease and pre-disease cells. It may also be useful to decrease the level of expression of 07CG27 even in those disease cells in which a wild-type gene is expressed at an elevated level.
  • Gene therapy would be carried out according to generally accepted methods, for example, as described in further detail in U.S. Patent No. 5,747,282 and references cited therein, all incorporated by reference herein.
  • Expression vectors in the context of gene therapy are meant to include those constructs containing sequences sufficient to express a polynucleotide that has been cloned therein.
  • the construct contains viral sequences sufficient to support packaging of the construct. If the polynucleotide encodes an antisense polynucleotide or a ribozyme, expression will produce the antisense polynucleotide or ribozyme. Thus in this context, expression does not require that a protein product be synthesized.
  • the vector also contains a promoter functional in eukaryotic cells.
  • the cloned polynucleotide sequence is under control of this promoter. Suitable eukaryotic promoters include those described above.
  • the expression vector may also include sequences, such as selectable markers and other sequences conventionally used.
  • Receptor-mediated gene transfer is accomplished by the conjugation of DNA (usually in the form of covalently closed supercoiled plasmid) to a protein ligand via polylysine.
  • Ligands are chosen on the basis of the presence of the corresponding ligand receptors on the cell surface of the target cell/tissue type.
  • These ligand-DNA conjugates can be injected directly into the blood if desired and are directed to the target tissue where receptor binding and internalization of the DNA-protein complex occurs.
  • coinfection with adenovirus can be included to disrupt endosome function.
  • cells and animals which carry a mutant 07CG27 allele can be used as model systems to study and test for substances which have potential as therapeutic agents. These may be isolated from individuals with 07CG27 mutations, either somatic or germline. Alternatively, the cell line can be engineered to carry the mutation in the 07CG27 allele, as described above. After a test substance is applied to the cells, the phenotype of the cell is determined, using assays known in the art. Animals for testing therapeutic agents can be selected after mutagenesis of whole animals or after treatment of germline cells or zygotes.
  • Such treatments include insertion of mutant 07CG27 alleles, usually from a second animal species, as well as insertion of disrupted homologous genes.
  • the endogenous 07CG27 gene(s) of the animals may be disrupted by insertion or deletion mutation or other genetic alterations using conventional techniques (Capecchi, 1989; Valancius and Smithies, 1991; Hasty et al, 1991; Shinkai et al, 1992; Mombaerts et al, 1992; Philpott et al, 1992; Snouwaert et al, 1992; Donehower et al, 1992) to produce knockout or transplacement animals.
  • a transplacement is similar to a knockout because the endogenous gene is replaced, but in the case of a transplacement the replacement is by another version of the same gene.
  • transgenic animals are produced which contain a functional transgene encoding a functional 07CG27 polypeptide or variants thereof.
  • Transgenic animals expressing 07CG27 transgenes, recombinant cell lines derived from such animals and transgenic embryos may be useful in methods for screening for and identifying agents that induce or repress function of 07CG27.
  • Transgenic animals of the present invention also can be used as models for studying indications such as disease.
  • a 07CG27 transgene is introduced into a non-human host to produce a transgenic animal expressing a human or murine 07CG27 gene.
  • the transgenic animal is produced by the integration of the transgene into the genome in a manner that permits the expression of the transgene.
  • Methods for producing transgenic animals are generally described by Wagner and Hoppe (U.S. Patent No. 4,873,191; which is incorporated herein by reference), Brinster et al.
  • transgenic animals may be produced from the fertilized eggs from a number of animals including, but not limited to reptiles, amphibians, birds, mammals, and fish. Within a particularly preferred embodiment, transgenic mice are generated which overexpress 07CG27 or express a mutant form of the polypeptide.
  • the absence of a 07CG27 in "knockout" mice permits the study of the effects that loss of 07CG27 protein has on a cell in vivo. Knock-out mice also provide a model for the development of 07CG27-related cancers.
  • transgenic animals and cell lines derived from such animals may find use in certain testing experiments.
  • transgenic animals and cell lines capable of expressing wild-type or mutant 07CG27 may be exposed to test substances. These test substances can be screened for the ability to reduce overepression of wild-type 07CG27 or impair the expression or function of mutant 07CG27.
  • Pharmaceutical Compositions and Routes of Administration The 07CG27 polypeptides, antibodies, peptides and nucleic acids of the present invention can be formulated in pharmaceutical compositions, which are prepared according to conventional pharmaceutical compounding techniques. See, for example, Remington's Pharmaceutical Sciences. 18th Ed. (1990, Mack Publishing Co., Easton, PA).
  • the composition may contain the active agent or pharmaceutically acceptable salts of the active agent.
  • compositions may comprise, in addition to one of the active substances, a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known in the art. Such materials should be non- toxic and should not interfere with the efficacy of the active ingredient.
  • the carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., intravenous, oral, intrathecal, epineural or parenteral.
  • the compounds can be formulated into solid or liquid preparations such as capsules, pills, tablets, lozenges, melts, powders, suspensions or emulsions.
  • any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, suspending agents, and the like in the case of oral liquid preparations (such as, for example, suspensions, elixirs and solutions); or carriers such as starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations (such as, for example, powders, capsules and tablets).
  • tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be sugar-coated or enteric-coated by standard techniques.
  • the active agent can be encapsulated to make it stable to passage through the gastrointestinal tract while at the same time allowing for passage across the blood brain barrier. See for example, WO 96/11698.
  • the compound may be dissolved in a pharmaceutical carrier and administered as either a solution or a suspension.
  • suitable carriers are water, saline, dextrose solutions, fructose solutions, ethanol, or oils of animal, vegetative or synthetic origin.
  • the carrier may also contain other ingredients, for example, preservatives, suspending agents, solubilizing agents, buffers and the like.
  • the compounds When the compounds are being administered intrathecally, they may also be dissolved in cerebrospinal fluid.
  • the active agent is preferably administered in a therapeutically effective amount.
  • the actual amount administered, and the rate and time-course of administration, will depend on the nature and severity of the condition being treated. Prescription of treatment, e.g. decisions on dosage, timing, etc., is within the responsibility of general practitioners or specialists, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners. Examples of techniques and protocols can be found in Remington 's Pharmaceutical Sciences.
  • targeting therapies may be used to deliver the active agent more specifically to certain types of cell, by the use of targeting systems such as antibodies or cell specific ligands. Targeting may be desirable for a variety of reasons, e.g. if the agent is unacceptably toxic, or if it would otherwise require too high a dosage, or if it would not otherwise be able to enter the target cells.
  • these agents could be produced in the target cell, e.g. in a viral vector such as described above or in a cell based delivery system such as described in U.S. Patent No. 5,550,050 and published PCT application Nos. WO 92/19195, WO 94/25503, WO 95/01203, WO 95/05452, WO 96/02286, WO 96/02646, WO 96/40871, WO 96/40959 and WO 97/12635, designed for implantation in a patient.
  • the vector could be targeted to the specific cells to be treated, or it could contain regulatory elements which are more tissue specific to the target cells.
  • the cell based delivery system is designed to be implanted in a patient's body at the desired target site and contains a coding sequence for the active agent.
  • the agent could be administered in a precursor form for conversion to the active form by an activating agent produced in, or targeted to, the cells to be treated. See for example, EP 425,731 A and WO 90/07936.
  • Extensive cancer prone kindreds were ascertained from a defined population providing a large set of extended kindreds with multiple cases of prostate cancer and many relatives available to study for analysis of candidate genes in the HPCl region.
  • Each kindred was extended through all available connecting relatives, and to all informative first degree relatives of each proband or cancer case. For these kindreds, additional prostate cancer cases and individuals with cancer at other sites of interest (e.g., bladder) who also appeared in the kindreds were identified through the tumor registry linked files. All prostate cancers reported in the kindred which were not confirmed in the Utah Cancer Registry were verified. Medical records or death certificates were obtained for confirmation of all cancers. Each key connecting individual and all informative individuals were invited to participate by providing a blood sample from which DNA was extracted. We also sampled spouses, siblings, and offspring of deceased cases so that the genotype of the deceased cases could be inferred from the genotypes of their relatives.
  • Each of the Utah pedigrees studied represents the descendants of a single founder for whom a significant excess of prostate cancer cases was observed among all descendants. Since all affected descendants are studied, the resulting kindreds represent a collection of both closely and distantly related prostate cancer cases.
  • the criteria for selection of kindreds to analyze for 07CG27 linkage were: 1) genotypes available, or inferable, for 6 or more prostate cancer cases, and 2) at least 3 genotyped cases within a second degree of relationship to another genotyped case.
  • the Utah kindreds are 5 - 7 generations deep, and contain between 8 and 29 prostate cancer cases. They are all Caucasian of Northern European ancestry. The median age-of-onset for each kindred ranged from 64 to 76, similar to that estimated for the general population. Five percent of cases were diagnosed before age 55. For each kindred analyzed, the number of prostate cancer cases, the median age and range of age-of-onset, and the number of cases and family members sampled and included in this analysis were detailed. EXAMPLE 2 Selection of Kindreds Which are Linked to Chromosome 1 Nuclear pellets were extracted from 16 ml of ACD blood, and DNA extracted with phenol and chloroform, precipitated with ethanol, and resuspended in Tris-EDTA.
  • the markers used for genotyping were short tandem repeat (STR) loci at lq24-25 which flanked the most likely HPCl location as indicated in Smith et al. (1996).
  • the most likely location as suggested in Smith et al. (1996) is at D1S254.
  • Amplification of 20 ng genomic DNA was performed according to standard PCR procedures, with minor modifications to optimize product clarity, in a total reaction mix of 10 ml. Radiolabeled PCR products were electrophoresed on standard 6% polyacrylamide denaturing sequencing gels. Gels were then dried and autoradiographed. A total of over 200 prostate cancer cases and approximately 800 of their relatives were genotyped for the markers.
  • Multipoint linkage analysis was performed using VITESSE (O'Connell et al., 1995). The size of the pedigrees and the lack of genotyping of the higher generations due to the late age-of-onset, made more-than-three-point analyses impossible.
  • Multipoint linkage analysis resulted in a maximum heterogeneity Lod score of +1.20 at D1S254 with an estimate that 5% of kindreds were linked. Multipoint heterogeneity analysis in the most likely interval excluded linkage (Lod scores less than -2.00) for alpha greater than 0.33.
  • Cancers of sites other than prostate would also be expected to occur in individuals in these kindreds. Some individuals hypothesized to be sharing the segregating chromosome 1 haplotype were affected with cancer at another site. Lod scores for linkage for a phenotype of cancer of any site did not differ significantly from those for prostate alone, although most individuals with cancer of another site were not included in the sampling.
  • Genomic clone contig assembly in the HPCl region started from a publicly available integrated map of chromosome 1, the WICGR Chr 1 map of Nov. 19, 1996.
  • YACs located in the interval between D1S202 and D1S238 were ordered from Genome Systems.
  • Primer pairs for the markers located in the interval between D1S202 and D1S238 were synthesized and used to screen a BAC library at Myriad. Markers that were negative on that BAC library were used to screen the BAC and PAC libraries at Genome Systems.
  • DNA preps were prepared from the BACs and PACs that contained these markers. End sequences were obtained by dye terminator sequencing with vector primers on ABI 377 sequencers.
  • BAC or PAC end markers were designed from these sequences. These new markers were checked against the YACs to make sure that they mapped within the interval. If the map data were ambiguous, the markers were also checked against a radiation hybrid panel. These new markers were checked against the already identified BACs/PACs to determine the positions of these clones relative to each other. The outside markers from each clone contig were used to screen the Myriad BAC library; those that were negative on that BAC library were used to screen the BAC and PAC libraries at Genome Systems. Repeated cycles of library screening and marker development allowed us to build a BAC/PAC contig that spanned the minimal recombinant interval. EXAMPLE 4 Genomic sequencing Two different types of genomic sequencing sublibraries were prepared from BAC or PAC clones in the candidate region. Random-Sheared Sequencing Sub-libraries
  • BAC or PAC DNA was sheared by sonication.
  • the sonicated DNA was incubated with mung-bean nuclease (Pharmacia Biotech) followed by treatment with a Pfu polishing kit (Stratagene).
  • the DNA fragments were size fractionated on a 0.8% TAE agarose gel, and fragments in the size range of 1.0 - 1.6 kb were excised under longwave (365 nm) ultraviolet light.
  • the excised gel slice was rotated 180 degrees relative to the original direction of electrophoresis and then placed into a new gel tray containing 1.0% GTG-Seaplaque low-melting temperature agarose (FMC corporation) before the gel solidified.
  • Electrophoresis was repeated for the same time and voltage as the first run, resulting in a concentration of the DNA fragments in a small volume of agarose, and the gel slice containing the DNA fragments was once again excised from the gel.
  • the DNA fragments were purified from the agarose by incubating the gel slice with beta-agarose (New England Biolabs), followed by removal of the agarose monomers using disposable microconcentrators (Amicon) that employ a 50,000 Daltons molecular weight cutoff filter.
  • DNA fragments were ligated into the Hinc II site of the plasmid pMYG2, a pBluescript (Stratagene) derivative where the polylinker has been replaced by a different polylinker MYG2.
  • the vector was prepared by digestion with Hinc II followed by dephosphorylation with calf alkaline phosphatase (Boehringer Mannheim).
  • Ligated products were transformed into DH5 ⁇ E. coli competent cells (Life Technologies, Inc.) and plated on LB plates containing ampicillin, IPTG, and Bluo-gal (Sigma; Life Technologies, Inc.) .
  • White colonies were used to inoculate individual wells of 1 ml 96-well microtiter plates (Beckman) containing 200 microhters of LB media supplemented with ampicillin at 50 micrograms per milliliter. The plates were incubated for 16-20 hours in a shaking incubator at 37 degrees Celsius. After incubation, 20 microhters of dimethyl sulfoxide was added to each well and the plates stored frozen. The inserts of random-sheared clones were amplified from E.
  • BAC or PAC DNA was partially digested with the restriction enzyme Sau 3 A, and fragments in the size range of 5-8 kb were size fractionated and recovered from the agarose gel as described above for random-sheared fragments.
  • Sau 3 A fragments were ligated into the Bam HI site of pMYGl, a pBluescript (Stratagene) derivative where the polylinker has been replaced by a different polylinker MYG1.
  • the vector was prepared by digestion with Bam HI and dephosphorylation with shrimp alkaline phosphatase (Amersham). The ligated products were transformed and plated as described above for random-sheared clones.
  • bacterial colonies were screened using a plasmid preparation procedure that has been adapted for use in a 96-well format.
  • White colonies were picked into individual wells of 2 ml 96-well plates (Continental Laboratory Products) containing 1 ml LB media supplemented with 200 micrograms per milliliter ampicillin.
  • the plates were incubated 16-20 hours in a shaking incubator at 37 degrees Celsius.
  • a bacterial stock of these clones was prepared by transferring 100 microhters of the 1 ml cultures to another 96-well plate containing 200 microhters of LB media supplemented with ampicillin.
  • the remaining cells were pelleted by centrifugation and the pellets resuspended in 200 microhters of LB media.
  • One hundred microhters of the concentrated cells were transferred to a 96-well thermowell PCR plate (Costar), and the cells were once again pelleted.
  • the pelleted cells were resuspended in lysis buffer [250 mM Tris-HCl, pH 8.0, 50 mM EDTA, pH 8.0, 8% sucrose, 5% Triton X-100, 1 mM tartrazine, and 666 micrograms per milliliter lysozyme], and the plates were covered with thermowell lids (Costar) and incubated in a MJ Research fhermocycler for 2 minutes at 100 degrees Celsius followed by 2 minutes at 25 degrees Celsius.
  • lysis buffer 250 mM Tris-HCl, pH 8.0, 50 mM EDTA, pH 8.0, 8% sucrose, 5% Triton X-100, 1 mM tartrazine, and 666 micrograms per milliliter lysozyme
  • the bacterial stocks of clones with inserts in the 5-8 kb size range were used to inoculate 3 ml cultures of LB media supplemented with ampicillin, which were incubated overnight in a shaking incubator at 37 degrees Celsius. Plasmid DNA was prepared from these cultures using the Autogen robotic plasmid preparation machine (Integrated Separation Systems). The resulting DNA templates are subjected to DNA sequencing from both ends with Ml 3 forward or reverse fluorescent energy transfer (FET) dye-labeled primers on ABI 377 sequencers. DNA sequencing gel files were examined for lane tracking accuracy and adjusted where necessary before data extraction.
  • FET fluorescent energy transfer
  • ABI sample files resulting from gel files were converted to the Standard Chromatogram Format (SCF) [Dear and Staden] and trimmed of sequencing vector (pMYGl or pMYG2). Trimmed sequences were assembled using Acem.bly (Gold-Mieg et al, 1995; Durbin and Thierry-Mieg, 1991). Contiguous sequence resulting from automatic assembly was screened for residual vector sequence (both sequencing vector and cloning vector) as well as for bacterial contamination using BLAST (Altschul et al, 1990).
  • Remaining sequences were arranged according to the relative position and orientation of assembled Sau3AI partial digest clone sequence reads as well as sequence similarity to overlapping genomic clones. Repetitive sequence was masked from the sequence contigs using xblast (Claverie and States, 1993). These masked sequences were placed in a Genetic Data Environment (GDE) (Smith et al., 1994) local database for subsequent similarity searches. Similarities among genomic DNA sequences and hybrid-selected cDNA clones as well as GenBank entries—both DNA and protein—were identified using BLAST. DNA sequences were also characterized with respect to short period repeats, CpG content, and long open reading frames.
  • GDE Genetic Data Environment
  • Method 1 cDNA preparation and selection.
  • Poly (A) enriched RNA from human mammary gland, prostate, testis, fetal brain, and placenta tissues and from total RNA of the cell line Caco-2 (ATCC HTB 37) were reverse transcribed using the tailed random primer RXGN6 and M-MLV Reverse Transcriptase (Life Technologies, Inc.).
  • First strand cDNA was poly(A) tailed, 2nd strand synthesis was primed with the oligo RXGT12 , and then the ds cDNA was expanded by amplification with the primer RXG.
  • Hybrid selection was carried out for two consecutive rounds of hybridization to immobilized BAC, PAC or gel purified YAC DNA as described previously. [Parimoo et al, 1991; Rommens et al, 1994]. Individual gel purified YACs or groups of two to four overlapping BAC and/or PAC clones were used in individual selection experiments. Hybridizing cDNA was collected, passed over a G50 Fine Sephadex column and amplified using tailed primers. The products were then digested with EcoRI, size selected on agarose gels, and ligated into pBluescript (Stratagene) that had been digested with EcoRI and treated with calf alkaline phosphatase (Boehringer Mannheim). Ligation products were transformed into competent DH5 ⁇ E. coli cells (Life Technologies, Inc.).
  • Plasmids from 25 to 50 clones from each selection experiment that did not hybridize in prescreening were isolated for further analysis.
  • the retrieved cDNA fragments were verified to originate from individual starting genomic clones by hybridization to restriction digests of DNAs of the starting clones, of a hamster hybrid cell line that contains chromosome 1 as its only human material, and to human genomic DNA.
  • the clones were tentatively assigned into groups based on the overlapping or non-overlapping intervals of the genomic clones.
  • Method 2 cDNA Preparation.
  • Poly(A) enriched RNA from human mammary gland, fetal brain, lymphocyte, pancreas, prostate, stomach, and thymus were reverse-transcribed using the tailed random primer XN12 and Superscript II reverse transcriptase (Gibco BRL). After second strand synthesis and end polishing, the ds cDNA was purified on Sepharose CL-4B columns (Pharmacia). cDNAs were "anchored" by ligation of a double-stranded oligo RP (RP-2 annealed to RL-1) to their 5' ends (5' relative to mRNA) using T4 DNA ligase.
  • Anchored ds cDNA was then repurified on Sepharose CL-4B columns. Selection was performed by a modified procedure of Lovett et al. (1991). cDNAs from mammary gland, fetal brain, lymphocyte, pancreas, prostate, stomach, and thymus tissues were first expanded by amplification using a nested version of RP, RP.A and XPCR, and purified by fractionation on Sepharose CL-4B. Selection probes were prepared from purified Pis, BACs or PACs by digestion with Hinfl and Exonuclease III. The single-stranded probe was photolabelled with photobiotin (Gibco BRL) according to the manufacturer's recommendations.
  • photobiotin Gibco BRL
  • Probe, cDNA and C 0 t-1 DNA and poly A DNA were hybridized in 2.4M TEA-C1, lOmM NaPO4, ImM EDTA.
  • Hybridized cDNAs were captured on streptavidin-paramagnetic particles (Dynal), eluted, and reamplified with a further nested version of RP, RP.B and XPCR, and gel purified.
  • the selected, amplified cDNA was hybridized with an additional aliquot of probe, C 0 t- 1 DNA and poly A DNA.
  • Captured and eluted products were amplified again with RP.B and XPCR, size-selected by gel electrophoresis, and cloned into dephosphorylated Hindi cut pUC18. Ligation products were transformed into XL2-Blue ultra-competent cells (Stratagene).
  • Insert-containing clones were identified by blue/white selection on Xgal or Bluo-gal plates. Inserts were amplified by colony PCR with vector primers and then sequenced on ABI 377 sequencers. Alignment of these cDNA sequences to corresponding genomic sequences, and parsing of the revealed exons across those genomic sequences, allowed initial characterization of genes located within the region.
  • Inter-exon PCR Following sequence analysis of hybrid selected clones that originated from the HPCl region, several primers were designed to try to amplify HPCl region products from fetal brain, breast, pancreas, prostate, stomach, and thymus cDNAs. Amplification was by hot start PCR; conditions used were an initial denaturation step at 95°C for 30 sec followed by a pause at 80°C while the polymerase/ nucleotide mixture was added to the template/primer mixtures. The hot start was followed by 35 cycles of denaturation at 96°C (4 s), annealing at
  • cDNAs were "anchored" by ligation of a double-stranded oligo RP (RP-2 annealed to RL-1) to their 5' ends (5' relative to mRNA) using T4 DNA ligase. Anchored ds cDNA was then repurified on Sepharose CL-4B columns.
  • the 5' sequences of candidate genes within the HPCl region were amplified using differenct primer combinations, and PCR products were fractionated on an agarose gel, gel purified, and captured on streptavidin-paramagnetic particles (Dynal). Reamplifications were performed if necessary using nested primers. These PCR reactions gave several bands on an agarose gel; the PCR products were gel purified and sequenced in the reverse direction with dye terminator chemistry on an ABI 377 sequencer.
  • 3' RACE The 3' end exons of candidate genes within the HPCl region was identified by a modified RACE protocol called biotin capture 3' RACE.
  • Poly(A) enriched RNA from prostate was reverse-transcribed using a tailed random primer and Superscript II reverse transcriptase (Life Technologies).
  • the first strand (heteroduplex) cDNA was purified by fractionation on a Sepharose CL-6B column.
  • the 3' sequence of candidate genes within the HPCl region was amplified with a biotinylated forward primer and an the anchor primer. PCR products amplified with these primers were fractionated on an agarose gel, gel purified, and captured on streptavidin- paramagnetic particles (Dynal). Captured material was reamplified as necessary using a nested phosphorylated forward primer PF5 and a tailed random primer.
  • PCR products were gel purified, ligated into the vector pMYG2, and transformed into DH5 ⁇ cells. Colony PCR products were sequenced using using dye terminator chemistry on an ABI 377 sequencer.
  • EXAMPLE 7 cDNA library screening Radioactive probes prepared from hybrid selected clones representative of candidate gene transcripts within the HPCl region were used as probes to screen a total of 5.5 x 10 6 recombinant phage from a human prostate ⁇ gtl l cDNA library (HL1131b, Clontech). Prehybridization and hybridization was performed at 42°C in 50% formamide, 5X SSPE, 0.1% SDS, 5X Denhardt's mixture, 0.2 mg/ml denatured salmon sperm DNA and 2 mg/ml poly (A). Dextran sulfate (4% v/v) was included in the hybridization solution only.
  • the filters were rinsed in 2X SSC for 10 minutes at room temperature and then rinsed in 2X SSC/0.1% SDS for 30 minutes at 60°C followed by two washes in IX SSC/0.1% SDS for 20 minutes each at 60°C.
  • the positive phage were retested for second and third screenings, as required, to obtain purified plaques for sequencing. Inserts were amplified by phage PCR with vector primers and then sequenced using dye terminator chemistry on ABI 377 sequencers.
  • Genomic DNA Using genomic DNAs from prostate kindred members, prostate cancer affecteds, and tumor cell lines as templates, nested PCR amplifications were performed to generate PCR products of the candidate genes in the HPCl region that were screened for mutations.
  • One to 10 ng of genomic DNA were subjected to a 23-26 cycle primary amplification, after which the PCR products were diluted 60-fold and reamplified using nested
  • PCR samples were sequenced with Ml 3 forward or reverse fluorescent energy transfer (FET) dye- labeled primers on ABI 377 sequencers. Chromatograms were analyzed for the presence of polymorphisms or sequence aberrations in either the Macintosh program Sequencer (Gene Codes) or the Java program Mutscreen (Myriad, proprietary).
  • FET fluorescent energy transfer
  • cDNA Total RNA prepared from either tumor cell lines or prostate kindred lymphocytes was treated with DNase I (Boehringer Mannheim) to remove contaminating genomic DNA, and then reverse transcribed to heteroduplex cDNA with a mix of Nio random primers and a tailed oligo dT primer, and Superscript II reverse transcriptase (Life Technologies). This cDNA was used as the template for nested PCR amplifications to generate the cDNA PCR products of the candidate genes that were screened for HPCl mutations.
  • DNase I Boehringer Mannheim
  • cDNAs were amplified by hot start PCRs using TaqPlus DNA polymerase (Stratagene). Conditions used were an initial denaturation step at 95°C for 30 sec followed by a pause at 80°C while the polymerase/nucleotide mixture was added to the template/primer mixtures. The hot start was followed by cycles of denaturation at
  • PCR products were gel purified and then sequenced with Ml 3 forward or reverse fluorescent energy transfer (FET) dye- labeled primers on ABI 377 sequencers. The sequences of these products were analyzed in GDE to determine their exon structure. Chromatograms were analyzed for the presence of polymorphisms or sequence aberrations in either the Macintosh program Sequencher (Gene Codes) or the Java program Mutscreen (Myriad, proprietary).
  • FET fluorescent energy transfer
  • the cDNA sequence of the 07CG27 gene is set forth in SEQ ID NO:l, with the corresponding protein sequence set forth in SEQ ID NO:2.
  • the 07CG27 gene comprises 33 exons as shown in Table 1, with the base numbering relative to SEQ ID NO:l.
  • the structure and function of 07CG27 gene are determined according to the following methods. Biological Studies. Mammalian expression vectors containing 07CG27 cDNA are constructed and transfected into appropriate prostate carcinoma cells with lesions in the gene. Wild-type 07CG27 cDNA as well as altered 07CG27 cDNA are utilized. The altered 07CG27 cDNA can be obtained from altered 07CG27 alleles or produced as described below. Phenotypic reversion in cultures (e.g., cell morphology, doubling time, anchorage-independent growth) and in animals (e.g., tumorigenicity) is examined. The studies will employ both wild- type and mutant forms of the gene.
  • Biological Studies Mammalian expression vectors containing 07CG27 cDNA are constructed and transfected into appropriate prostate carcinoma cells with lesions in the gene. Wild-type 07CG27 cDNA as well as altered 07CG27 cDNA are utilized. The altered 07CG27 cDNA can be obtained from altered 07CG27 alleles or produced as described below. Phenotyp
  • This procedure can be repeated to generate antibodies against mutant forms of the 07CG27 protein.
  • These antibodies in conjunction with antibodies to wild type 07CG27, are used to detect the presence and the relative level of the mutant forms in various tissues and biological fluids.
  • EXAMPLE 11 Generation of Monoclonal Antibodies Specific for 07CG27 Monoclonal antibodies are generated according to the following protocol. Mice are immunized with immunogen comprising intact 07CG27 or 07CG27 peptides (wild type or mutant) conjugated to keyhole limpet hemocyanin using glutaraldehyde or EDC as is well known.
  • the immunogen is mixed with an adjuvant.
  • Each mouse receives four injections of 10 to
  • mice 100 mg of immunogen and after the fourth injection blood samples are taken from the mice to determine if the serum contains antibody to the immunogen. Serum titer is determined by ELISA or RIA. Mice with sera indicating the presence of antibody to the immunogen are selected for hybridoma production.
  • Spleens are removed from immune mice and a single cell suspension is prepared (see
  • Peptides that bind to the 07CG27 product are isolated from both chemical and phage- displayed random peptide libraries as follows.
  • Fragments of the 07CG27 gene product are expressed as GST and His-tag fusion proteins in both E. coli and SF9 cells.
  • the fusion protein is isolated using either a glutathione matrix (for GST fusions proteins) or nickel chelation matrix (for His-tag fusion proteins).
  • This target fusion protein preparation is either screened directly as described below, or eluted with glutathione or imidizole.
  • the target protein is immobilized to either a surface such as polystyrene; or a resin such as agarose; or solid supports using either direct absorption, covalent linkage reagents such as glutaraldehyde, or linkage agents such as biotin-avidin.
  • Two types of random peptide libraries of varying lengths are generated: synthetic peptide libraries that may contain derivatized residues, for example by phosphorylation or myristylation, and phage-displayed peptide libraries which may be phosphorylated. These libraries are incubated with immobilized 07CG27 gene product in a variety of physiological buffers. Next, unbound peptides are removed by repeated washes, and bound peptides recovered by a variety of elution reagents such as low or high pH, strong denaturants, glutathione, or imidizole. Recovered synthetic peptide mixtures are sent to commercial services for peptide micro- sequencing to identify enriched residues.
  • Recovered phage are amplified, rescreened, plaque purified, and then sequenced to determined the identity of the displayed peptides.
  • Use of 07CG27 binding peptides Peptides identified from the above screens are synthesized in larger quantities as biotin conjugates by commercial services. These peptides are used in both solid and solution phase competition assays with 07CG27 and its interacting partners identified in yeast 2-hybrid screens. Versions of these peptides that are fused to membrane-permeable motifs (Lin et al., 1995; Rojas et al., 1996) will be chemically synthesized, added to cultured cells and the effects on growth, apoptosis, differentiation, cofactor response, and internal changes will be assayed.
  • EXAMPLE 13 Sandwich Assay for 07CG27 Monoclonal antibody is attached to a solid surface such as a plate, tube, bead, or particle.
  • the antibody is attached to the well surface of a 96-well ELISA plate.
  • 100 ml sample e.g., serum, urine, tissue cytosol
  • the sample is incubated for 2 hrs at room temperature.
  • the sample fluid is decanted, and the solid phase is washed with buffer to remove unbound material.
  • 100 ml of a second monoclonal antibody to a different determinant on the 07CG27 peptide/protein is added to the solid phase.
  • This antibody is labeled with a detector molecule (e.g., 125-1, enzyme, fluorophore, or a chromophore) and the solid phase with the second antibody is incubated for two hrs at room temperature. The second antibody is decanted and the solid phase is washed with buffer to remove unbound material.
  • a detector molecule e.g., 125-1, enzyme, fluorophore, or a chromophore
  • the amount of bound label which is proportional to the amount of 07CG27 peptide/protein present in the sample, is quantitated. Separate assays are performed using monoclonal antibodies which are specific for the wild-type 07CG27 as well as monoclonal antibodies specific for each of the mutations identified in 07CG27.
  • Sequence encoding all or portions of 07CG27 are ligated to pAS2-l (Clontech) such that the coding sequence of 07CG27 is in- frame with coding sequence for the GAL4p DNA-binding domain.
  • This plasmid construct is introduced into the yeast reporter strain Y190 by transformation.
  • a library of activation domain fusion plasmids prepared from human prostate cDNA (Clontech) is then introduced into strain Y190 carrying the pAS2-l -based fusion construct. Transformants are spread onto 20 - 150 mm plates of yeast minimal media lacking leucine, tryptophan, and histidine, and containing 25 mM 3-amino-l,2,4-triazole.
  • yeast colonies are assayed for expression of the lacZ reporter gene by ⁇ -galactosidase filter assay. Colonies that both grow in the absence of histidine and are positive for production of ⁇ -galactosidase are chosen for further characterization.
  • the activation domain plasmid is purified from positive colonies by the smash-and-grab technique. These plasmids are introduced into E. coli DH5 ⁇ by electroporation and purified from E. coli by the alkaline lysis method. To test for the specificity of the interaction, specific activation domain plasmids are cotransformed into strain Y190 with plasmids encoding various DNA-binding domain fusion proteins, including fusions to 07CG27 and human lamin C. Transformants from these experiments are assayed for expression of the HIS3 and lacZ reporter genes. Positives that express reporter genes with 07CG27 constructs and not with lamin C constructs encode bona fide 07CG27 interacting proteins. These proteins are identified and characterized by sequence analysis of the insert of the appropriate activation domain plasmid.

Abstract

A human gene which is here named 07CG27 has been identified which is a marker for prostate cancer.

Description

TITLE OF THE INVENTION 07CG27 GENE
FIELD OF THE INVENTION
The present invention relates generally to the field of human genetics. Specifically, the present invention relates to methods and materials used to isolate and detect a human prostate cancer marker gene (identified as 07CG27 herein). The invention further relates to the screening of drugs for therapy. Finally, the invention relates to the screening of the 07CG27 gene for mutations, which are useful for diagnosing the predisposition to disease.
The publications and other materials used herein to illuminate the background of the invention, and in particular, cases to provide additional details respecting the practice, are incorporated herein by reference, and for convenience, are referenced by author and date in the following text and respectively grouped in the appended List of References.
BACKGROUND OF THE INVENTION
The genetics of cancer is complicated, involving gain or loss of function of three loosely defined classes of genes: (1) dominant, positive regulators of the transformed state (oncogenes); (2) recessive, negative regulators of the transformed state (tumor suppressor genes); (3) recessive genes involved in maintenance of genome integrity (caretaker genes) (Kinzler and Vogelstein, 1997). Over one hundred oncogenes have been characterized. About a dozen tumor suppressor genes and a similar number of caretaker genes have been identified; the number of genes falling into these last two classes is expected to increase beyond fifty (Knudson, 1993).
The involvement of so many genes underscores the complexity of the growth control mechanisms that operate in cells to maintain the integrity of normal tissue. This complexity is manifest in another way. So far, no single gene has been shown to participate in the development of all, or even the majority of, human cancers. The most common oncogenic mutations are in the H-ras gene, found in 10-15% of all solid tumors (Anderson et al., 1992). The most frequently mutated tumor predisposition genes are the TP53 gene, homozygously deleted or mutated in roughly 50% of all tumors, and CDKN2, which was homozygously deleted in 46% of tumor cell lines examined (Kamb et al, 1994). Without a target that is common to all transformed cells, the dream of a "magic bullet" that can destroy or revert cancer cells while leaving normal tissue unharmed is improbable. The hope for a new generation of specifically targeted antitumor drugs may rest on the ability to identify oncogenes, tumor suppressor, and caretaker genes that play general roles in the process of oncogenesis.
Specific germline alleles of certain oncogenes, tumor suppressor, and caretaker genes are causally associated with predisposition to cancer. This set of genes is referred to as tumor predisposition genes. Some of the tumor predisposition genes which have been cloned and characterized influence susceptibility to: 1) Retinoblastoma (RBI); 2) Wilms' tumor (WT1); 3) Li-Fraumeni (TP53); 4) Familial adenomatous polyposis (APC); 5) Neurofibromatosis type 1 (NFl); 6) Neurofibromatosis type 2 (NF2); 7) von Hippel-Lindau syndrome (VHL); 8) Multiple endocrine neoplasia type 2A (MEN2A); 9) Melanoma (CDKN2 and CDK4); 10) Breast and ovarian cancer (BRCA1 and BRCA2); 11) Cowden disease (MMACl); 12) Multiple endocrine neoplasia (MEN1); 13) Nevoid basal cell carcinoma syndrome (PTC); 14) Tuberous sclerosis 2 (TSC2); 15) Xeroderma pigmentosum (genes involved in nucleotide excision repair); 16) Hereditary nonpolyposis colorectal cancer (genes involved in mismatch repair).
Tumor predisposition loci that have been mapped genetically but not yet isolated include genes for: Lynch cancer family syndrome 2 (LCFS2); Neuroblastoma (NB); Beckwith- Wiedemann syndrome (BWS); Renal cell carcinoma (RCC); and Tuberous sclerosis 1 (TSC1). Tumor predisposition genes that have been characterized to date encode products with similarities to a variety of protein types, including DNA binding proteins (WT1), ancillary transcription regulators (RBI), GTPase activating proteins or GAPs (NFl), cytoskeletal components (NF2), membrane bound receptor kinases (MEN2A), cell cycle regulators (CDKN2 and CDK4), tyrosine phosphatases (MMACl), as well as others with no obvious similarity to proteins of known function (BRCA2).
In many cases, the tumor predisposition gene originally identified through genetic studies has been shown to be lost or mutated in some sporadic tumors. This result suggests that regions of chromosomal aberration, whether germline, in tumors, or in tumor cell lines, may signify the position of important tumor predisposition genes involved both in genetic predisposition to cancer and in sporadic cancer.
Prostate cancer is the most common cancer in men in many western countries, and the second leading cause of cancer deaths in men. It accounts for more than 40,000 deaths in the US annually. The number of deaths is likely to continue rising over the next 10 to 15 years. In the US, prostate cancer is estimated to cost $1.5 billion per year in direct medical expenses. In addition to the burden of suffering, it is a major public-health issue. Numerous studies have provided evidence for familial clustering of prostate cancer, indicating that family history is a major risk factor for this disease (Cannon et al., 1982; Steinberg et al., 1990; Carter et al, 1993). Prostate cancer has long been recognized to be, in part, a familial disease. Numerous investigators have examined the evidence for genetic inheritance and concluded that the data are most consistent with dominant inheritance for a major susceptibility locus or loci. Woolf (1960), described a relative risk of 3.0 of developing prostate cancer among first-degree relatives of prostate cancer cases in Utah using death certificate data. Relative risks ranging from 3 to 11 for first-degree relatives of prostate cancer cases have been reported (Cannon et al., 1982; Woolf, 1960; Fincham et al., 1990; Meikle et al., 1985; Krain, 1974; Morganti et al., 1956; Goldgar et al., 1994). Carter et al. (1992) performed segregation analysis on families ascertained through a single prostate cancer proband. The analysis suggested Mendelian inheritance in a subset of families through autosomal dominant inheritance of a rare (q=0.003), high-risk allele with estimated cumulative risk of prostate cancer for carriers of 88% by age 85. Inherited prostate cancer susceptibility accounted for a significant proportion of early-onset disease, and overall was responsible for 9% of prostate cancer occurrence by age 85. Recent results demonstrate that at least two loci exist which convey susceptibility to prostate cancer as well as other cancers. These loci are HPCl on chromosome lq, (Smith et al., 1996), HPCX on chromosome Xp (Xu et al., 1998), and one or more loci responsible for the unmapped residual.
Smith et al., (1996) indicated that the inherited prostate susceptibility in kindreds with early age onset is linked to chromosome 1 (the HPCl locus or region). Most strategies for cloning a chromosome 1 -linked prostate cancer predisposing gene require precise genetic localization studies. The simplest model for the functional role of a prostate cancer predisposing gene holds that alleles of prostate cancer predisposing gene that predispose to cancer are recessive to wild type alleles; that is, cells that contain at least one wild type allele are not cancerous. However, cells that contain one wild type allele and one predisposing allele may occasionally suffer loss of the wild type allele either by random mutation or by chromosome loss during cell division (nondisjunction). All the progeny of such a mutant cell lack the wild type function of the gene and may develop into tumors. According to this model, predisposing alleles of the gene are recessive, yet susceptibility to cancer is inherited in a dominant fashion: men who possess one predisposing allele (and one wild type allele) risk developing cancer, because their prostate cells may spontaneously lose the wild type allele. This model applies to both tumor suppressor and caretaker genes described above. By inference this model may also explain the HPCl function, as has recently been suggested (Smith et al, 1996). A second possibility is that prostate cancer predisposing alleles are truly dominant; that is, a wild type allele cannot overcome the tumor-forming role of the predisposing allele. Thus, a cell that carries both wild type and mutant alleles would not necessarily lose the wild type copy before giving rise to malignant cells. Instead, prostate cells in predisposed individuals would undergo some other stochastic change(s) leading to cancer. If a prostate cancer predisposing alleles are recessive, the prostate cancer predisposing gene is expected to be expressed in normal prostate tissue but not functionally expressed in prostate tumors. In contrast, if prostate cancer predisposing alleles are dominant, the wild type gene may or may not be expressed in normal prostate tissue. However, the predisposing allele will likely be expressed in prostate tumor cells. Evidence for a prostate cancer susceptibility locus (identified in the literature as HPCl) on the long arm of chromosome 1, which is hypothesized to explain approximately 35% of families, was recently presented (Smith et al., 1996). Although several groups report evidence supporting this localization, it has not yet been confirmed statistically. Both the original Smith et al. report and a subsequent analysis of additional families (Cooney et al., 1997), suggest that the bulk of linkage evidence comes from African-American high-risk kindreds. In addition, it appears that this gene predisposes (although not exclusively) primarily to early onset prostate cancer. The chromosome 1 linkage of HPCl has not been statistically confirmed; however, a report by Cooney et al. (1997) as well as a report by Neuhausen et al. (1977) are suggestive of confirmation, with less-than-significant indications of linkage at the location suggested to harbor HPCl.
Identification of a prostate cancer predisposition locus would permit the early detection of susceptible individuals and greatly increase our ability to understand the initial steps which lead to cancer. Cloning prostate cancer genes would also be important in the development of better diagnostic and prognostic products, as well as better cancer therapies. SUMMARY OF THE INVENTION
The present invention relates generally to the field of human genetics. Specifically, the present invention relates to methods and materials used to isolate and detect a human prostate cancer marker gene located within the HPCl region (identified herein as the 07CG27 gene). The invention further relates to the screening of drugs for therapy. Finally, the invention relates to the screening of the 07CG27 gene for mutations or for overexpression, which are useful for diagnosing the predisposition to disease. The 07CG27 gene is a homo log of two known genes HLA-B associated transcript 1 (BAT1) and BAT2. The 07CG27 gene is useful as a marker for the HPCl locus and as a marker for prostate cancer.
DETAILED DESCRIPTION OF THE INVENTION
The present invention relates generally to the field of human genetics. Specifically, the present invention relates to methods and materials used to isolate and detect a human prostate cancer marker gene (identified herein as the 07CG27 gene). The invention further relates to the screening of drugs for therapy. Finally, the invention relates to the screening of the 07CG27 gene for mutations or overexpression, which are useful for diagnosing the predisposition to disease. The 07CG27 gene is a homolog of two known genes HLA-B associated transcript 1 (BAT1) and BAT2. The 07CG27 gene is useful as a marker for the HPCl locus and as a marker for prostate cancer. The present invention provides an isolated polynucleotide comprising all, or a portion of the 07CG27 locus or of a mutated 07CG27 locus, preferably at least eight bases and not more than about 300 kb in length. Such polynucleotides may be antisense polynucleotides. The present invention also provides a recombinant construct comprising such an isolated polynucleotide, for example, a recombinant construct suitable for expression in a transformed host cell.
Also provided by the present invention are methods of detecting a polynucleotide comprising a portion of the 07CG27 locus or its expression product in an analyte. Such methods may further comprise the step of amplifying the portion of the 07CG27 locus, and may further include a step of providing a set of polynucleotides which are primers for amplification of said portion of the 07CG27 locus. The method is useful for either diagnosis of the predisposition to disease or the prognosis of disease. The present invention further provides an isolated polypeptide comprising all or part of the polypeptide encoded by the 07CG27 gene or a mutated form of the polypeptide. An analysis of the sequence of the 07CG27 polypeptide shows similarities with Guansoine Exchange Factor (GEF) proteins. Thus, the 07CG27 polypeptide is a GEF protein and functions as a ras activator. The present invention also provides isolated antibodies, preferably monoclonal antibodies, which specifically bind to an isolated polypeptide comprised of at least five amino acid residues encoded by the 07CG27 locus.
The present invention also provides kits for detecting in an analyte a polynucleotide comprising a portion of the 07CG27 locus, the kits comprising a polynucleotide complementary to the portion of the 07CG27 locus packaged in a suitable container, and instructions for its use.
The present invention further provides methods of preparing a polynucleotide comprising polymerizing nucleotides to yield a sequence comprised of at least eight consecutive nucleotides of the 07CG27 locus; and methods of preparing a polypeptide comprising polymerizing amino acids to yield a sequence comprising at least five amino acids encoded within the 07CG27 locus. The present invention further provides methods of screening the 07CG27 gene to identify mutations. Such methods may further comprise the step of amplifying a portion of the 07CG27 locus, and may further include a step of providing a set of polynucleotides which are primers for amplification of said portion of the 07CG27 locus. Such methods may also include a step of providing the complete set of short polynucleotides defined by the sequence of 07CG27 or discrete subsets of that sequence, all single-base substitutions of that sequence or discrete subsets of that sequence, all 1-, 2-, 3-, or 4-base deletions of that sequence or discrete subsets of that sequence, and all 1-, 2-, 3-, or 4-base insertions in that sequence or discrete subsets of that sequence. The method is useful for identifying mutations for use in either diagnosis of a predisposition to disease or the prognosis of disease. The present invention further provides methods of screening suspected 07CG27 mutant alleles to identify mutations in the 07CG27 gene.
In addition, the present invention provides methods to screen drugs for inhibition of 07CG27 gene product function as an anticancer therapy. Since the 07CG27 gene is an oncogene which acts as an activator of ras, any small molecule which interrupts this interaction will have anti-cancer activity. Thus, such drugs are useful for therapy.
It is a discovery of the present invention that the 07CG27 locus which is a marker for prostate cancer, is a gene encoding an 07CG27 protein, which has been found to have no significant homology with publicly available protein or DNA sequences. This gene is termed 07CG27 herein. The mutational events of the 07CG27 locus can involve deletions, insertions and point mutations within the coding sequence and the non-coding sequence. The preliminary evidence is that 07CG27 is an oncogene. Strategy For The Molecular Cloning Of 07CG27
Starting from the HPCl region on chromosome 1 of the human genome, a region which contains a genetic locus, 07CG27, which causes susceptibility to cancer, including prostate cancer, has been identified.
The region containing the 07CG27 locus was identified using a variety of genetic techniques. Genetic mapping techniques initially defined the 07CG27 region in terms of recombination with genetic markers. Based upon studies of large extended families ("kindreds") with multiple cases of prostate cancer, a chromosomal region has been pinpointed that contains the 07CG27 gene, as well as putative susceptibility alleles in the 07CG27 locus. Population Resources Large, well-documented Utah kindreds are especially important in providing good resources for human genetic studies. Each large kindred independently gives evidence whether or not a 07CG27 predisposing allele is segregating in that family. Recombinants informative for localization and isolation of the 07CG27 locus could be obtained only from kindreds large enough to confirm the presence of a susceptibility allele. Large sibships are especially important for studying prostate cancer, since penetrance of the 07CG27 predisposing allele is reduced both by age and sex, making informative sibships difficult to find. Furthermore, large sibships are essential for constructing haplotypes of deceased individuals by inference from the haplotypes of their close relatives.
Genetic Mapping Given a set of informative families, genetic markers are essential for linking a disease to a region of a chromosome. Such markers include restriction fragment length polymorphisms (RFLPs) (Botstein et al, 1980), markers with a variable number of tandem repeats (VNTRs) (Jeffreys et al, 1985; Nakamura et al, 1987), and an abundant class of DNA polymorphisms based on short tandem repeats (STRs), especially repeats of CpA (Weber and May, 1989; Litt et al, 1989). To generate a genetic map, one selects potential genetic markers and tests them using DNA extracted from members of the kindreds being studied. Genetic markers useful in searching for a genetic locus associated with a disease can be selected on an ad hoc basis, by densely covering a specific chromosome, or by detailed analysis of a specific region of a chromosome. A preferred method for selecting genetic markers linked with a disease involves evaluating the degree of informativeness of kindreds to determine the ideal distance between genetic markers of a given degree of polymorphism, then selecting markers from known genetic maps which are ideally spaced for maximal efficiency. Informativeness of kindreds is measured by the probability that the markers will be heterozygous in unrelated individuals. It is also most efficient to use STR markers which are detected by amplification of the target nucleic acid sequence using PCR; such markers are highly informative, easy to assay (Weber and May, 1989), and can be assayed simultaneously using multiplexing strategies (Skolnick and Wallace, 1988), greatly reducing the number of experiments required.
Once linkage has been established, one needs to find markers that flank the disease locus, i.e., one or more markers proximal to the disease locus, and one or more markers distal to the disease locus. Where possible, candidate markers can be selected from a known genetic map. Where none is known, new markers can be identified by the STR technique. Contig assembly Given a genetically defined interval flanked by meiotic recombinants, one needs to generate a contig of genomic clones that spans that interval. Publicly available resources, such as the Whitehead integrated maps of the human genome (e.g., the WICGR Chr 1 map of Nov. 19, 1996) provide aligned chromosome maps of genetic markers, other sequence tagged sites (STSs), radiation hybrid map data, and CEPH yeast artificial chromosome (YAC) clones. From the map data, one can often identify a set of yeast artificial chromosomes (YACs) that span the genetically defined interval. Oligonucleotide primer pairs for the markers located in the interval can be synthesized and used to screen libraries of bacterial artificial chromosomes (BACs) and PI artificial chromosomes (PACs). Successive rounds of BAC/PAC library screening with BAC or PAC end markers enables the completion of a BAC/PAC clone contig that spans the genetically defined interval.
Genomic sequencing Given a tiling path of BAC and PAC clones across a defined interval, one useful gene finding strategy is to generate an almost complete genomic sequence of that interval. Random genomic clone sublibraries can be prepared from each BAC or PAC clone in the tiling path. Individual sublibrary clones sufficient in number to generate an, on average, 6x redundant sequence of each BAC or PAC can then be end-sequenced with vector primers. These sequences can be assembled into sequence contigs, and these contigs placed in a local genomic sequence database. One can search the genomic sequence contigs for sequence similarity with known genes and expressed sequence tags (ESTs), examine them for the presence of long open translational reading frames, and characterize them for CpG dinucleotide frequency.
Hybrid selection
Given a tiling path of BAC and PAC clones across a defined interval, another useful gene finding strategy is to obtain cDNA clones cognate to the tiling path BACs and PACs. One preferred cDNA cloning strategy is hybrid selection. cDNA can be prepared from a number of human tissues and human cell lines in such a manner that the cDNA molecules have PCR primer binding sites (anchors) at each end. This cDNA can be affinity captured with the tiling path BACs and PAGsr-Captured cDNA can then be PCR amplified using the anchor primers and then cloned. Individual clones can then be end-sequenced with vector primers. The sequences of these cDNA clones can_be analyzed for similarity to genomic sequence contigs generated from BACs and PACs on the tiling path. One can then identify individual exons of genes in the genetically defined interval by parsing the sequences of true-positive hybrid selected clones across these genomic sequence contigs.
RACE and inter-exon PCR While hybrid selection is an efficient approach to the initial identification of novel genes located within a defined interval of the genome, the approach is not often the most efficient way to complete the cloning of those genes. Rapid amplification of cDNA ends (RACE) provides a PCR based method to identify new 5' and 3' cDNA sequences. cDNA can be prepared from a number of human tissues in a manner such that the cDNA molecules have PCR primer binding sites (anchors) at their 5' ends, 3' ends, or both. PCR amplification from this cDNA with 5' end anchor primers and gene specific reverse primers can generate 5' RACE products. Similarly, PCR amplification with 3' end anchor primers and gene specific forward primers can generate 3' RACE products. cDNA cloning techniques can also miss exons that lie between already known exons of a gene; for instance, this can easily occur if a particular exon is only included in a relatively rare splice variant of a transcript. Combinatorial inter-exon PCR is an effective strategy for detecting these exons. One can design a forward primer based on sequences from the first known exon of the gene and a set of reverse primers, one based on the sequence of each of the downstream exons (or any subset thereof) of the gene. Then one can PCR amplify from cDNA of tissues and cell lines thought to express the gene, using all the combinations of the forward primer with each reverse primer. Combinations as complex as a forward primer from each exon paired with a reverse primer from each exon, subject only to the limitation that the forward primer should be from an exon upstream of the exon from which the reverse primer was designed, can be tried. PCR products which differ in length from the expected product can be gel purified. In either RACE or combinatorial inter-exon PCRs, the PCR products can either be gel purified and then sequenced directly or first cloned and then sequenced. cDNA library screening Another useful strategy for finding new 5', 3', or internal sequences is cDNA library screening. One can make or purchase bacteriophage 1 cDNA libraries prepared from RNA from tissues or cell lines thought to express the gene. One then screens plaque lifts from those libraries with labeled nucleic acid probes based on the currently known sequences of the gene of interest. Individual positive clones are purified, and then the clone inserts can be sequenced. Mutation screening
Proof that any particular gene located within the genetically defined interval is 07CG27 is obtained by finding sequences in DNA or RNA extracted from affected kindred members which create abnormal 07CG27 gene products or abnormal levels of 07CG27 gene product. Such 07CG27 predisposing alleles will co-segregate with the disease in large kindreds. They will also be present at a much higher frequency in non-kindred individuals with prostate cancer than in individuals in the general population. Finally, since tumors often mutate somatically at loci which are in other instances mutated in the germline, we expect to see normal germline 07CG27 alleles mutated into sequences which are identical or similar to 07CG27 predisposing alleles in DNA extracted from tumor tissue. Whether one is comparing 07CG27 sequences from tumor tissue to 07CG27 alleles from the germline of the same individuals, or one is comparing germline 07CG27 alleles from cancer cases to those from unaffected individuals, the key is to find mutations which are serious enough to cause obvious disruption to the normal function of the gene product. These mutations can take a number of forms. The most severe forms would be frame shift mutations or large deletions which would cause the gene to code for an abnormal protein or one which would significantly alter protein expression. Less severe disruptive mutations would include small in-frame deletions and nonconservative base pair substitutions which would have a significant effect on the protein produced, such as changes to or from a cysteine residue, from a basic to an acidic amino acid or vice versa, from a hydrophobic to hydrophilic amino acid or vice versa, or other mutations which would affect secondary, tertiary or quaternary protein structure. Small deletions or base pair substitutions could also significantly alter protein expression by changing the level of transcription, splice pattern, mRNA stability, or translation efficiency of the 07CG27 transcript. Silent mutations or those resulting in conservative amino acid substitutions would not generally be expected to disrupt protein function.
Useful Diagnostic Techniques According to the diagnostic and prognostic method of the present invention, alteration of the wild-type 07CG27 locus is detected. In addition, the method can be performed by detecting the wild-type 07CG27 locus and confirming the lack of a predisposition to cancer at the 07CG27 locus. "Alteration of a wild-type gene" encompasses all forms of mutations including deletions, insertions and point mutations in the coding and noncoding regions. Deletions may be of the entire gene or of only a portion of the gene. Point mutations may result in stop codons, frameshift mutations or amino acid substitutions. Somatic mutations are those which occur only in certain tissues, e.g., in the tumor tissue, and are not inherited in the germline. Germline mutations can be found in any of a body's tissues and are inherited. The finding of 07CG27 mutations thus provides diagnostic information. A 07CG27 allele which is not deleted (e.g., found on the sister chromosome to a chromosome carrying an 07CG27 deletion) can be screened for other mutations, such as insertions, small deletions, and point mutations. Point mutational events may occur in regulatory regions, such as in the promoter of the gene, or in intron regions or at intron/exon junctions.
Useful diagnostic techniques include, but are not limited to fluorescent in situ hybridization (FISH), direct DNA sequencing, PFGE analysis, Southern blot analysis, single stranded conformation analysis (SSCA), RNase protection assay, allele-specific oligonucleotide (ASO), dot blot analysis and PCR-SSCP, as discussed in detail further below. Also useful is the recently developed technique of DNA microchip technology.
Predisposition to cancers, such as prostate cancer, and the other cancers identified herein, can be ascertained by testing any tissue of a human for mutations of the 07CG27 gene. For example, a person who has inherited a germline 07CG27 mutation would be prone to develop cancers. This can be determined by testing DNA from any tissue of the person's body. Most simply, blood can be drawn and DNA extracted from the cells of the blood. In addition, prenatal diagnosis can be accomplished by testing fetal cells, placental cells or amniotic cells for mutations of the 07CG27 gene. Alteration of a wild-type 07CG27 allele, whether, for example, by point mutation or deletion, can be detected by any of the means discussed herein.
There are several methods that can be used to detect DNA sequence variation. Direct DNA sequencing, either manual sequencing or automated fluorescent sequencing can detect sequence variation. For a gene as large as 07CG27, manual sequencing is very labor-intensive, but under optimal conditions, mutations in the coding sequence of a gene are rarely missed. Another approach is the single-stranded conformation polymorphism assay (SSCA) (Orita et al, 1989). This method does not detect all sequence changes, especially if the DNA fragment size is greater than 200 bp, but can be optimized to detect most DNA sequence variation. The reduced detection sensitivity is a disadvantage, but the increased throughput possible with SSCA makes it an attractive, viable alternative to direct sequencing for mutation detection on a research basis. The fragments which have shifted mobility on SSCA gels are then sequenced to determine the exact nature of the DNA sequence variation. Other approaches based on the detection of mis- matches between the two complementary DNA strands include clamped denaturing gel electrophoresis (CDGE) (Sheffield et al, 1991), heteroduplex analysis (HA) (White et al, 1992) and chemical mismatch cleavage (CMC) (Grompe et al, 1989). None of the methods described above will detect large deletions, duplications or insertions, nor will they detect a regulatory mutation which affects transcription or translation of the protein. Other methods which might detect these classes of mutations such as a protein truncation assay or the asymmetric assay, detect only specific types of mutations and would not detect missense mutations. A review of currently available methods of detecting DNA sequence variation can be found in a recent review by Grompe (1993). Once a mutation is known, an allele specific detection approach such as allele specific oligonucleotide (ASO) hybridization can be utilized to rapidly screen large numbers of other samples for that same mutation.
In order to detect the alteration of the wild-type 07CG27 gene in a tissue, it is helpful to isolate the tissue free from surrounding normal tissues. Means for enriching tissue preparation for disease cells are known in the art. For example, the tissue may be isolated from paraffin or cryostat sections. Disease cells may also be separated from normal cells by flow cytometry. These techniques, as well as other techniques for separating disease cells from normal cells, are well known in the art. If the disease tissue is highly contaminated with normal cells, detection of mutations is more difficult. Detection of point mutations may be accomplished by molecular cloning of the 07CG27 allele(s) and sequencing the allele(s) using techniques well known in the art. Alternatively, the gene sequences can be amplified directly from a genomic DNA preparation from the tumor tissue, using known techniques. The DNA sequence of the amplified sequences can then be determined.
There are six well known methods for a more complete, yet still indirect, test for confirming the presence of a susceptibility allele: 1) single-stranded conformation analysis (SSCA) (Orita et al, 1989); 2) denaturing gradient gel electrophoresis (DGGE) (Wartell et al, 1990; Sheffield et al, 1989); 3) RNase protection assays (Finkelstein et al, 1990; Kinszler et al, 1991); 4) allele-specific oligonucleotides (ASOs) (Conner et al, 1983); 5) the use of proteins which recognize nucleotide mismatches, such as the E. coli mutS protein (Modrich, 1991); and 6) allele-specific PCR (Rano and Kidd, 1989). For allele-specific PCR, primers are used which hybridize at their 3' ends to a particular 07CG27 mutation. If the particular 07CG27 mutation is not present, an amplification product is not observed. Amplification Refractory Mutation System (ARMS) can also be used, as disclosed in European Patent Application Publication No. 0332435 and in Newton et al, 1989. Insertions and deletions of genes can also be detected by cloning, sequencing and amplification. In addition, restriction fragment length polymorphism (RFLP) probes for the gene or surrounding marker genes can be used to score alteration of an allele or an insertion in a polymorphic fragment. Such a method is particularly useful for screening relatives of an affected individual for the presence of the 07CG27 mutation found in that individual. Other techniques for detecting insertions and deletions as known in the art can be used.
In the first three methods (SSCA, DGGE and RNase protection assay), a new electrophoretic band appears. SSCA detects a band which migrates differentially because the sequence change causes a difference in single-strand, intramolecular base pairing. RNase protection involves cleavage of the mutant polynucleotide into two or more smaller fragments. DGGE detects differences in migration rates of mutant sequences compared to wild-type sequences, using a denaturing gradient gel. In an allele-specific oligonucleotide assay, an oligonucleotide is designed which detects a specific sequence, and the assay is performed by detecting the presence or absence of a hybridization signal. In the mutS assay, the protein binds only to sequences that contain a nucleotide mismatch in a heteroduplex between mutant and wild-type sequences. Mismatches, according to the present invention, are hybridized nucleic acid duplexes in which the two strands are not 100%) complementary. Lack of total homo logy may be due to deletions, insertions, inversions or substitutions. Mismatch detection can be used to detect point mutations in the gene or in its mRNA product. While these techniques are less sensitive than sequencing, they are simpler to perform on a large number of tumor samples. An example of a mismatch cleavage technique is the RNase protection method. In the practice of the present invention, the method involves the use of a labeled riboprobe which is complementary to the human wild-type 07CG27 gene coding sequence. The riboprobe and either mRNA or DNA isolated from the tumor tissue are annealed (hybridized) together and subsequently digested with the enzyme RNase A which is able to detect some mismatches in a duplex RNA structure. If a mismatch is detected by RNase A, it cleaves at the site of the mismatch. Thus, when the annealed RNA preparation is separated on an elecfrophoretic gel matrix, if a mismatch has been detected and cleaved by RNase A, an RNA product will be seen which is smaller than the full length duplex RNA for the riboprobe and the mRNA or DNA. The riboprobe need not be the full length of the 07CG27 mRNA or gene but can be a segment of either. If the riboprobe comprises only a segment of the 07CG27 mRNA or gene, it will be desirable to use a number of these probes to screen the whole mRNA sequence for mismatches.
In similar fashion, DNA probes can be used to detect mismatches, through enzymatic or chemical cleavage. See, e.g., Cotton et al, 1988; Shenk et al, 1975; Novack et al, 1986. Alternatively, mismatches can be detected by shifts in the elecfrophoretic mobility of mismatched duplexes relative to matched duplexes. See, e.g., Cariello, 1988. With either riboprobes or DNA probes, the cellular mRNA or DNA which might contain a mutation can be amplified using PCR (see below) before hybridization. Changes in DNA of the 07CG27 gene can also be detected using Southern hybridization, especially if the changes are gross rearrangements, such as deletions and insertions.
DNA sequences of the 07CG27 gene which have been amplified by use of PCR may also be screened using allele-specific probes. These probes are nucleic acid oligomers, each of which contains a region of the 07CG27 gene sequence harboring a known mutation. For example, one oligomer may be about 30 nucleotides in length (although shorter and longer oligomers are also usable as well recognized by those of skill in the art), corresponding to a portion of the 07CG27 gene sequence. By use of a battery of such allele-specific probes, PCR amplification products can be screened to identify the presence of a previously identified mutation in the 07CG27 gene. Hybridization of allele-specific probes with amplified 07CG27 sequences can be performed, for example, on a nylon filter. Hybridization to a particular probe under high stringency hybridization conditions indicates the presence of the same mutation in the tumor tissue as in the allele-specific probe. The newly developed technique of nucleic acid analysis via microchip technology is also applicable to the present invention. In this technique, literally thousands of distinct oligonucleotide probes are built up in an array on a silicon chip. Nucleic acid to be analyzed is fluorescently labeled and hybridized to the probes on the chip. It is also possible to study nucleic acid-protein interactions using these nucleic acid microchips. Using this technique one can determine the presence of mutations or even sequence the nucleic acid being analyzed or one can measure expression levels of a gene of interest. The method is one of parallel processing of many, even thousands, of probes at once and can tremendously increase the rate of analysis. Several papers have been published which use this technique. Some of these are Hacia et al., 1996; Shoemaker et al., 1996; Chee et al., 1996; Lockhart et al., 1996; DeRisi et al., 1996; Lipshutz et al., 1995. This method has already been used to screen people for mutations in the breast cancer gene BRCA1 (Hacia et al., 1996). This new technology has been reviewed in a news article in Chemical and Engineering News (Borman, 1996) and been the subject of an editorial (Nature Genetics, 1996). Also see Fodor (1997).
The most definitive test for mutations in a candidate locus is to directly compare genomic 07CG27 sequences from diseased patients with those from a control population. Alternatively, one could sequence messenger RNA after amplification, e.g., by PCR, thereby eliminating the necessity of determining the exon structure of the candidate gene.
Mutations from cancer patients falling outside the coding region of 07CG27 can be detected by examining the non-coding regions, such as introns and regulatory sequences near or within the 07CG27 gene. An early indication that mutations in noncoding regions are important may come from Northern blot experiments that reveal messenger RNA molecules of abnormal size or abundance in cancer patients as compared to control individuals.
Alteration of 07CG27 mRNA expression can be detected by any techniques known in the art. These include Northern blot analysis, PCR amplification and RNase protection. Diminished mRNA expression indicates an alteration of the wild-type 07CG27 gene. Alteration of wild-type 07CG27 genes can also be detected by screening for alteration of wild-type 07CG27 protein. For example, monoclonal antibodies immunoreactive with 07CG27 can be used to screen a tissue. Lack of cognate antigen would indicate a 07CG27 mutation. Antibodies specific for products of mutant alleles could also be used to detect mutant 07CG27 gene product. Such immunological assays can be done in any convenient formats known in the art. These include Western blots, immunohistochemical assays and ELISA assays. Any means for detecting an altered 07CG27 protein can be used to detect alteration of wild-type 07CG27 genes. Functional assays, such as protein binding determinations, can be used. In addition, assays can be used which detect 07CG27 biochemical function. Finding a mutant 07CG27 gene product indicates alteration of a wild- type 07CG27 gene.
Mutant 07CG27 genes or gene products can also be detected in other human body samples, such as serum, stool, urine and sputum. The same techniques discussed above for detection of mutant 07CG27 genes or gene products in tissues can be applied to other body samples. In addition, the 07CG27 gene product itself may be secreted into the extracellular space and found in these body samples even in the absence of disease cells. By screening such body samples, a simple early diagnosis can be achieved for disease. In addition, the progress of therapy can be monitored more easily by testing such body samples for mutant 07CG27 genes or gene products.
The methods of diagnosis of the present invention are applicable to any disease in which 07CG27 has a role in its pathogenesis. The diagnostic method of the present invention is useful for clinicians, so they can decide upon an appropriate course of treatment. The primer pairs of the present invention are useful for determination of the nucleotide sequence of a particular 07CG27 allele using PCR. The pairs of single-stranded DNA primers can be annealed to sequences within or surrounding the 07CG27 gene on chromosome 1 in order to prime amplifying DNA synthesis of the 07CG27 gene itself. A complete set of these primers allows synthesis of all of the nucleotides of the 07CG27 gene coding sequences, i.e., the exons. The set of primers preferably allows synthesis of both intron and exon sequences. Allele-specific primers can also be used. Such primers anneal only to particular 07CG27 mutant alleles, and thus will only amplify a product in the presence of the mutant allele as a template.
In order to facilitate subsequent cloning of amplified sequences, primers may have restriction enzyme site sequences appended to their 5' ends. Thus, all nucleotides of the primers are derived from 07CG27 sequences or sequences adjacent to 07CG27, except for the few nucleotides necessary to form a restriction enzyme site. Such enzymes and sites are well known in the art. The primers themselves can be synthesized using techniques which are well known in the art. Generally, the primers can be made using oligonucleotide synthesizing machines which are commercially available. Given the sequence of the 07CG27 open reading frame shown in SEQ ID NO:l, design of particular primers is well within the skill of the art.
The nucleic acid probes provided by the present invention are useful for a number of purposes. They can be used in Southern hybridization to genomic DNA and in the RNase protection method for detecting point mutations already discussed above. The probes can be used to detect PCR amplification products. They may also be used to detect mismatches with the
07CG27 gene or mRNA using other techniques.
It has been discovered that individuals with the wild-type 07CG27 gene do not have disease which results from the 07CG27 allele. However, mutations which interfere with the function of the 07CG27 protein are involved in the pathogenesis of disease. Thus, the presence of an altered (or a mutant) 07CG27 gene which produces a protein having a loss of function, or altered function, directly correlates to an increased risk of disease. In order to detect an 07CG27 gene mutation, a biological sample is prepared and analyzed for a difference between the sequence of the 07CG27 allele being analyzed and the sequence of the wild-type 07CG27 allele. Mutant 07CG27 alleles can be initially identified by any of the techniques described above. The mutant alleles are then sequenced to identify the specific mutation of the particular mutant allele. Alternatively, mutant 07CG27 alleles can be initially identified by identifying mutant (altered) 07CG27 proteins, using conventional techniques. The mutant alleles are then sequenced to identify the specific mutation for each allele. The mutations, especially those which lead to an altered function of the 07CG27 protein, are then used for the diagnostic methods of the present invention. It has also been discovered that the 07CG27 gene is a marker for prostate cancer.
Definitions The present invention employs the following definitions: "Amplification of Polynucleotides" utilizes methods such as the polymerase chain reaction (PCR), ligation amplification (or ligase chain reaction, LCR) and amplification methods based on the use of Q-beta replicase. Also useful are strand displacement amplification (SDA) and nucleic acid sequence based amplification (NASBA). These methods are well known and widely practiced in the art. See, e.g., U.S. Patents 4,683,195 and 4,683,202 and Innis et al, 1990 (for PCR); and Wu et al, 1989a (for LCR); U.S. Patents 5,270,184 and 5,455,166 (for SDA); Spargo et al., 1996 (for thermophilic SDA) and U.S. Patent 5,409,818, Fahy et al., 1991 and Compton, 1991 for 3SR and NASBA. Reagents and hardware for conducting PCR are commercially available. Primers useful to amplify sequences from the 07CG27 region are preferably complementary to, and hybridize specifically to sequences in the 07CG27 region or in regions that flank a target region therein. 07CG27 sequences generated by amplification may be sequenced directly. Alternatively, but less desirably, the amplified sequence(s) may be cloned prior to sequence analysis. A method for the direct cloning and sequence analysis of enzymatically amplified genomic segments has been described by Scharf, 1986.
"Analyte polynucleotide" and "analyte strand" refer to a single- or double-stranded polynucleotide which is suspected of containing a target sequence, and which may be present in a variety of types of samples, including biological samples. "Antibodies." The present invention also provides polyclonal and/or monoclonal antibodies and fragments thereof, and immunologic binding equivalents thereof, which are capable of specifically binding to the 07CG27 polypeptides and fragments thereof or to polynucleotide sequences from the 07CG27 region, particularly from the 07CG27 locus or a portion thereof. The term "antibody" is used both to refer to a homogeneous molecular entity, or a mixture such as a serum product made up of a plurality of different molecular entities. Polypeptides may be prepared synthetically in a peptide synthesizer and coupled to a carrier molecule (e.g., keyhole limpet hemocyanin) and injected over several months into rabbits. Rabbit sera is tested for immunoreactivity to the 07CG27 polypeptide or fragment. Monoclonal antibodies may be made by injecting mice with the protein polypeptides, fusion proteins or fragments thereof. Monoclonal antibodies will be screened by ELISA and tested for specific immunoreactivity with 07CG27 polypeptide or fragments thereof. See, Harlow and Lane, 1988. These antibodies will be useful in assays as well as pharmaceuticals.
Once a sufficient quantity of desired polypeptide has been obtained, it may be used for various purposes. A typical use is the production of antibodies specific for binding. These antibodies may be either polyclonal or monoclonal, and may be produced by in vitro or in vivo techniques well known in the art. For production of polyclonal antibodies, an appropriate target immune system, typically mouse or rabbit, is selected. Substantially purified antigen is presented to the immune system in a fashion determined by methods appropriate for the animal and by other parameters well known to immunologists. Typical sites for injection are in footpads, intramuscularly, intraperitoneally, or intradermally. Of course, other species may be substituted for mouse or rabbit. Polyclonal antibodies are then purified using techniques known in the art, adjusted for the desired specificity. An immunological response is usually assayed with an immunoassay. Normally, such immunoassays involve some purification of a source of antigen, for example, that produced by the same cells and in the same fashion as the antigen. A variety of immunoassay methods are well known in the art. See, e.g., Harlow and Lane, 1988, or Goding, 1986. Monoclonal antibodies with affinities of 10" M"1 or preferably 10"9 to 10"10 M"1 or stronger will typically be made by standard procedures as described, e.g., in Harlow and Lane, 1988 or Goding, 1986. Briefly, appropriate animals will be selected and the desired immunization protocol followed. After the appropriate period of time, the spleens of such animals are excised and individual spleen cells fused, typically, to immortalized myeloma cells under appropriate selection conditions. Thereafter, the cells are clonally separated and the supernatants of each clone tested for their production of an appropriate antibody specific for the desired region of the antigen.
Other suitable techniques involve in vitro exposure of lymphocytes to the antigenic polypeptides, or alternatively, to selection of libraries of antibodies in phage or similar vectors. See Huse et al, 1989. The polypeptides and antibodies of the present invention may be used with or without modification. Frequently, polypeptides and antibodies will be labeled by joining, either covalently or non-covalently, a substance which provides for a detectable signal. A wide variety of labels and conjugation techniques are known and are reported extensively in both the scientific and patent literature. Suitable labels include radionuclides, enzymes, substrates, cofactors, inhibitors, fluorescent agents, chemiluminescent agents, magnetic particles and the like. Patents teaching the use of such labels include U.S. Patents 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149 and 4,366,241. Also, recombinant immunoglobulins may be produced (see U.S. Patent 4,816,567).
"Binding partner" refers to a molecule capable of binding a ligand molecule with high specificity, as for example, an antigen and an antigen-specific antibody or an enzyme and its inhibitor. In general, the specific binding partners must bind with sufficient affinity to immobilize the analyte copy/complementary strand duplex (in the case of polynucleotide hybridization) under the isolation conditions. Specific binding partners are known in the art and include, for example, biotin and avidin or streptavidin, IgG and protein A, the numerous, known receptor- ligand couples, and complementary polynucleotide strands. In the case of complementary polynucleotide binding partners, the partners are normally at least about 15 bases in length, and may be at least 40 bases in length. It is well recognized by those of skill in the art that lengths shorter than 15, between 15 and 40, and greater than 40 bases may also be used. The polynucleotides may be composed of DNA, RNA, or synthetic nucleotide analogs.
A "biological sample" refers to a sample of tissue or fluid suspected of containing an analyte polynucleotide or polypeptide from an individual including, but not limited to, e.g., plasma, serum, spinal fluid, lymph fluid, the external sections of the skin, respiratory, intestinal, and genitourinary tracts, tears, saliva, blood cells, tumors, organs, tissue and samples of in vitro cell culture constituents.
As used herein, the terms "diagnosing" or "prognosing," as used in the context of neoplasia, are used to indicate 1) the classification of lesions as neoplasia, 2) the determination of the severity of the neoplasia, or 3) the monitoring of the disease progression, prior to, during and after treatment.
"Encode". A polynucleotide is said to "encode" a polypeptide if, in its native state or when manipulated by methods well known to those skilled in the art, it can be transcribed and/or translated to produce the mRNA for and/or the polypeptide or a fragment thereof. The anti- sense strand is the complement of such a nucleic acid, and the encoding sequence can be deduced therefrom.
"Isolated" or "substantially pure". An "isolated" or "substantially pure" nucleic acid (e.g., an RNA, DNA or a mixed polymer) is one which is substantially separated from other cellular components which naturally accompany a native human sequence or protein, e.g., ribosomes, polymerases, many other human genome sequences and proteins. The term embraces a nucleic acid sequence or protein which has been removed from its naturally occurring environment, and includes recombinant or cloned DNA isolates and chemically synthesized analogs or analogs biologically synthesized by heterologous systems.
"07CG27 Allele" refers to normal alleles of the 07CG27 locus as well as alleles carrying variations that predispose individuals to develop disease. Such predisposing alleles are also called "07CG27 predisposing alleles".
"07CG27 Locus", "07CG27 Gene", "07CG27 Nucleic Acids" or "07CG27
Polynucleotide" each refer to polynucleotides, all of which are in the 07CG27 region, that are likely to be expressed in normal tissue, certain alleles of which predispose an individual to develop disease. Mutations at the 07CG27 locus may be involved in the initiation and/or progression of disease. The 07CG27 locus is intended to include coding sequences, intervening sequences and regulatory elements controlling transcription and/or translation. The 07CG27 locus is intended to include all allelic variations of the DNA sequence.
These terms, when applied to a nucleic acid, refer to a nucleic acid which encodes an
07CG27 polypeptide, fragment, homolog or variant, including, e.g., protein fusions or deletions. The nucleic acids of the present invention will possess a sequence which is either derived from, or substantially similar to a natural 07CG27-encoding gene or one having substantial homology with a natural 07CG27-encoding gene or a portion thereof.
The polynucleotide compositions of this invention include RNA, cDNA, genomic DNA, synthetic forms, and mixed polymers, both sense and antisense strands, and may be chemically or biochemically modified or may contain non-natural or derivatized nucleotide bases, as will be readily appreciated by those skilled in the art. Such modifications include, for example, labels, methylation, substitution of one or more of the naturally occurring nucleotides with an analog, internucleotide modifications such as uncharged linkages (e.g., methyl phosphonates, phosphotriesters, phosphoramidates, carbamates, etc.), charged linkages (e.g., phosphorothioates, phosphorodithioates, etc.), pendent moieties (e.g., polypeptides), intercalators (e.g., acridine, psoralen, etc.), chelators, alkylators, and modified linkages (e.g., alpha anomeric nucleic acids, etc.). Also included are synthetic molecules that mimic polynucleotides in their ability to bind to a designated sequence via hydrogen bonding and other chemical interactions. Such molecules are known in the art and include, for example, those in which peptide linkages substitute for phosphate linkages in the backbone of the molecule.
The present invention provides recombinant nucleic acids comprising all or part of the 07CG27 region. The recombinant construct may be capable of replicating autonomously in a host cell. Alternatively, the recombinant construct may become integrated into the chromosomal DNA of the host cell. Such a recombinant polynucleotide comprises a polynucleotide of genomic, cDNA, semi-synthetic, or synthetic origin which, by virtue of its origin or manipulation, 1) is not associated with all or a portion of a polynucleotide with which it is associated in nature; 2) is linked to a polynucleotide other than that to which it is linked in nature; or 3) does not occur in nature.
Therefore, recombinant nucleic acids comprising sequences otherwise not naturally occurring are provided by this invention. Although the wild-type sequence may be employed, it will often be altered, e.g., by deletion, substitution or insertion. cDNA or genomic libraries of various types may be screened as natural sources of the nucleic acids of the present invention, or such nucleic acids may be provided by amplification of sequences resident in genomic DNA or other natural sources, e.g., by PCR. The choice of cDNA libraries normally corresponds to a tissue source which is abundant in mRNA for the desired proteins. Phage libraries are normally preferred, but other types of libraries may be used. Clones of a library are spread onto plates, transferred to a substrate for screening, denatured and probed for the presence of desired sequences.
The DNA sequences used in this invention will usually comprise at least about five codons (15 nucleotides), more usually at least about 7-15 codons, and most preferably, at least about 35 codons. One or more introns may also be present. This number of nucleotides is usually about the minimal length required for a successful probe that would hybridize specifically with an 07CG27-encoding sequence.
Techniques for nucleic acid manipulation are described generally, for example, in Sambrook et al, 1989 or Ausubel et al, 1992. Reagents useful in applying such techniques, such as restriction enzymes and the like, are widely known in the art and commercially available from such vendors as New England BioLabs, Boehringer Mannheim, Amersham, Promega Biotec, U. S. Biochemicals, New England Nuclear, and a number of other sources. The recombinant nucleic acid sequences used to produce fusion proteins of the present invention may be derived from natural or synthetic sequences. Many natural gene sequences are obtainable from various cDNA or from genomic libraries using appropriate probes. See, GenBank, National Institutes of Health.
"07CG27 Region" refers to a portion of human chromosome 1 containing the 07CG27 gene.
As used herein, the terms "07CG27 locus", "07CG27 allele" and "07CG27 region" all refer to the double-stranded DNA comprising the locus, allele, or region, as well as either of the single-stranded DNAs comprising the locus, allele or region.
As used herein, a "portion" of the 07CG27 locus or region or allele is defined as having a minimal size of at least about eight nucleotides, or preferably about 15 nucleotides, or more preferably at least about 25 nucleotides, and may have a minimal size of at least about 40 nucleotides. This definition includes all sizes in the range of 8-40 nucleotides as well as greater than 40 nucleotides. Thus, this definition includes nucleic acids of 8, 12, 15, 20, 25, 40, 60, 80, 100, 200, 300, 400, 500 nucleotides, or nucleic acids having any number of nucleotides within these ranges of values (e.g., 9, 10, 11, 16, 23, 30, 38, 50, 72, 121, etc., nucleotides), or nucleic acids having more than 500 nucleotides. The present invention includes all novel nucleic acids having at least 8 nucleotides derived from any of SEQ ID NOs:l and 3-21 and any combination of these sequences as described in further detail below, its complement or functionally equivalent nucleic acid sequences. The present invention .does not include nucleic acids which exist in the prior art. That is, the present invention includes all nucleic acids having at least 8 nucleotides derived from any of SEQ ID NOs:l and 3-21 and any combination of these sequences as described in further detail below with the proviso that it does not include nucleic acids existing in the prior art. "07CG27 protein" or "07CG27 polypeptide" refers to a protein or polypeptide encoded by the 07CG27 locus, variants or fragments thereof. The term "polypeptide" refers to a polymer of amino acids and its equivalent and does not refer to a specific length of the product; thus, peptides, oligopeptides and proteins are included within the definition of a polypeptide. This term also does not refer to, or exclude modifications of the polypeptide, for example, glycosylations, acetylations, phosphorylations, and the like. Included within the definition are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), polypeptides with substituted linkages as well as other modifications known in the art, both naturally and non-naturally occurring. Ordinarily, such polypeptides will be at least about 50% homologous to the native 07CG27 sequence, preferably in excess of about 90%, and more preferably at least about 95% or 98% or more homologous. Also included are proteins encoded by DNA which hybridize under high or low stringency conditions, to 07CG27-encoding nucleic acids and closely related polypeptides or proteins retrieved by antisera to the 07CG27 protein(s).
An 07CG27 polypeptide may be that derived from any of the exons described herein which may be in isolated and/or purified form, free or substantially free of material with which it is naturally associated. The polypeptide may, if produced by expression in a prokaryotic cell or produced synthetically, lack native post-translational processing, such as glycosylation. Alternatively, the present invention is also directed to polypeptides which are sequence variants, alleles or derivatives of an 07CG27 polypeptide. Such polypeptides may have an amino acid sequence which differs from that derived form any of the exons described herein by one or more of addition, substitution, deletion or insertion of one or more amino acids. Preferred such polypeptides have 07CG27 function. Substitutional variants typically contain the exchange of one amino acid for another at one or more sites within the protein, and may be designed to modulate one or more properties of the polypeptide, such as stability against proteolytic cleavage, without the loss of other functions or properties. Amino acid substitutions may be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophihcity, and/or the amphipathic nature of the residues involved. Preferred substitutions are ones which are conservative, that is, one amino acid is replaced with one of similar shape and charge. Conservative substitutions are well known in the art and typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and tyrosine, phenylalanine.
Certain amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies or binding sites on substrate molecules or binding sites on proteins interacting with an 07CG27 polypeptide. Since it is the interactive capacity and nature of a protein which defines that protein's biological functional activity, certain amino acid substitutions can be made in a protein sequence, and its underlying DNA coding sequence, and nevertheless obtain a protein with like properties. In making such changes, the hydropathic index of amino acids may be considered. The importance of the hydrophobic amino acid index in conferring interactive biological function on a protein is generally understood in the art (Kyte and Doolittle, 1982). Alternatively, the substitution of like amino acids can be made effectively on the basis of hydrophihcity. The importance of hydrophihcity in conferring interactive biological function of a protein is generally understood in the art (U.S. Patent 4,554,101). The use of the hydrophobic index or hydrophihcity in designing polypeptides is further discussed in U.S. Patent 5,691,198. The length of polypeptide sequences compared for homology will generally be at least about 16 amino acids, usually at least about 20 residues, more usually at least about 24 residues, typically at least about 28 residues, and preferably more than about 35 residues.
"Operably linked" refers to a juxtaposition wherein the components so described are in a relationship permitting them to function in their intended manner. For instance, a promoter is operably linked to a coding sequence if the promoter affects its transcription or expression.
The term peptide mimetic or mimetic is intended to refer to a substance which has the essential biological activity of an 07CG27 polypeptide. A peptide mimetic may be a peptide- containing molecule that mimics elements of protein secondary structure (Johnson et al., 1993). The underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions, such as those of antibody and antigen, enzyme and substrate or scaffolding proteins. A peptide mimetic is designed to permit molecular interactions similar to the natural molecule. A mimetic may not be a peptide at all, but it will retain the essential biological activity of a natural 07CG27 polypeptide.
"Probes". Polynucleotide polymorphisms associated with 07CG27 alleles which predispose to certain cancers or are associated with most cancers are detected by hybridization with a polynucleotide probe which forms a stable hybrid with that of the target sequence, under highly stringent to moderately stringent hybridization and wash conditions. If it is expected that the probes will be perfectly complementary to the target sequence, high stringency conditions will be used. Hybridization stringency may be lessened if some mismatching is expected, for example, if variants are expected with the result that the probe will not be completely complementary. Conditions are chosen which rule out nonspecific/adventitious bindings, that is, which minimize noise. (It should be noted that throughout this disclosure, if it is simply stated that "stringent" conditions are used that is meant to be read as "high stringency" conditions are used.) Since such indications identify neutral DNA polymorphisms as well as mutations, these indications need further analysis to demonstrate detection of a 07CG27 predisposing allele. Probes for 07CG27 alleles may be derived from the sequences of the 07CG27 region or its cDNAs. The probes may be of any suitable length, which span all or a portion of the 07CG27 region, and which allow specific hybridization to the 07CG27 region. If the target sequence contains a sequence identical to that of the probe, the probes may be short, e.g., in the range of about 8-30 base pairs, since the hybrid will be relatively stable under even highly stringent conditions. If some degree of mismatch is expected with the probe, i.e., if it is suspected that the probe will hybridize to a variant region, a longer probe may be employed which hybridizes to the target sequence with the requisite specificity.
The probes will include an isolated polynucleotide attached to a label or reporter molecule and may be used to isolate other polynucleotide sequences, having sequence similarity by standard methods. For techniques for preparing and labeling probes see, e.g., Sambrook et al, 1989 or Ausubel et al, 1992. Other similar polynucleotides may be selected by using homologous polynucleotides. Alternatively, polynucleotides encoding these or similar polypeptides may be synthesized or selected by use of the redundancy in the genetic code. Various codon substitutions may be introduced, e.g., by silent changes (thereby producing various restriction sites) or to optimize expression for a particular system. Mutations may be introduced to modify the properties of the polypeptide, perhaps to change ligand-binding affinities, interchain affinities, or the polypeptide degradation or turnover rate.
Probes comprising synthetic oligonucleotides or other polynucleotides of the present invention may be derived from naturally occurring or recombinant single- or double-stranded polynucleotides, or be chemically synthesized. Probes may also be labeled by nick translation, Klenow fill-in reaction, or other methods known in the art. Portions of the polynucleotide sequence having at least about eight nucleotides, usually at least about 15 nucleotides, and fewer than about 6 kb, usually fewer than about 1.0 kb, from a polynucleotide sequence encoding 07CG27 are preferred as probes. Thus, this definition includes probes of 8, 12, 15, 20, 25, 40, 60, 80, 100, 200, 300, 400 or 500 nucleotides or probes having any number of nucleotides within these ranges of values (e.g., 9, 10, 11, 16, 23, 30, 38, 50, 72, 121, etc., nucleotides), or probes having more than 500 nucleotides. The probes may also be used to determine whether mRNA encoding 07CG27 is present in a cell or tissue. The present invention includes all novel probes having at least 8 nucleotides derived from SEQ ID NO:l, its complement or functionally equivalent nucleic acid sequences. The present invention does not include probes which exist in the prior art. That is, the present invention includes all probes having at least 8 nucleotides derived from SEQ ID NO: 1 with the proviso that they do not include probes existing in the prior art.
Similar considerations and nucleotide lengths are also applicable to primers which may be used for the amplification of all or part of the 07CG27 gene. Thus, a definition for primers includes primers of 8, 12, 15, 20, 25, 40, 60, 80, 100, 200, 300, 400, 500 nucleotides, or primers having any number of nucleotides within these ranges of values (e.g., 9, 10, 11, 16, 23, 30, 38, 50, 72, 121, etc. nucleotides), or primers having more than 500 nucleotides, or any number of nucleotides between 500 and 9000. The primers may also be used to determine whether mRNA encoding 07CG27 is present in a cell or tissue. The present invention includes all novel primers having at least 8 nucleotides derived from the 07CG27 locus for amplifying the 07CG27 gene, its complement or functionally equivalent nucleic acid sequences. The present invention does not include primers which exist in the prior art. That is, the present invention includes all primers having at least 8 nucleotides with the proviso that it does not include primers existing in the prior art.
"Protein modifications or fragments" are provided by the present invention for 07CG27 polypeptides or fragments thereof which are substantially homologous to primary structural sequence but which include, e.g., in vivo or in vitro chemical and biochemical modifications or which incorporate unusual amino acids. Such modifications include, for example, acetylation, carboxylation, phosphorylation, glycosylation, ubiquitination, labeling, e.g., with radionuclides, and various enzymatic modifications, as will be readily appreciated by those well skilled in the art. A variety of methods for labeling polypeptides and of substituents or labels useful for such purposes are well known in the art, and include radioactive isotopes such as P, ligands which bind to labeled antiligands (e.g., antibodies), fluorophores, chemiluminescent agents, enzymes, and antiligands which can serve as specific binding pair members for a labeled ligand. The choice of label depends on the sensitivity required, ease of conjugation with the primer, stability requirements, and available instrumentation. Methods of labeling polypeptides are well known in the art. See Sambrook et al, 1989 or Ausubel et al, 1992.
Besides substantially full-length polypeptides, the present invention provides for biologically active fragments of the polypeptides. Significant biological activities include ligand-binding, immunological activity and other biological activities characteristic of 07CG27 polypeptides. Immunological activities include both immunogenic function in a target immune system, as well as sharing of immunological epitopes for binding, serving as either a competitor or substitute antigen for an epitope of the 07CG27 protein. As used herein, "epitope" refers to an antigenic determinant of a polypeptide. An epitope could comprise three amino acids in a spatial conformation which is unique to the epitope. Generally, an epitope consists of at least five such amino acids, and more usually consists of at least 8-10 such amino acids. Methods of determining the spatial conformation of such amino acids are known in the art.
For immunological purposes, tandem-repeat polypeptide segments may be used as immunogens, thereby producing highly antigenic proteins. Alternatively, such polypeptides will serve as highly efficient competitors for specific binding. Production of antibodies specific for 07CG27 polypeptides or fragments thereof is described below.
The present invention also provides for fusion polypeptides, comprising 07CG27 polypeptides and fragments. Homologous polypeptides may be fusions between two or more 07CG27 polypeptide sequences or between the sequences of 07CG27 and a related protein. Likewise, heterologous fusions may be constructed which would exhibit a combination of properties or activities of the derivative proteins. For example, ligand-binding or other domains may be "swapped" between different new fusion polypeptides or fragments. Such homologous or heterologous fusion polypeptides may display, for example, altered strength or specificity of binding. Fusion partners include immunoglobulins, bacterial b-galactosidase, trpE, protein A, b- lactamase, alpha amylase, alcohol dehydrogenase and yeast alpha mating factor. See Godowski et al, 1988.
Fusion proteins will typically be made by either recombinant nucleic acid methods, as described below, or may be chemically synthesized. Techniques for the synthesis of polypeptides are described, for example, in Merrifield, 1963.
"Protein purification" refers to various methods for the isolation of the 07CG27 polypeptides from other biological material, such as from cells transformed with recombinant nucleic acids encoding 07CG27, and are well known in the art. For example, such polypeptides may be purified by immunoaffinity chromatography employing, e.g., the antibodies provided by the present invention. Various methods of protein purification are well known in the art, and include those described in Deutscher, 1990 and Scopes, 1982.
The terms "isolated", "substantially pure", and "substantially homogeneous" are used interchangeably to describe a protein or polypeptide which has been separated from components which accompany it in its natural state. A monomeric protein is substantially pure when at least about 60 to 75% of a sample exhibits a single polypeptide sequence. A substantially pure protein will typically comprise about 60 to 90% W/W of a protein sample, more usually about 95%, and preferably will be over about 99% pure. Protein purity or homogeneity may be indicated by a number of means well known in the art, such as polyacrylamide gel electrophoresis of a protein sample, followed by visualizing a single polypeptide band upon staining the gel. For certain purposes, higher resolution may be provided by using HPLC or other means well known in the art which are utilized for purification.
A 07CG27 protein is substantially free of naturally associated components when it is separated from the native contaminants which accompany it in its natural state. Thus, a polypeptide which is chemically synthesized or synthesized in a cellular system different from the cell from which it naturally originates will be substantially free from its naturally associated components. A protein may also be rendered substantially free of naturally associated components by isolation, using protein purification techniques well known in the art.
A polypeptide produced as an expression product of an isolated and manipulated genetic sequence is an "isolated polypeptide," as used herein, even if expressed in a homologous cell type. Synthetically made forms or molecules expressed by heterologous cells are inherently isolated molecules.
"Recombinant nucleic acid" is a nucleic acid which is not naturally occurring, or which is made by the artificial combination of two otherwise separated segments of sequence. This artificial combination is often accomplished by either chemical synthesis means, or by the artificial manipulation of isolated segments of nucleic acids, e.g., by genetic engineering techniques. Such is usually done to replace a codon with a redundant codon encoding the same or a conservative amino acid, while typically introducing or removing a sequence recognition site. Alternatively, it is performed to join together nucleic acid segments of desired functions to generate a desired combination of functions. "Regulatory sequences" refers to those sequences normally within 100 kb of the coding region of a locus, but they may also be more distant from the coding region, which affect the expression of the gene (including transcription of the gene, and translation, splicing, stability or the like of the messenger RNA).
"Substantial homology or similarity". A nucleic acid or fragment thereof is "substantially homologous" ("or substantially similar") to another if, when optimally aligned (with appropriate nucleotide insertions or deletions) with the other nucleic acid (or its complementary strand), there is nucleotide sequence identity in at least about 60% of the nucleotide bases, usually at least about 70%, more usually at least about 80%, preferably at least about 90%), and more preferably at least about 95-98% of the nucleotide bases. Identity means the degree of sequence relatedness between two polypeptide or two polynucleotides sequences as determined by the identity of the match between two strings of such sequences. Identity can be readily calculated. While there exist a number of methods to measure identity between two polynucleotide or polypeptide sequences, the term "identity" is well known to skilled artisans (Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A. M., and Griffin, H. G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991). Methods commonly employed to determine identity between two sequences include, but are not limited to those disclosed in Guide to Huge Computers. Martin J. Bishop, ed., Academic Press, San Diego, 1994, and Carillo, H., and Lipman, D. (1988). Preferred methods to determine identity are designed to give the largest match between the two sequences tested. Such methods are codified in computer programs. Preferred computer program methods to determine identity between two sequences include, but are not limited to, GCG program package (Devereux et al. (1984), BLASTP, BLASTN, FASTA (Atschul et al. (1990)). Alternatively, substantial homology or similarity exists when a nucleic acid or fragment thereof will hybridize to another nucleic acid (or a complementary strand thereof) under selective hybridization conditions, to a strand, or to its complement. Selectivity of hybridization exists when hybridization which is substantially more selective than total lack of specificity occurs. Typically, selective hybridization will occur when there is at least about 55% homology over a stretch of at least about 14 nucleotides, preferably at least about 65%, more preferably at least about 75%, and most preferably at least about 90%. See, Kanehisa, 1984. The length of homology comparison, as described, may be over longer stretches, and in certain embodiments will often be over a stretch of at least about nine nucleotides, usually at least about 20 nucleotides, more usually at least about 24 nucleotides, typically at least about 28 nucleotides, more typically at least about 32 nucleotides, and preferably at least about 36 or more nucleotides.
Nucleic acid hybridization will be affected by such conditions as salt concentration, temperature, or organic solvents, in addition to the base composition, length of the complementary strands, and the number of nucleotide base mismatches between the hybridizing nucleic acids, as will be readily appreciated by those skilled in the art. Stringent temperature conditions will generally include temperatures in excess of 30°C, typically in excess of 37°C, and preferably in excess of 45°C. Stringent salt conditions will ordinarily be less than 1000 mM, typically less than 500 mM, and preferably less than 200 mM. However, the combination of parameters is much more important than the measure of any single parameter. See, e.g., Wetmur and Davidson, 1968. See also Ausubel et al, 1992. Probe sequences may also hybridize specifically to duplex DNA under certain conditions to form triplex or other higher order DNA complexes. The preparation of such probes and suitable hybridization conditions are well known in the art.
The terms "substantial homology" or "substantial identity", when referring to polypeptides, indicate that the polypeptide or protein in question exhibits at least about 30% identity with an entire naturally-occurring protein or a portion thereof, usually at least about 70% identity, and preferably at least about 95% identity.
Homology, for polypeptides, is typically measured using sequence analysis software. See, e.g., the Sequence Analysis Software Package of the Genetics Computer Group, University of Wisconsin Biotechnology Center, 910 University Avenue, Madison, Wisconsin 53705 and the references cited above with respect to nucleic acid homology. Protein analysis software matches similar sequences using measures of homology assigned to various substitutions, deletions and other modifications. Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine. Methods commonly employed to determine identity between two sequences include, but are not limited to those disclosed in Guide to Huge Computers, Martin J. Bishop, ed., Academic Press, San Diego, 1994, and Carillo, H., and Lipman, D. (1988). Preferred methods to determine identity are designed to give the largest match between the two sequences tested. Such methods are codified in computer programs. Preferred computer program methods to determine identity between two sequences include, but are not limited to, GCG program package (Devereux et al. (1984), BLASTP, BLASTN, FASTA (Atschul et al. (1990)).
"Substantially similar function" refers to the function of a modified nucleic acid or a modified protein, with reference to the wild-type 07CG27 nucleic acid or wild-type 07CG27 polypeptide. The modified polypeptide will be substantially homologous to the wild-type 07CG27 polypeptide and will have substantially the same function. The modified polypeptide may have an altered amino acid sequence and/or may contain modified amino acids. In addition to the similarity of function, the modified polypeptide may have other useful properties, such as a longer half-life. The similarity of function (activity) of the modified polypeptide may be substantially the same as the activity of the wild-type 07CG27 polypeptide. Alternatively, the similarity of function (activity) of the modified polypeptide may be higher than the activity of the wild-type 07CG27 polypeptide. The modified polypeptide is synthesized using conventional techniques, or is encoded by a modified nucleic acid and produced using conventional techniques. The modified nucleic acid is prepared by conventional techniques. A nucleic acid with a function substantially similar to the wild-type 07CG27 gene function produces the modified protein described above. A polypeptide "fragment," "portion" or "segment" is a stretch of amino acid residues of at least about five to seven contiguous amino acids, often at least about seven to nine contiguous amino acids, typically at least about nine to 13 contiguous amino acids and, most preferably, at least about 20 to 30 or more contiguous amino acids.
The polypeptides of the present invention, if soluble, may be coupled to a solid-phase support, e.g., nitrocellulose, nylon, column packing materials (e.g., Sepharose beads), magnetic beads, glass wool, plastic, metal, polymer gels, cells, or other substrates. Such supports may take the form, for example, of beads, wells, dipsticks, or membranes.
"Target region" refers to a region of the nucleic acid which is amplified and/or detected. The term "target sequence" refers to a sequence with which a probe or primer will form a stable hybrid under desired conditions.
The practice of the present invention employs, unless otherwise indicated, conventional techniques of chemistry, molecular biology, microbiology, recombinant DNA, genetics, and immunology. See, e.g., Maniatis et al, 1982; Sambrook et al, 1989; Ausubel et al, 1992; Glover, 1985; Anand, 1992; Guthrie and Fink, 1991. A general discussion of techniques and materials for human gene mapping, including mapping of human chromosome 1, is provided, e.g., in White and Lalouel, 1988.
Preparation Of Recombinant Or Chemically Synthesized Nucleic Acids. Vectors. Transformation And Host Cells
Large amounts of the polynucleotides of the present invention may be produced by replication in a suitable host cell. Natural or synthetic polynucleotide fragments coding for a desired fragment will be incorporated into recombinant polynucleotide constructs, usually DNA constructs, capable of introduction into and replication in a prokaryotic or eukaryotic cell.
Usually the polynucleotide constructs will be suitable for replication in a unicellular host, such as yeast or bacteria, but may also be intended for introduction to (with and without integration within the genome) cultured mammalian or plant or other eukaryotic cell lines. The purification of nucleic acids produced by the methods of the present invention is described, e.g., in
Sambrook et al, 1989 or Ausubel et al, 1992. The polynucleotides of the present invention may also be produced by chemical synthesis, e.g., by the phosphoramidite method described by Beaucage and Carruthers, 1981 or the triester method according to Matteucci and Caruthers, 1981, and may be performed on commercial, automated oligonucleotide synthesizers. A double-stranded fragment may be obtained from the single-stranded product of chemical synthesis either by synthesizing the complementary strand and annealing the strands together under appropriate conditions or by adding the complementary strand using DNA polymerase with an appropriate primer sequence.
Polynucleotide constructs prepared for introduction into a prokaryotic or eukaryotic host may comprise a replication system recognized by the host, including the intended polynucleotide fragment encoding the desired polypeptide, and will preferably also include transcription and translational initiation regulatory sequences operably linked to the polypeptide encoding segment. Expression vectors may include, for example, an origin of replication or autonomously replicating sequence (ARS) and expression control sequences, a promoter, an enhancer and necessary processing information sites, such as ribosome-binding sites, RNA splice sites, polyadenylation sites, transcriptional terminator sequences, and mRNA stabilizing sequences. Secretion signals may also be included where appropriate, whether from a native 07CG27 protein or from other receptors or from secreted polypeptides of the same or related species, which allow the protein to cross and/or lodge in cell membranes, and thus attain its functional topology, or be secreted from the cell. Such vectors may be prepared by means of standard recombinant techniques well known in the art and discussed, for example, in Sambrook et al, 1989 or Ausubel et al. 1992.
An appropriate promoter and other necessary vector sequences will be selected so as to be functional in the host, and may include, when appropriate, those naturally associated with 07CG27 genes. Examples of workable combinations of cell lines and expression vectors are described in Sambrook et al, 1989 or Ausubel et al, 1992; see also, e.g., Metzger et al, 1988. Many useful vectors are known in the art and may be obtained from such vendors as Stratagene, New England BioLabs, Promega Biotech, and others. Promoters such as the tip, lac and phage promoters, tRNA promoters and glycolytic enzyme promoters may be used in prokaryotic hosts. Useful yeast promoters include promoter regions for metallothionein, 3-phosphoglycerate kinase or other glycolytic enzymes such as enolase or glyceraldehyde-3-phosphate dehydrogenase, enzymes responsible for maltose and galactose utilization, and others. Vectors and promoters suitable for use in yeast expression are further described in Hitzeman et al, EP 73, 675 A. Appropriate non-native mammalian promoters might include the early and late promoters from SV40 (Fiers et al, 1978) or promoters derived from murine Moloney leukemia virus, mouse tumor virus, avian sarcoma viruses, adenovirus II, bovine papilloma virus or polyoma. In addition, the construct may be joined to an amplifiable gene (e.g., DHFR) so that multiple copies of the gene may be made. For appropriate enhancer and other expression control sequences, see also Enhancers and Eukaryotic Gene Expression, Cold Spring Harbor Press, Cold Spring Harbor, New York (1983). See also, e.g., U.S. Patent Nos. 5,691,198; 5,735,500; 5,747,469 and 5,436,146.
While such expression vectors may replicate autonomously, they may also replicate by being inserted into the genome of the host cell, by methods well known in the art.
Expression and cloning vectors will likely contain a selectable marker, a gene encoding a protein necessary for survival or growth of a host cell transformed with the vector. The presence of this gene ensures growth of only those host cells which express the inserts. Typical selection genes encode proteins that a) confer resistance to antibiotics or other toxic substances, e.g. ampicillin, neomycin, methotrexate, etc.; b) complement auxotrophic deficiencies, or c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli. The choice of the proper selectable marker will depend on the host cell, and appropriate markers for different hosts are well known in the art.
The vectors containing the nucleic acids of interest can be transcribed in vitro, and the resulting RNA introduced into the host cell by well-known methods, e.g., by injection (see, Kubo et al, 1988), or the vectors can be introduced directly into host cells by methods well known in the art, which vary depending on the type of cellular host, including electroporation; transfection employing calcium chloride, rubidium chloride, calcium phosphate, DEAE-dextran, or other substances; microprojectile bombardment; lipofection; infection (where the vector is an infectious agent, such as a retroviral genome); and other methods. See generally, Sambrook et al, 1989 and Ausubel et al, 1992. The introduction of the polynucleotides into the host cell by any method known in the art, including, inter alia, those described above, will be referred to herein as "transformation." The cells into which have been introduced nucleic acids described above are meant to also include the progeny of such cells. Large quantities of the nucleic acids and polypeptides of the present invention may be prepared by expressing the 07CG27 nucleic acids or portions thereof in vectors or other expression vehicles in compatible prokaryotic or eukaryotic host cells. The most commonly used prokaryotic hosts are strains of Escherichia coli, although other prokaryotes, such as Bacillus subtilis or Pseudomonas may also be used.
Mammalian or other eukaryotic host cells, such as those of yeast, filamentous fungi, plant, insect, or amphibian or avian species, may also be useful for production of the proteins of the present invention. Propagation of mammalian cells in culture is per se well known. See, Jakoby and Pastan, 1979. Examples of commonly used mammalian host cell lines are VERO and HeLa cells, Chinese hamster ovary (CHO) cells, and WI38, BHK, and COS cell lines. An example of a commonly used insect cell line is SF9. However, it will be appreciated by the skilled practitioner that other cell lines may be appropriate, e.g., to provide higher expression, desirable glycosylation patterns, or other features.
Clones are selected by using markers depending on the mode of the vector construction.
The marker may be on the same or a different DNA molecule, preferably the same DNA molecule. In prokaryotic. hosts, the transformant may be selected, e.g., by resistance to ampicillin, tetracycline or other antibiotics. Production of a particular product based on temperature sensitivity may also serve as an appropriate marker.
Prokaryotic or eukaryotic cells transformed with the polynucleotides of the present invention will be useful not only for the production of the nucleic acids and polypeptides of the present invention, but also, for example, in studying the characteristics of 07CG27 polypeptides.
Antisense polynucleotide sequences are useful in preventing or diminishing the expression of the 07CG27 locus, as will be appreciated by those skilled in the art. For example, polynucleotide vectors containing all or a portion of the 07CG27 locus or other sequences from the 07CG27 region (particularly those flanking the 07CG27 locus) may be placed under the control of a promoter in an antisense orientation and introduced into a cell. Expression of such an antisense construct within a cell will interfere with 07CG27 transcription and/or translation and/or replication.
The probes and primers based on the 07CG27 gene sequences disclosed herein are used to identify homologous 07CG27 gene sequences and proteins in other species. These 07CG27 gene sequences and proteins are used in the diagnostic/prognostic, therapeutic and drug screening methods described herein for the species from which they have been isolated. Methods of Use: Nucleic Acid Diagnosis and Diagnostic Kits
In order to detect the presence of an 07CG27 allele predisposing an individual to disease, a biological sample such as blood is prepared and analyzed for the presence or absence of predisposing alleles of 07CG27. Results of these tests and interpretive information are returned to the health care provider for communication to the tested individual. Such diagnoses may be performed by diagnostic laboratories, or, alternatively, diagnostic kits are manufactured and sold to health care providers or to private individuals for self-diagnosis. Initially, the screening method involves amplification of the relevant 07CG27 sequences.
In another preferred embodiment of the invention, the screening method involves a non-PCR based strategy. Such screening methods include two-step label amplification methodologies that are well known in the art. Both PCR and non-PCR based screening strategies can detect target sequences with a high level of sensitivity. The most popular method used today is target amplification. Here, the target nucleic acid sequence is amplified with polymerases. One particularly preferred method using polymerase- driven amplification is the polymerase chain reaction (PCR). The polymerase chain reaction and other polymerase-driven amplification assays can achieve over a million-fold increase in copy number through the use of polymerase-driven amplification cycles. Once amplified, the resulting nucleic acid can be sequenced or used as a substrate for DNA probes.
When the probes are used to detect the presence of the target sequences (for example, in screening for disease susceptibility), the biological sample to be analyzed, such as blood or serum, may be treated, if desired, to extract the nucleic acids. The sample nucleic acid may be prepared in various ways to facilitate detection of the target sequence; e.g. denaturation, restriction digestion, electrophoresis or dot blotting. The targeted region of the analyte nucleic acid usually must be at least partially single-stranded to form hybrids with the targeting sequence of the probe. If the sequence is naturally single-stranded, denaturation will not be required. However, if the sequence is double-stranded, the sequence will probably need to be denatured. Denaturation can be carried out by various techniques known in the art. Analyte nucleic acid and probe are incubated under conditions which promote stable hybrid formation of the target sequence in the probe with the putative targeted sequence in the analyte. The region of the probes which is used to bind to the analyte can be made completely complementary to the targeted region of human chromosome 1. Therefore, high stringency conditions are desirable in order to prevent false positives. However, conditions of high stringency are used only if the probes are complementary to regions of the chromosome which are unique in the genome. The stringency of hybridization is determined by a number of factors during hybridization and during the washing procedure, including temperature, ionic strength, base composition, probe length, and concentration of formamide. These factors are outlined in, for example, Maniatis et al, 1982 and Sambrook et al, 1989. Under certain circumstances, the formation of higher order hybrids, such as triplexes, quadraplexes, etc., may be desired to provide the means of detecting target sequences. Detection, if any, of the resulting hybrid is usually accomplished by the use of labeled probes. Alternatively, the probe may be unlabeled, but may be detectable by specific binding with a ligand which is labeled, either directly or indirectly. Suitable labels, and methods for labeling probes and ligands are known in the art, and include, for example, radioactive labels which may be incorporated by known methods (e.g., nick translation, random priming or kinasing), biotin, fluorescent groups, chemilummescent groups (e.g., dioxetanes, particularly triggered dioxetanes), enzymes, antibodies and the like. Variations of this basic scheme are known in the art, and include those variations that facilitate separation of the hybrids to be detected from extraneous materials and/or that amplify the signal from the labeled moiety. A number of these variations are reviewed in, e.g., Matthews and Kricka, 1988; Landegren et al, 1988; Mittlin, 1989; U.S. Patent 4,868,105, and in EPO Publication No. 225,807.
As noted above, non-PCR based screening assays are also contemplated in this invention. This procedure hybridizes a nucleic acid probe (or an analog such as a methyl phosphonate backbone replacing the normal phosphodiester), to the low level DNA target. This probe may have an enzyme covalently linked to the probe, such that the covalent linkage does not interfere with the specificity of the hybridization. This enzyme-probe-conjugate-target nucleic acid complex can then be isolated away from the free probe enzyme conjugate and a substrate is added for enzyme detection. Enzymatic activity is observed as a change in color development or luminescent output resulting in a 10 -10 increase in sensitivity. For an example relating to the preparation of oligodeoxynucleotide-alkaline phosphatase conjugates and their use as hybridization probes see Jablonski et al, 1986.
Two-step label amplification methodologies are known in the art. These assays work on the principle that a small ligand (such as digoxigenin, biotin, or the like) is attached to a nucleic acid probe capable of specifically binding 07CG27. Allele specific probes are also contemplated within the scope of this example and exemplary allele specific probes include probes encompassing the predisposing or potentially predisposing mutations summarized in herein.
In one example, the small ligand attached to the nucleic acid probe is specifically recognized by an antibody-enzyme conjugate. In one embodiment of this example, digoxigenin is attached to the nucleic acid probe. Hybridization is detected by an antibody-alkaline phosphatase conjugate which turns over a chemilummescent substrate. For methods for labeling nucleic acid probes according to this embodiment see Martin et al, 1990. In a second example, the small ligand is recognized by a second ligand-enzyme conjugate that is capable of specifically complexing to the first ligand. A well known embodiment of this example is the biotin-avidin type of interactions. For methods for labeling nucleic acid probes and their use in biotin-avidin based assays see Rigby et al, 1977 and Nguyen et al, 1992.
It is also contemplated within the scope of this invention that the nucleic acid probe assays of this invention will employ a cocktail of nucleic acid probes capable of detecting 07CG27. Thus, in one example to detect the presence of 07CG27 in a cell sample, more than one probe complementary to 07CG27 is employed and in particular the number of different probes is alternatively 2, 3, or 5 different nucleic acid probe sequences. In another example, to detect the presence of mutations in the 07CG27 gene sequence in a patient, more than one probe complementary to 07CG27 is employed where the cocktail includes probes capable of binding to the allele-specific mutations identified in populations of patients with alterations in 07CG27. In this embodiment, any number of probes can be used, and will preferably include probes corresponding to the major gene mutations identified as predisposing an individual to prostate cancer. Some candidate probes contemplated within the scope of the invention include probes that include the allele-specific mutations identified herein and those that have the 07CG27 regions corresponding to SEQ ID NO:l both 5' and 3' to a mutation site.
Methods of Use: Peptide Diagnosis and Diagnostic Kits The neoplastic condition of lesions can also be detected on the basis of the alteration of wild-type 07CG27 polypeptide. Such alterations can be determined by sequence analysis in accordance with conventional techniques. More preferably, antibodies (polyclonal or monoclonal) are used to detect differences in, or the absence of, 07CG27 peptides. The antibodies may be prepared as discussed above under the heading "Antibodies" and as further shown in the Examples infra. Other techniques for raising and purifying antibodies are well known in the art and any such techniques may be chosen to achieve the preparations claimed in this invention. In a preferred embodiment of the invention, antibodies will immunoprecipitate 07CG27 proteins from solution as well as react with 07CG27 protein on Western or immunoblots of polyacrylamide gels. In another preferred embodiment, antibodies will detect 07CG27 proteins in paraffin or frozen tissue sections, using immunocytochemical techniques. Preferred embodiments relating to methods for detecting 07CG27 or its mutations include enzyme linked immunosorbent assays (ELISA), radioimmunoassays (RIA), immunoradiometric assays (IRMA) and immunoenzymatic assays (IEMA), including sandwich assays using monoclonal and/or polyclonal antibodies. Exemplary sandwich assays are described by David et al. in U.S. Patent Nos. 4,376,110 and 4,486,530, hereby incorporated by reference, and exemplified in Example 15.
Methods of Use: Drug Screening This invention is particularly useful for screening compounds by using a wild-type or mutant 07CG27 polypeptide or binding fragment thereof in any of a variety of drug screening techniques.
The 07CG27 polypeptide or fragment employed in such a test may either be free in solution, affixed to a solid support, or borne on a cell surface. One method of drug screening utilizes eucaryotic or procaryotic host cells which are stably transformed with recombinant polynucleotides expressing the polypeptide or fragment, preferably in competitive binding assays. Such cells, either in viable or fixed form, can be used for standard binding assays. One may measure, for example, for the formation of complexes between an 07CG27 polypeptide or fragment and the agent being tested, or examine the degree to which the formation of a complex between an 07CG27 polypeptide or fragment and a known ligand, e.g. ras, is interfered with by the agent being tested. Thus, the present invention provides methods of screening for drugs comprising contacting such an agent with an 07CG27 polypeptide or fragment thereof and assaying (i) for the presence of a complex between the agent and the 07CG27 polypeptide or fragment, or (ii) for the presence of a complex between the 07CG27 polypeptide or fragment and a ligand, by methods well known in the art. In such competitive binding assays the 07CG27 polypeptide or fragment is typically labeled. Free 07CG27 polypeptide or fragment is separated from that present in a proteimprotein complex, and the amount of free (i.e., uncomplexed) label is a measure of the binding of the agent being tested to 07CG27 or its interference with 07CG27:ligand binding, respectively. Since 07CG27 is a GEF protein and activates ras, the effect of a drug candidate can be determined by measuring ras activation by a wild-type or mutant 07CG27 polypeptide.
Another technique for drug screening provides high throughput screening for compounds having suitable binding affinity to the 07CG27 polypeptides and is described in detail in Geysen, PCT published application WO 84/03564, published on September 13, 1984. Briefly stated, large numbers of different small peptide test compounds are synthesized on a solid substrate, such as plastic pins or some other surface. The peptide test compounds are reacted with 07CG27 polypeptide and washed. Bound 07CG27 polypeptide is then detected by methods well known in the art.
Purified 07CG27 can be coated directly onto plates for use in the aforementioned drug screening techniques. However, non-neutralizing antibodies to the polypeptide can be used to capture antibodies to immobilize the 07CG27 polypeptide on the solid phase.
This invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of specifically binding the 07CG27 polypeptide compete with a test compound for binding to the 07CG27 polypeptide or fragments thereof. In this manner, the antibodies can be used to detect the presence of any peptide which shares one or more antigenic determinants of the 07CG27 polypeptide.
A further technique for drug screening involves the use of host eukaryotic cell lines or cells (such as described above) which have a wild-type or mutant 07CG27 gene. The host cell lines or cells are grown in the presence of drug compound. The ras activation occurring in the host cells is measured to determine if the compound is capable of regulating the interaction of 07CG27 and ras.
Briefly, a method of screening for a substance which modulates activity of a polypeptide may include contacting one or more test substances with the polypeptide in a suitable reaction medium, testing the activity of the treated polypeptide and comparing that activity with the activity of the polypeptide in comparable reaction medium untreated with the test substance or substances. A difference in activity between the treated and untreated polypeptides is indicative of a modulating effect of the relevant test substance or substances. Prior to or as well as being screened for modulation of activity, test substances may be screened for ability to interact with the polypeptide, e.g., in a yeast two-hybrid system (e.g., Bartel et al., 1993; Fields and Song, 1989; Chevray and Nathans, 1992; Lee et al, 1995). This system may be used as a coarse screen prior to testing a substance for actual ability to modulate activity of the polypeptide. Alternatively, the screen could be used to screen test substances for binding to an 07CG27 specific binding partner, or to find mimetics of an 07CG27 polypeptide. Methods of Use: Rational Drug Design The goal of rational drug design is to produce structural analogs of biologically active polypeptides of interest or of small molecules with which they interact (e.g., agonists, antagonists, inhibitors) in order to fashion drugs which are, for example, more active or stable forms of the polypeptide, or which, e.g., enhance or interfere with the function of a polypeptide in vivo. See, e.g., Hodgson, 1991. In one approach, one first determines the three-dimensional structure of a protein of interest (e.g., 07CG27 polypeptide) or, for example, of the 07CG27- receptor or ligand complex, by x-ray crystallography, by computer modeling or most typically, by a combination of approaches. Less often, useful information regarding the structure of a polypeptide may be gained by modeling based on the structure of homologous proteins. An example of rational drug design is the development of HIV protease inhibitors (Erickson et al, 1990). In addition, peptides (e.g., 07CG27 polypeptide) are analyzed by an alanine scan (Wells, 1991). In this technique, an amino acid residue is replaced by Ala, and its effect on the peptide's activity is determined. Each of the amino acid residues of the peptide is analyzed in this manner to determine the important regions of the peptide.
It is also possible to isolate a target-specific antibody, selected by a functional assay, and then to solve its crystal structure. In principle, this approach yields a pharmacore upon which subsequent drug design can be based. It is possible to bypass protein crystallography altogether by generating anti-idiotypic antibodies (anti-ids) to a functional, pharmacologically active antibody. As a mirror image of a mirror image, the binding site of the anti-ids would be expected to be an analog of the original receptor. The anti-id could then be used to identify and isolate peptides from banks of chemically or biologically produced banks of peptides. Selected peptides would then act as the pharmacore.
Thus, one may design drugs which have, e.g., improved 07CG27 polypeptide activity or stability or which act as inhibitors, agonists, antagonists, etc. of 07CG27 polypeptide activity. By virtue of the availability of cloned 07CG27 sequences, sufficient amounts of the 07CG27 polypeptide may be made available to perform such analytical studies as x-ray crystallography. In addition, the knowledge of the 07CG27 protein sequence provided herein will guide those employing computer modeling techniques in place of, or in addition to x-ray crystallography. Following identification of a substance which modulates or affects polypeptide activity, the substance may be investigated further. Furthermore, it may be manufactured and/or used in preparation, i.e., manufacture or formulation, or a composition such as a medicament, pharmaceutical composition or drug. These may be administered to individuals.
Thus, the present invention extends in various aspects not only to a substance identified using a nucleic acid molecule as a modulator of polypeptide activity, in accordance with what is disclosed herein, but also a pharmaceutical composition, medicament, drug or other composition comprising such a substance, a method comprising administration of such a composition comprising such a substance, a method comprising administration of such a composition to a patient, e.g., for treatment of prostate cancer, use of such a substance in the manufacture of a composition for administration, e.g., for treatment of prostate cancer, and a method of making a pharmaceutical composition comprising admixing such a substance with a pharmaceutically acceptable excipient, vehicle or carrier, and optionally other ingredients.
A substance identified as a modulator of polypeptide function may be peptide or non- peptide in nature. Non-peptide " small molecules" are often preferred for many in vivo pharmaceutical uses. Accordingly, a mimetic or mimic of the substance (particularly if a peptide) may be designed for pharmaceutical use.
The designing of mimetics to a known pharmaceutically active compound is a known approach to the development of pharmaceuticals based on a " lead" compound. This might be desirable where the active compound is difficult or expensive to synthesize or where it is unsuitable for a particular method of administration, e.g., pure peptides are unsuitable active agents for oral compositions as they tend to be quickly degraded by proteases in the alimentary canal. Mimetic design, synthesis and testing is generally used to avoid randomly screening large numbers of molecules for a target property.
There are several steps commonly taken in the design of a mimetic from a compound having a given target property. First, the particular parts of the compound that are critical and/or important in determining the target property are determined. In the case of a peptide, this can be done by systematically varying the amino acid residues in the peptide, e.g., by substituting each residue in turn. Alanine scans of peptide are commonly used to refine such peptide motifs. These parts or residues constituting the active region of the compound are known as its " pharmacophore" . Once the pharmacophore has been found, its structure is modeled according to its physical properties, e.g., stereochemistry, bonding, size and/or charge, using data from a range of sources, e.g., spectroscopic techniques, x-ray diffraction data and NMR. Computational analysis, similarity mapping (which models the charge and/or volume of a pharmacophore, rather than the bonding between atoms) and other techniques can be used in this modeling process.
In a variant of this approach, the three-dimensional structure of the ligand and its binding partner are modeled. This can be especially useful where the ligand and/or binding partner change conformation on binding, allowing the model to take account of this in the design of the mimetic.
A template molecule is then selected onto which chemical groups which mimic the pharmacophore can be grafted. The template molecule and the chemical groups grafted onto it can conveniently be selected so that the mimetic is easy to synthesize, is likely to be pharmacologically acceptable, and does not degrade in vivo, while retaining the biological activity of the lead compound. Alternatively, where the mimetic is peptide-based, further stability can be achieved by cyclizing the peptide, increasing its rigidity. The mimetic or mimetics found by this approach can then be screened to see whether they have the target property, or to what extent they exhibit it. Further optimization or modification can then be carried out to arrive at one or more final mimetics for in vivo or clinical testing.
Methods of Use: Antisensense Therapy The present invention contemplates an antisense polynucleotide up to about 50 nucleotides in length that hybridizes with mRNA molecules that encode a 07CG27 polypeptide, and the use of one or more of those polynucleotides in treating disease. See U.S. Patent Nos. 5,891,858 and 5,885,970, incorporated herein by reference, for further details. The antisense polynucleotide is useful for treating disease caused by a mutant 07CG27 as well as overexpression of a wild-type 07CG27.
In one embodiment an antisense polynucleotide is contacted with a cancer cells. The contact is carried out in vivo in a host mammal, and contact is effected by administration to the mammal of a pharmaceutical composition containing the polynucleotide dissolved or dispersed in a physiologically tolerable diluent so that a body fluid such as blood or lymph provides at least a portion of the aqueous medium. In vivo contact is maintained until the polynucleotide is eliminated from the mammal's body by a normal bodily function such as excretion in the urine or feces or enzymatic breakdown.
A polynucleotide can also be administered in the form of liposomes. As is shown in the art, liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by monoor multi-lamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used. The present compositions in liposome form can contain stabilizers, preservatives, excipients, and the like in addition to the agent. The preferred lipids are phospholipids and phosphatidyl cholines (lecithins), both natural and synthetic.
An antisense polynucleotide can also be administered by gene therapy. The polynucleotide may be introduced into the cell in a vector such that the polynucleotide remains extrachromosomal. In such a situation, the polynucleotide will be expressed by the cell from the extrachromosomal location. Vectors for introduction of polyucleotides for extrachromosomal maintenance are known in the art, and any suitable vector may be used. Methods for introducing DNA into cells such as electroporation, calcium phosphate coprecipitation and viral transduction are known in the art, and the choice of method is within the competence of the routineer.
The antisense polynucleotide, may be employed in gene therapy methods in order to decrease the amount of the expression products of 07CG27 in disease cells, especially in those cases where 07CG27 is overexpressed. Such gene therapy is particularly appropriate for use in both disease and pre-disease cells. It may also be useful to decrease the level of expression of 07CG27 even in those disease cells in which a wild-type gene is expressed at an elevated level.
Gene therapy would be carried out according to generally accepted methods, for example, as described in further detail in U.S. Patent No. 5,747,282 and references cited therein, all incorporated by reference herein. Expression vectors in the context of gene therapy are meant to include those constructs containing sequences sufficient to express a polynucleotide that has been cloned therein. In viral expression vectors, the construct contains viral sequences sufficient to support packaging of the construct. If the polynucleotide encodes an antisense polynucleotide or a ribozyme, expression will produce the antisense polynucleotide or ribozyme. Thus in this context, expression does not require that a protein product be synthesized. In addition to the polynucleotide cloned into the expression vector, the vector also contains a promoter functional in eukaryotic cells. The cloned polynucleotide sequence is under control of this promoter. Suitable eukaryotic promoters include those described above. The expression vector may also include sequences, such as selectable markers and other sequences conventionally used.
Gene transfer techniques which target DNA directly to, e.g. prostate tissues, e.g., epithelial cells of the prostate, are preferred. Receptor-mediated gene transfer, for example, is accomplished by the conjugation of DNA (usually in the form of covalently closed supercoiled plasmid) to a protein ligand via polylysine. Ligands are chosen on the basis of the presence of the corresponding ligand receptors on the cell surface of the target cell/tissue type. These ligand-DNA conjugates can be injected directly into the blood if desired and are directed to the target tissue where receptor binding and internalization of the DNA-protein complex occurs. To overcome the problem of intracellular destruction of DNA, coinfection with adenovirus can be included to disrupt endosome function.
Methods of Use: Transformed Hosts; Trans genic/Knockour Animals and Models Similarly, cells and animals which carry a mutant 07CG27 allele can be used as model systems to study and test for substances which have potential as therapeutic agents. These may be isolated from individuals with 07CG27 mutations, either somatic or germline. Alternatively, the cell line can be engineered to carry the mutation in the 07CG27 allele, as described above. After a test substance is applied to the cells, the phenotype of the cell is determined, using assays known in the art. Animals for testing therapeutic agents can be selected after mutagenesis of whole animals or after treatment of germline cells or zygotes. Such treatments include insertion of mutant 07CG27 alleles, usually from a second animal species, as well as insertion of disrupted homologous genes. Alternatively, the endogenous 07CG27 gene(s) of the animals may be disrupted by insertion or deletion mutation or other genetic alterations using conventional techniques (Capecchi, 1989; Valancius and Smithies, 1991; Hasty et al, 1991; Shinkai et al, 1992; Mombaerts et al, 1992; Philpott et al, 1992; Snouwaert et al, 1992; Donehower et al, 1992) to produce knockout or transplacement animals. A transplacement is similar to a knockout because the endogenous gene is replaced, but in the case of a transplacement the replacement is by another version of the same gene. After test substances have been administered to the animals, the phenotype must be assessed. If the test substance prevents or suppresses the disease, then the test substance is a candidate therapeutic agent for the treatment of disease. These animal models provide an extremely important testing vehicle for potential therapeutic products.
In one embodiment of the invention, transgenic animals are produced which contain a functional transgene encoding a functional 07CG27 polypeptide or variants thereof. Transgenic animals expressing 07CG27 transgenes, recombinant cell lines derived from such animals and transgenic embryos may be useful in methods for screening for and identifying agents that induce or repress function of 07CG27. Transgenic animals of the present invention also can be used as models for studying indications such as disease.
In one embodiment of the invention, a 07CG27 transgene is introduced into a non-human host to produce a transgenic animal expressing a human or murine 07CG27 gene. The transgenic animal is produced by the integration of the transgene into the genome in a manner that permits the expression of the transgene. Methods for producing transgenic animals are generally described by Wagner and Hoppe (U.S. Patent No. 4,873,191; which is incorporated herein by reference), Brinster et al. 1985; which is incorporated herein by reference in its entirety) and in "Manipulating the Mouse Embryo; A Laboratory Manual" 2nd edition (eds., Hogan, Beddington, Costantimi and Long, Cold Spring Harbor Laboratory Press, 1994; which is incorporated herein by reference in its entirety).
It may be desirable to replace the endogenous 07CG27 by homologous recombination between the transgene and the endogenous gene; or the endogenous gene may be eliminated by deletion as in the preparation of "knock-out" animals. Typically, a 07CG27 gene flanked by genomic sequences is transferred by microinjection into a fertilized egg. The microinjected eggs are implanted into a host female, and the progeny are screened for the expression of the transgene. Transgenic animals may be produced from the fertilized eggs from a number of animals including, but not limited to reptiles, amphibians, birds, mammals, and fish. Within a particularly preferred embodiment, transgenic mice are generated which overexpress 07CG27 or express a mutant form of the polypeptide. Alternatively, the absence of a 07CG27 in "knockout" mice permits the study of the effects that loss of 07CG27 protein has on a cell in vivo. Knock-out mice also provide a model for the development of 07CG27-related cancers.
Methods for producing knockout animals are generally described by Shastry (1995, 1998) and Osterrieder and Wolf (1998). The production of conditional knockout animals, in which the gene is active until knocked out at the desired time is generally described by Feil et al. (1996), Gagneten et al. (1997) and Lobe and Nagy (1998). Each of these references is incorporated herein by reference.
As noted above, transgenic animals and cell lines derived from such animals may find use in certain testing experiments. In this regard, transgenic animals and cell lines capable of expressing wild-type or mutant 07CG27 may be exposed to test substances. These test substances can be screened for the ability to reduce overepression of wild-type 07CG27 or impair the expression or function of mutant 07CG27. Pharmaceutical Compositions and Routes of Administration The 07CG27 polypeptides, antibodies, peptides and nucleic acids of the present invention can be formulated in pharmaceutical compositions, which are prepared according to conventional pharmaceutical compounding techniques. See, for example, Remington's Pharmaceutical Sciences. 18th Ed. (1990, Mack Publishing Co., Easton, PA). The composition may contain the active agent or pharmaceutically acceptable salts of the active agent. These compositions may comprise, in addition to one of the active substances, a pharmaceutically acceptable excipient, carrier, buffer, stabilizer or other materials well known in the art. Such materials should be non- toxic and should not interfere with the efficacy of the active ingredient. The carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., intravenous, oral, intrathecal, epineural or parenteral.
For oral administration, the compounds can be formulated into solid or liquid preparations such as capsules, pills, tablets, lozenges, melts, powders, suspensions or emulsions. In preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, suspending agents, and the like in the case of oral liquid preparations (such as, for example, suspensions, elixirs and solutions); or carriers such as starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like in the case of oral solid preparations (such as, for example, powders, capsules and tablets). Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be sugar-coated or enteric-coated by standard techniques. The active agent can be encapsulated to make it stable to passage through the gastrointestinal tract while at the same time allowing for passage across the blood brain barrier. See for example, WO 96/11698. For parenteral administration, the compound may be dissolved in a pharmaceutical carrier and administered as either a solution or a suspension. Illustrative of suitable carriers are water, saline, dextrose solutions, fructose solutions, ethanol, or oils of animal, vegetative or synthetic origin. The carrier may also contain other ingredients, for example, preservatives, suspending agents, solubilizing agents, buffers and the like. When the compounds are being administered intrathecally, they may also be dissolved in cerebrospinal fluid.
The active agent is preferably administered in a therapeutically effective amount. The actual amount administered, and the rate and time-course of administration, will depend on the nature and severity of the condition being treated. Prescription of treatment, e.g. decisions on dosage, timing, etc., is within the responsibility of general practitioners or specialists, and typically takes account of the disorder to be treated, the condition of the individual patient, the site of delivery, the method of administration and other factors known to practitioners. Examples of techniques and protocols can be found in Remington 's Pharmaceutical Sciences.
Alternatively, targeting therapies may be used to deliver the active agent more specifically to certain types of cell, by the use of targeting systems such as antibodies or cell specific ligands. Targeting may be desirable for a variety of reasons, e.g. if the agent is unacceptably toxic, or if it would otherwise require too high a dosage, or if it would not otherwise be able to enter the target cells.
Instead of administering these agents directly, they could be produced in the target cell, e.g. in a viral vector such as described above or in a cell based delivery system such as described in U.S. Patent No. 5,550,050 and published PCT application Nos. WO 92/19195, WO 94/25503, WO 95/01203, WO 95/05452, WO 96/02286, WO 96/02646, WO 96/40871, WO 96/40959 and WO 97/12635, designed for implantation in a patient. The vector could be targeted to the specific cells to be treated, or it could contain regulatory elements which are more tissue specific to the target cells. The cell based delivery system is designed to be implanted in a patient's body at the desired target site and contains a coding sequence for the active agent. Alternatively, the agent could be administered in a precursor form for conversion to the active form by an activating agent produced in, or targeted to, the cells to be treated. See for example, EP 425,731 A and WO 90/07936.
EXAMPLES The present invention is described by reference to the following Examples, which are offered by way of illustration and are not intended , to limit the invention in any manner. Standard techniques well known in the art or the techniques specifically described below were utilized. EXAMPLE 1
Ascertain and Study Kindreds Likely to Have a Chromosome 1 -Linked Prostate Cancer Susceptibility Locus
Extensive cancer prone kindreds were ascertained from a defined population providing a large set of extended kindreds with multiple cases of prostate cancer and many relatives available to study for analysis of candidate genes in the HPCl region.
Each kindred was extended through all available connecting relatives, and to all informative first degree relatives of each proband or cancer case. For these kindreds, additional prostate cancer cases and individuals with cancer at other sites of interest (e.g., bladder) who also appeared in the kindreds were identified through the tumor registry linked files. All prostate cancers reported in the kindred which were not confirmed in the Utah Cancer Registry were verified. Medical records or death certificates were obtained for confirmation of all cancers. Each key connecting individual and all informative individuals were invited to participate by providing a blood sample from which DNA was extracted. We also sampled spouses, siblings, and offspring of deceased cases so that the genotype of the deceased cases could be inferred from the genotypes of their relatives.
Each of the Utah pedigrees studied represents the descendants of a single founder for whom a significant excess of prostate cancer cases was observed among all descendants. Since all affected descendants are studied, the resulting kindreds represent a collection of both closely and distantly related prostate cancer cases. The criteria for selection of kindreds to analyze for 07CG27 linkage were: 1) genotypes available, or inferable, for 6 or more prostate cancer cases, and 2) at least 3 genotyped cases within a second degree of relationship to another genotyped case.
The Utah kindreds are 5 - 7 generations deep, and contain between 8 and 29 prostate cancer cases. They are all Caucasian of Northern European ancestry. The median age-of-onset for each kindred ranged from 64 to 76, similar to that estimated for the general population. Five percent of cases were diagnosed before age 55. For each kindred analyzed, the number of prostate cancer cases, the median age and range of age-of-onset, and the number of cases and family members sampled and included in this analysis were detailed. EXAMPLE 2 Selection of Kindreds Which are Linked to Chromosome 1 Nuclear pellets were extracted from 16 ml of ACD blood, and DNA extracted with phenol and chloroform, precipitated with ethanol, and resuspended in Tris-EDTA. The markers used for genotyping were short tandem repeat (STR) loci at lq24-25 which flanked the most likely HPCl location as indicated in Smith et al. (1996). The most likely location as suggested in Smith et al. (1996) is at D1S254.
Amplification of 20 ng genomic DNA was performed according to standard PCR procedures, with minor modifications to optimize product clarity, in a total reaction mix of 10 ml. Radiolabeled PCR products were electrophoresed on standard 6% polyacrylamide denaturing sequencing gels. Gels were then dried and autoradiographed. A total of over 200 prostate cancer cases and approximately 800 of their relatives were genotyped for the markers.
In the kindreds which showed evidence of segregation, up to an additional 35 markers were used to identify and confirm segregation of multiple linked markers (haplotypes). Two- point linkage analysis was performed with the package LINKAGE (Lathrop et al., 1984; 1985) using the FASTLINK implementation (Cottingham et al., 1993; Schaffer et al., 1994). The statistical analysis for the inheritance of susceptibility to prostate cancer used the model described in Smith et al. (1996). This model assumed a rare autosomal dominant susceptibility locus and allowed for a 15% sporadic rate of prostate cancer. Marker allele frequencies were estimated from unrelated individuals present in the kindreds.
Linkage in the presence of heterogeneity was assessed by the admixture test (A-test) of
Ott (1986). HOMOG, which postulates two family types, linked and unlinked, was used.
Multipoint linkage analysis was performed using VITESSE (O'Connell et al., 1995). The size of the pedigrees and the lack of genotyping of the higher generations due to the late age-of-onset, made more-than-three-point analyses impossible.
The two-point Lod scores for the 29 kindreds combined were highly negative at the 3 markers examined, suggesting an overall lack of evidence for this susceptibility locus across all kindreds. Heterogeneity analysis of the three loci showed weak, non-significant evidence for one locus, explaining 5% of the pedigrees. The positive Lod score observed for D1S254 in analysis of heterogeneity, as well as the low estimate of alpha reported in Smith et al. (1996) suggested that there might be a subset of linked pedigrees within our data set. We examined three marker haplotypes in each kindred for evidence of a shared region among affecteds. For those kindreds which suggested such segregation, we genotyped samples for up to an additional 35 markers
Multipoint linkage analysis resulted in a maximum heterogeneity Lod score of +1.20 at D1S254 with an estimate that 5% of kindreds were linked. Multipoint heterogeneity analysis in the most likely interval excluded linkage (Lod scores less than -2.00) for alpha greater than 0.33.
Cancers of sites other than prostate would also be expected to occur in individuals in these kindreds. Some individuals hypothesized to be sharing the segregating chromosome 1 haplotype were affected with cancer at another site. Lod scores for linkage for a phenotype of cancer of any site did not differ significantly from those for prostate alone, although most individuals with cancer of another site were not included in the sampling.
EXAMPLE 3 Contig Assembly Genomic clone contig assembly in the HPCl region started from a publicly available integrated map of chromosome 1, the WICGR Chr 1 map of Nov. 19, 1996. YACs located in the interval between D1S202 and D1S238 were ordered from Genome Systems. Primer pairs for the markers located in the interval between D1S202 and D1S238 were synthesized and used to screen a BAC library at Myriad. Markers that were negative on that BAC library were used to screen the BAC and PAC libraries at Genome Systems. DNA preps were prepared from the BACs and PACs that contained these markers. End sequences were obtained by dye terminator sequencing with vector primers on ABI 377 sequencers. Primer pairs defining BAC or PAC end markers were designed from these sequences. These new markers were checked against the YACs to make sure that they mapped within the interval. If the map data were ambiguous, the markers were also checked against a radiation hybrid panel. These new markers were checked against the already identified BACs/PACs to determine the positions of these clones relative to each other. The outside markers from each clone contig were used to screen the Myriad BAC library; those that were negative on that BAC library were used to screen the BAC and PAC libraries at Genome Systems. Repeated cycles of library screening and marker development allowed us to build a BAC/PAC contig that spanned the minimal recombinant interval. EXAMPLE 4 Genomic sequencing Two different types of genomic sequencing sublibraries were prepared from BAC or PAC clones in the candidate region. Random-Sheared Sequencing Sub-libraries
BAC or PAC DNA was sheared by sonication. To generate blunt-ended fragments, the sonicated DNA was incubated with mung-bean nuclease (Pharmacia Biotech) followed by treatment with a Pfu polishing kit (Stratagene). The DNA fragments were size fractionated on a 0.8% TAE agarose gel, and fragments in the size range of 1.0 - 1.6 kb were excised under longwave (365 nm) ultraviolet light. The excised gel slice was rotated 180 degrees relative to the original direction of electrophoresis and then placed into a new gel tray containing 1.0% GTG-Seaplaque low-melting temperature agarose (FMC corporation) before the gel solidified. Electrophoresis was repeated for the same time and voltage as the first run, resulting in a concentration of the DNA fragments in a small volume of agarose, and the gel slice containing the DNA fragments was once again excised from the gel. The DNA fragments were purified from the agarose by incubating the gel slice with beta-agarose (New England Biolabs), followed by removal of the agarose monomers using disposable microconcentrators (Amicon) that employ a 50,000 Daltons molecular weight cutoff filter. DNA fragments were ligated into the Hinc II site of the plasmid pMYG2, a pBluescript (Stratagene) derivative where the polylinker has been replaced by a different polylinker MYG2. The vector was prepared by digestion with Hinc II followed by dephosphorylation with calf alkaline phosphatase (Boehringer Mannheim).
Ligated products were transformed into DH5α E. coli competent cells (Life Technologies, Inc.) and plated on LB plates containing ampicillin, IPTG, and Bluo-gal (Sigma; Life Technologies, Inc.) . White colonies were used to inoculate individual wells of 1 ml 96-well microtiter plates (Beckman) containing 200 microhters of LB media supplemented with ampicillin at 50 micrograms per milliliter. The plates were incubated for 16-20 hours in a shaking incubator at 37 degrees Celsius. After incubation, 20 microhters of dimethyl sulfoxide was added to each well and the plates stored frozen. The inserts of random-sheared clones were amplified from E. coli cultures by PCR with vector primers, and the PCR products were sequenced with Ml 3 forward or reverse fluorescent energy transfer (FET) dye-labeled primers on ABI 377 sequencers. Sau 3A Sequencing Sub-libraries BAC or PAC DNA was partially digested with the restriction enzyme Sau 3 A, and fragments in the size range of 5-8 kb were size fractionated and recovered from the agarose gel as described above for random-sheared fragments. Sau 3 A fragments were ligated into the Bam HI site of pMYGl, a pBluescript (Stratagene) derivative where the polylinker has been replaced by a different polylinker MYG1. The vector was prepared by digestion with Bam HI and dephosphorylation with shrimp alkaline phosphatase (Amersham). The ligated products were transformed and plated as described above for random-sheared clones.
To identify clones containing inserts in the size range of 5-8 kb, bacterial colonies were screened using a plasmid preparation procedure that has been adapted for use in a 96-well format. White colonies were picked into individual wells of 2 ml 96-well plates (Continental Laboratory Products) containing 1 ml LB media supplemented with 200 micrograms per milliliter ampicillin. The plates were incubated 16-20 hours in a shaking incubator at 37 degrees Celsius. A bacterial stock of these clones was prepared by transferring 100 microhters of the 1 ml cultures to another 96-well plate containing 200 microhters of LB media supplemented with ampicillin. The remaining cells were pelleted by centrifugation and the pellets resuspended in 200 microhters of LB media. One hundred microhters of the concentrated cells were transferred to a 96-well thermowell PCR plate (Costar), and the cells were once again pelleted. The pelleted cells were resuspended in lysis buffer [250 mM Tris-HCl, pH 8.0, 50 mM EDTA, pH 8.0, 8% sucrose, 5% Triton X-100, 1 mM tartrazine, and 666 micrograms per milliliter lysozyme], and the plates were covered with thermowell lids (Costar) and incubated in a MJ Research fhermocycler for 2 minutes at 100 degrees Celsius followed by 2 minutes at 25 degrees Celsius. Cell debris was pelleted by centrifugation, and 15 microhters of the supernatant containing the plasmid DNA was electrophoresed on a 0.6x TBE 0.8% agarose gel with appropriate supercoiled size standards to estimate the size of each clone.
The bacterial stocks of clones with inserts in the 5-8 kb size range were used to inoculate 3 ml cultures of LB media supplemented with ampicillin, which were incubated overnight in a shaking incubator at 37 degrees Celsius. Plasmid DNA was prepared from these cultures using the Autogen robotic plasmid preparation machine (Integrated Separation Systems). The resulting DNA templates are subjected to DNA sequencing from both ends with Ml 3 forward or reverse fluorescent energy transfer (FET) dye-labeled primers on ABI 377 sequencers. DNA sequencing gel files were examined for lane tracking accuracy and adjusted where necessary before data extraction. ABI sample files resulting from gel files were converted to the Standard Chromatogram Format (SCF) [Dear and Staden] and trimmed of sequencing vector (pMYGl or pMYG2). Trimmed sequences were assembled using Acem.bly (Thierry-Mieg et al, 1995; Durbin and Thierry-Mieg, 1991). Contiguous sequence resulting from automatic assembly was screened for residual vector sequence (both sequencing vector and cloning vector) as well as for bacterial contamination using BLAST (Altschul et al, 1990).
Remaining sequences were arranged according to the relative position and orientation of assembled Sau3AI partial digest clone sequence reads as well as sequence similarity to overlapping genomic clones. Repetitive sequence was masked from the sequence contigs using xblast (Claverie and States, 1993). These masked sequences were placed in a Genetic Data Environment (GDE) (Smith et al., 1994) local database for subsequent similarity searches. Similarities among genomic DNA sequences and hybrid-selected cDNA clones as well as GenBank entries—both DNA and protein—were identified using BLAST. DNA sequences were also characterized with respect to short period repeats, CpG content, and long open reading frames.
EXAMPLE 5 Hybrid selection Two distinct methods of hybrid selection were used in this work. Method 1 : cDNA preparation and selection. Poly (A) enriched RNA from human mammary gland, prostate, testis, fetal brain, and placenta tissues and from total RNA of the cell line Caco-2 (ATCC HTB 37) were reverse transcribed using the tailed random primer RXGN6 and M-MLV Reverse Transcriptase (Life Technologies, Inc.). First strand cDNA was poly(A) tailed, 2nd strand synthesis was primed with the oligo RXGT12 , and then the ds cDNA was expanded by amplification with the primer RXG. Hybrid selection was carried out for two consecutive rounds of hybridization to immobilized BAC, PAC or gel purified YAC DNA as described previously. [Parimoo et al, 1991; Rommens et al, 1994]. Individual gel purified YACs or groups of two to four overlapping BAC and/or PAC clones were used in individual selection experiments. Hybridizing cDNA was collected, passed over a G50 Fine Sephadex column and amplified using tailed primers. The products were then digested with EcoRI, size selected on agarose gels, and ligated into pBluescript (Stratagene) that had been digested with EcoRI and treated with calf alkaline phosphatase (Boehringer Mannheim). Ligation products were transformed into competent DH5α E. coli cells (Life Technologies, Inc.).
Characterization of Retrieved cDNAs. 200 to 300 individual colonies from each ligation (from each 250 kbases of genomic DNA) were picked and gridded into microtiter plates for ordering and storage. Cultures were replica transferred onto Hybond N membranes (Amersham) supported by LB agar with ampicillin. Colonies were allowed to propagate and were subsequently lysed with standard procedures. Initial analysis of the cDNA clones involved a prescreen for ribosomal sequences and subsequent cross screenings for detection of overlap and redundancy. Approximately 10-25% of the clones were eliminated as they hybridized strongly with radiolabeled cDNA obtained from total RNA. Plasmids from 25 to 50 clones from each selection experiment that did not hybridize in prescreening were isolated for further analysis. The retrieved cDNA fragments were verified to originate from individual starting genomic clones by hybridization to restriction digests of DNAs of the starting clones, of a hamster hybrid cell line that contains chromosome 1 as its only human material, and to human genomic DNA. The clones were tentatively assigned into groups based on the overlapping or non-overlapping intervals of the genomic clones.
Method 2: cDNA Preparation. Poly(A) enriched RNA from human mammary gland, fetal brain, lymphocyte, pancreas, prostate, stomach, and thymus were reverse-transcribed using the tailed random primer XN12 and Superscript II reverse transcriptase (Gibco BRL). After second strand synthesis and end polishing, the ds cDNA was purified on Sepharose CL-4B columns (Pharmacia). cDNAs were "anchored" by ligation of a double-stranded oligo RP (RP-2 annealed to RL-1) to their 5' ends (5' relative to mRNA) using T4 DNA ligase. Anchored ds cDNA was then repurified on Sepharose CL-4B columns. Selection was performed by a modified procedure of Lovett et al. (1991). cDNAs from mammary gland, fetal brain, lymphocyte, pancreas, prostate, stomach, and thymus tissues were first expanded by amplification using a nested version of RP, RP.A and XPCR, and purified by fractionation on Sepharose CL-4B. Selection probes were prepared from purified Pis, BACs or PACs by digestion with Hinfl and Exonuclease III. The single-stranded probe was photolabelled with photobiotin (Gibco BRL) according to the manufacturer's recommendations. Probe, cDNA and C0t-1 DNA and poly A DNA were hybridized in 2.4M TEA-C1, lOmM NaPO4, ImM EDTA. Hybridized cDNAs were captured on streptavidin-paramagnetic particles (Dynal), eluted, and reamplified with a further nested version of RP, RP.B and XPCR, and gel purified. The selected, amplified cDNA was hybridized with an additional aliquot of probe, C0t- 1 DNA and poly A DNA. Captured and eluted products were amplified again with RP.B and XPCR, size-selected by gel electrophoresis, and cloned into dephosphorylated Hindi cut pUC18. Ligation products were transformed into XL2-Blue ultra-competent cells (Stratagene).
Both methods: Insert-containing clones were identified by blue/white selection on Xgal or Bluo-gal plates. Inserts were amplified by colony PCR with vector primers and then sequenced on ABI 377 sequencers. Alignment of these cDNA sequences to corresponding genomic sequences, and parsing of the revealed exons across those genomic sequences, allowed initial characterization of genes located within the region.
EXAMPLE 6
Inter-exon PCR and RACE for the identification of new exons (5'. 3'. or internal) in the HPCl Region
Inter-exon PCR: Following sequence analysis of hybrid selected clones that originated from the HPCl region, several primers were designed to try to amplify HPCl region products from fetal brain, breast, pancreas, prostate, stomach, and thymus cDNAs. Amplification was by hot start PCR; conditions used were an initial denaturation step at 95°C for 30 sec followed by a pause at 80°C while the polymerase/ nucleotide mixture was added to the template/primer mixtures. The hot start was followed by 35 cycles of denaturation at 96°C (4 s), annealing at
60°C (10 s) and extension at 72°C (60 s). Parsing of these cDNA sequences across the genomic sequence of the HPCl region revealed several new exons of candidate genes within the HPCl region. 5' RACE: The 5' end exons of candidate genes within the HPCl were identified by a modified RACE protocol called biotin capture 5' RACE (Tavtigian et al., 1996). Poly(A) enriched RNA from prostate was reverse-transcribed using the tailed random primer XN12 and Superscript II reverse transcriptase (Gibco BRL). After second strand synthesis and end polishing, the ds cDNA was purified on Sepharose CL-4B columns (Pharmacia). cDNAs were "anchored" by ligation of a double-stranded oligo RP (RP-2 annealed to RL-1) to their 5' ends (5' relative to mRNA) using T4 DNA ligase. Anchored ds cDNA was then repurified on Sepharose CL-4B columns.
The 5' sequences of candidate genes within the HPCl region were amplified using differenct primer combinations, and PCR products were fractionated on an agarose gel, gel purified, and captured on streptavidin-paramagnetic particles (Dynal). Reamplifications were performed if necessary using nested primers. These PCR reactions gave several bands on an agarose gel; the PCR products were gel purified and sequenced in the reverse direction with dye terminator chemistry on an ABI 377 sequencer.
3' RACE: The 3' end exons of candidate genes within the HPCl region was identified by a modified RACE protocol called biotin capture 3' RACE. Poly(A) enriched RNA from prostate was reverse-transcribed using a tailed random primer and Superscript II reverse transcriptase (Life Technologies). The first strand (heteroduplex) cDNA was purified by fractionation on a Sepharose CL-6B column.
The 3' sequence of candidate genes within the HPCl region was amplified with a biotinylated forward primer and an the anchor primer. PCR products amplified with these primers were fractionated on an agarose gel, gel purified, and captured on streptavidin- paramagnetic particles (Dynal). Captured material was reamplified as necessary using a nested phosphorylated forward primer PF5 and a tailed random primer.
PCR products were gel purified, ligated into the vector pMYG2, and transformed into DH5α cells. Colony PCR products were sequenced using using dye terminator chemistry on an ABI 377 sequencer.
EXAMPLE 7 cDNA library screening Radioactive probes prepared from hybrid selected clones representative of candidate gene transcripts within the HPCl region were used as probes to screen a total of 5.5 x 106 recombinant phage from a human prostate λgtl l cDNA library (HL1131b, Clontech). Prehybridization and hybridization was performed at 42°C in 50% formamide, 5X SSPE, 0.1% SDS, 5X Denhardt's mixture, 0.2 mg/ml denatured salmon sperm DNA and 2 mg/ml poly (A). Dextran sulfate (4% v/v) was included in the hybridization solution only. The filters were rinsed in 2X SSC for 10 minutes at room temperature and then rinsed in 2X SSC/0.1% SDS for 30 minutes at 60°C followed by two washes in IX SSC/0.1% SDS for 20 minutes each at 60°C. The positive phage were retested for second and third screenings, as required, to obtain purified plaques for sequencing. Inserts were amplified by phage PCR with vector primers and then sequenced using dye terminator chemistry on ABI 377 sequencers.
EXAMPLE 8
Mutation screening
Both genomic DNA and cDNA were used as templates for mutation screening.
Genomic DNA: Using genomic DNAs from prostate kindred members, prostate cancer affecteds, and tumor cell lines as templates, nested PCR amplifications were performed to generate PCR products of the candidate genes in the HPCl region that were screened for mutations. One to 10 ng of genomic DNA were subjected to a 23-26 cycle primary amplification, after which the PCR products were diluted 60-fold and reamplified using nested
M13-tailed primers for another 20-25 cycles; either TaqPlus (Stratagene) or AmpliTaq Gold
(Perkin Elmer) was used in the PCRs. In general, the PCR conditions used were an initial denaturation step at 95°C for 1 min (TaqPlus) or 10 min (AmpliTaq Gold), followed by cycles of denaturation at 96°C (12 s), annealing at 55°C (15 s) and extension at 72°C (45-60 s). PCR products were sequenced with Ml 3 forward or reverse fluorescent energy transfer (FET) dye- labeled primers on ABI 377 sequencers. Chromatograms were analyzed for the presence of polymorphisms or sequence aberrations in either the Macintosh program Sequencer (Gene Codes) or the Java program Mutscreen (Myriad, proprietary). cDNA: Total RNA prepared from either tumor cell lines or prostate kindred lymphocytes was treated with DNase I (Boehringer Mannheim) to remove contaminating genomic DNA, and then reverse transcribed to heteroduplex cDNA with a mix of Nio random primers and a tailed oligo dT primer, and Superscript II reverse transcriptase (Life Technologies). This cDNA was used as the template for nested PCR amplifications to generate the cDNA PCR products of the candidate genes that were screened for HPCl mutations. Using the outer primer pair for each amplicon, 10 ng of cDNA were subjected to a 20 cycle primary amplification, after which the PCR products were diluted 100-fold and reamplified using nested M13-tailed primers for another 25-30 cycles. The cDNAs were amplified by hot start PCRs using TaqPlus DNA polymerase (Stratagene). Conditions used were an initial denaturation step at 95°C for 30 sec followed by a pause at 80°C while the polymerase/nucleotide mixture was added to the template/primer mixtures. The hot start was followed by cycles of denaturation at
96°C (4 s), annealing at 55°C (10 s) and extension at 72°C (60 s). PCR products were gel purified and then sequenced with Ml 3 forward or reverse fluorescent energy transfer (FET) dye- labeled primers on ABI 377 sequencers. The sequences of these products were analyzed in GDE to determine their exon structure. Chromatograms were analyzed for the presence of polymorphisms or sequence aberrations in either the Macintosh program Sequencher (Gene Codes) or the Java program Mutscreen (Myriad, proprietary).
EXAMPLE 9 Analysis of the 07CG27 Gene
The cDNA sequence of the 07CG27 gene is set forth in SEQ ID NO:l, with the corresponding protein sequence set forth in SEQ ID NO:2. The 07CG27 gene comprises 33 exons as shown in Table 1, with the base numbering relative to SEQ ID NO:l.
TABLE 1
Exons ofthe 07CG27
Starting Ending Starting Ending
Exon Base Base Exon Base Base
1 1 167 18 5197 5351
2 168 336 19 5352 6255
3 337 514 20 6256 6341
4 515 624 21 6342 6530
5 625 750 22 6531 6782
6 751 974 23 6783 6875
7 975 1057 24 6876 6977
8 1058 1191 25 6978 7179
9 1192 1279 26 7180 7383 10 1280 1424 27 7384 7496 11 1425 1478 28 7497 7633 12 1479 2097 29 7634 7865 13 2098 2267 30 7866 7973 14 2268 2517 31 7974 8192 15 2518 2728 32 8193 8296 16 2729 4987 33 8297 8423 17 4988 5196
The structure and function of 07CG27 gene are determined according to the following methods. Biological Studies. Mammalian expression vectors containing 07CG27 cDNA are constructed and transfected into appropriate prostate carcinoma cells with lesions in the gene. Wild-type 07CG27 cDNA as well as altered 07CG27 cDNA are utilized. The altered 07CG27 cDNA can be obtained from altered 07CG27 alleles or produced as described below. Phenotypic reversion in cultures (e.g., cell morphology, doubling time, anchorage-independent growth) and in animals (e.g., tumorigenicity) is examined. The studies will employ both wild- type and mutant forms of the gene.
Molecular Genetics Studies. In vitro mutagenesis is performed to construct deletion mutants and missense mutants (by single base-pair substitutions in individual codons and alanine scanning mutagenesis). The mutants are used in biological, biochemical and biophysical studies. Mechanism Studies. The ability of 07CG27 protein to bind to known and unknown DNA sequences is examined. Its ability to transactivate promoters is analyzed by transient reporter expression systems in mammalian cells. Conventional procedures such as particle- capture and yeast two-hybrid system are used to discover and identify any functional partners. The nature and functions of the partners are characterized. These partners in turn are targets for drug discovery.
Structural Studies. Recombinant proteins are produced in E. coli, yeast, insect and/or mammalian cells and are used in crystallographical and NMR studies. Molecular modeling of the proteins is also employed. These studies facilitate structure-driven drug design. As noted above, 07CG27 shares domain(s) in common with guansosine exchange factor
(GΕF) proteins. Preliminary studies have confirmed that 07CG27 is a ras activator.
EXAMPLE 10
Generation of Polyclonal Antibody against 07CG27 Segments of 07CG27 coding sequence are expressed as fusion protein in E. coli. The overexpressed proteins are purified by gel elution and used to immunize rabbits and mice using a procedure similar to the one described by Harlow and Lane, 1988. This procedure has been shown to generate Abs against various other proteins (for example, see Kraemer, et al, 1993).
Briefly, a stretch of 07CG27 coding sequence was cloned as a fusion protein in plasmid PET5A (Novagen, Inc., Madison, Wl). After induction with IPTG, the overexpression of a fusion protein with the expected molecular weight is verified by SDS/PAGE. Fusion proteins are purified from the gel by electroelution. The identification of the protein as the 07CG27 fusion product is verified by protein sequencing at the N-terminus. Next, the purified protein is used as immunogen in rabbits. Rabbits are immunized with 100 mg of the protein in complete Freund's adjuvant and boosted twice in 3 week intervals, first with 100 mg of immunogen in incomplete Freund's adjuvant followed by 100 mg of immunogen in PBS. Antibody containing serum is collected two weeks thereafter.
This procedure can be repeated to generate antibodies against mutant forms of the 07CG27 protein. These antibodies, in conjunction with antibodies to wild type 07CG27, are used to detect the presence and the relative level of the mutant forms in various tissues and biological fluids.
EXAMPLE 11 Generation of Monoclonal Antibodies Specific for 07CG27 Monoclonal antibodies are generated according to the following protocol. Mice are immunized with immunogen comprising intact 07CG27 or 07CG27 peptides (wild type or mutant) conjugated to keyhole limpet hemocyanin using glutaraldehyde or EDC as is well known.
The immunogen is mixed with an adjuvant. Each mouse receives four injections of 10 to
100 mg of immunogen and after the fourth injection blood samples are taken from the mice to determine if the serum contains antibody to the immunogen. Serum titer is determined by ELISA or RIA. Mice with sera indicating the presence of antibody to the immunogen are selected for hybridoma production.
Spleens are removed from immune mice and a single cell suspension is prepared (see
Harlow and Lane, 1988). Cell fusions are performed essentially as described by Kohler and
Milstein, 1975. Briefly, P3.65.3 myeloma cells (American Type Culture Collection, Rockville, MD) are fused with immune spleen cells using polyethylene glycol as described by Harlow and
Lane, 1988. Cells are plated at a density of 2x10 cells/well in 96 well tissue culture plates. Individual wells are examined for growth and the supernatants of wells with growth are tested for the presence of 07CG27 specific antibodies by ELISA or RIA using wild type or mutant 07CG27 target protein. Cells in positive wells are expanded and subcloned to establish and confirm monoclonality. Clones with the desired specificities are expanded and grown as ascites in mice or in a hollow fiber system to produce sufficient quantities of antibody for characterization and assay development.
EXAMPLE 12 Isolation of 07CG27 Binding Peptides
Peptides that bind to the 07CG27 product are isolated from both chemical and phage- displayed random peptide libraries as follows.
Fragments of the 07CG27 gene product are expressed as GST and His-tag fusion proteins in both E. coli and SF9 cells. The fusion protein is isolated using either a glutathione matrix (for GST fusions proteins) or nickel chelation matrix (for His-tag fusion proteins). This target fusion protein preparation is either screened directly as described below, or eluted with glutathione or imidizole. The target protein is immobilized to either a surface such as polystyrene; or a resin such as agarose; or solid supports using either direct absorption, covalent linkage reagents such as glutaraldehyde, or linkage agents such as biotin-avidin. Two types of random peptide libraries of varying lengths are generated: synthetic peptide libraries that may contain derivatized residues, for example by phosphorylation or myristylation, and phage-displayed peptide libraries which may be phosphorylated. These libraries are incubated with immobilized 07CG27 gene product in a variety of physiological buffers. Next, unbound peptides are removed by repeated washes, and bound peptides recovered by a variety of elution reagents such as low or high pH, strong denaturants, glutathione, or imidizole. Recovered synthetic peptide mixtures are sent to commercial services for peptide micro- sequencing to identify enriched residues. Recovered phage are amplified, rescreened, plaque purified, and then sequenced to determined the identity of the displayed peptides. Use of 07CG27 binding peptides Peptides identified from the above screens are synthesized in larger quantities as biotin conjugates by commercial services. These peptides are used in both solid and solution phase competition assays with 07CG27 and its interacting partners identified in yeast 2-hybrid screens. Versions of these peptides that are fused to membrane-permeable motifs (Lin et al., 1995; Rojas et al., 1996) will be chemically synthesized, added to cultured cells and the effects on growth, apoptosis, differentiation, cofactor response, and internal changes will be assayed. EXAMPLE 13 Sandwich Assay for 07CG27 Monoclonal antibody is attached to a solid surface such as a plate, tube, bead, or particle. Preferably, the antibody is attached to the well surface of a 96-well ELISA plate. 100 ml sample (e.g., serum, urine, tissue cytosol) containing the 07CG27 peptide/protein (wild-type or mutant) is added to the solid phase antibody. The sample is incubated for 2 hrs at room temperature. Next the sample fluid is decanted, and the solid phase is washed with buffer to remove unbound material. 100 ml of a second monoclonal antibody (to a different determinant on the 07CG27 peptide/protein) is added to the solid phase. This antibody is labeled with a detector molecule (e.g., 125-1, enzyme, fluorophore, or a chromophore) and the solid phase with the second antibody is incubated for two hrs at room temperature. The second antibody is decanted and the solid phase is washed with buffer to remove unbound material.
The amount of bound label, which is proportional to the amount of 07CG27 peptide/protein present in the sample, is quantitated. Separate assays are performed using monoclonal antibodies which are specific for the wild-type 07CG27 as well as monoclonal antibodies specific for each of the mutations identified in 07CG27.
EXAMPLE 14
Two-hybrid Assay to Identify Proteins that Interact with 07CG27
Sequence encoding all or portions of 07CG27 are ligated to pAS2-l (Clontech) such that the coding sequence of 07CG27 is in- frame with coding sequence for the GAL4p DNA-binding domain. This plasmid construct is introduced into the yeast reporter strain Y190 by transformation. A library of activation domain fusion plasmids prepared from human prostate cDNA (Clontech) is then introduced into strain Y190 carrying the pAS2-l -based fusion construct. Transformants are spread onto 20 - 150 mm plates of yeast minimal media lacking leucine, tryptophan, and histidine, and containing 25 mM 3-amino-l,2,4-triazole. After one week incubation at 30°C, yeast colonies are assayed for expression of the lacZ reporter gene by β-galactosidase filter assay. Colonies that both grow in the absence of histidine and are positive for production of β-galactosidase are chosen for further characterization.
The activation domain plasmid is purified from positive colonies by the smash-and-grab technique. These plasmids are introduced into E. coli DH5α by electroporation and purified from E. coli by the alkaline lysis method. To test for the specificity of the interaction, specific activation domain plasmids are cotransformed into strain Y190 with plasmids encoding various DNA-binding domain fusion proteins, including fusions to 07CG27 and human lamin C. Transformants from these experiments are assayed for expression of the HIS3 and lacZ reporter genes. Positives that express reporter genes with 07CG27 constructs and not with lamin C constructs encode bona fide 07CG27 interacting proteins. These proteins are identified and characterized by sequence analysis of the insert of the appropriate activation domain plasmid.
This procedure is repeated with mutant forms of the 07CG27 gene, to identify proteins that interact with only the mutant protein or to determine whether a mutant form of the 07CG27 protein can or cannot interact with a protein known to interact with wild-type 07CG27.
While the invention has been disclosed in this patent application by reference to the details of preferred embodiments of the invention, it is to be understood that the disclosure is intended in an illustrative rather than in a limiting sense, as it is contemplated that modifications will readily occur to those skilled in the art, within the spirit of the invention and the scope of the appended claims.
LIST OF REFERENCES IN SPECIFICATION Altschul, S.F., et al. (1990). J. Mol Biol 215: 195-197. Altschul, S.F., et al. (1997). Nucl Acids Res. 25:3389-3402.
Anand, R. (1992). Techniques for the Analysis of Complex Genomes, (Academic Press). Anderson, J.A., et al. (1992). J. Otolaryngology 21:321.
Ausubel, F.M., et al. (1992). Current Protocols in Molecular Biology, (J. Wiley and Sons,
NY). Bartel, P.L., et al. (1993). "Using the 2-hybrid system to detect protein-protein interactions." In: Cellular Interactions in Development: A Practical Approach. Oxford University Press, pp. 153-179.
Beaucage and Carruthers (1981). Tetra. Letts. 22:1859-1862.
Botstein, et al. (1980). Am. J. Hum. Genet. 32:314-331. Brinster et al. (1985). Proc. Natl Acad. Sci. USA 82:4438-4442.
Cannon, L., et al. (1982). Cancer Surveys 1:47-69.
Capecchi, M.R. (1989). Science 244:1288.
Cariello (1988). Human Genetics 42:726. Chevray, P.M. and Nathans, D.N. (1992). Proc. Natl Acad. Sci. USA 89:5789-5793.
Claverie, J.M. and States, D.J. (1993). Computers and Chemistry 17: 191-201.
Conner, B.J., et al. (1983). Proc. Natl. Acad. Sci. USA 80:278-282.
Cotton, et al. (1988). Proc. Natl. Acad. Sci. USA 85:4397-4401. Cottingham, R..W., et al. (1993). Am. J. Hum. Genet. 53:252-263.
Dear, S. and Staden, R. (1992). DNA Sequence 107-110.
Deutscher, M. (1990). Meth. Enzymology 182 (Academic Press, San Diego, Cal.).
Donehower, L.A., et al. (1992). Nature 356:215.
Durbin, R. and Thierry-Mieg, J. (1991). A C. elegans Database. Documentation, code and data available from anonymous FTP servers at lirmm.lirmm.fr, cele.mrc-lmb.cam.ac.uk and ncbi.nlm.nih.gov.
Enhancers and Eurkaryotic Gene Expression, Cold Spring Harbor Press, Cold Spring Harbor, New York (1983).
Erickson, J. et al, (1990). Science 249:527-533. Fahy, E., et al. (1991). PCR Methods Appl. 1:25-33.
Fain, P.R. (1992). Cytogen. Cell Genet. 60:178.
Feil et al., (1996). Proc. Natl. Acad. Sci. USA 93:10887-10890.
Fields, S. and Song, O-K. (1989). Nature 340:245-246.
Fiers, et al. (1978). Nature 273:113. Fincham, S.M., et al., Wijayasinghe, C. (1990). The Prostate 17:189-206.
Finkelstein, J., et al. (1990). Genomics 7:167-172.
Fodor, S.P.A. (1997). DNA Sequencing. Massively Parallel Genomics. Science 277:393-395.
Gagneten et al. (1997). Nucl Acids Res. 25:3326-3331.
Glover, D. (1985). DNA Cloning, I and II (Oxford Press). Go, R.C.P., et al. (1983). J. Natl Cancer Inst. 71:455-461.
Goding (1986). Monoclonal Antibodies: Principles and Practice, 2d ed. (Academic Press, NY).
Godowski, et al. (1988). Science 241:812-816.
Goldgar, D.E., et al. (1994). J. Natl. Can. Inst. 86:3:200-209.
Goldgar, D.E., et al (1993). Am. J. Hum. Genet. 52:743-748. Grompe, M., (1993). Nature Genetics 5:111-117.
Grompe, M., et al, (1989). Proc. Natl. Acad. Sci. USA 86:5855-5892.
Guthrie, G. and Fink, G.R. (1991). Guide to Yeast Genetics and Molecular Biology (Academic Press).
Harlow and Lane (1988). Antibodies: A Laboratory Manual (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY Hasty, P., K., et al. (1991). Nature 350:243.
Hodgson, J. (1991). Bio/Technology 9:19-21.
Hogan et al. (eds) (1994). Manipulating the Mouse Embryo: A Laboratory Manual, 2nd Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY. Huse, et /. (1989). Science 246:1275-1281.
Innis, et al. (1990). PCR Protocols: A Guide to Methods and Applications (Academic Press, San Diego, CA).
Jablonski, E., et al. (1986). Nuc. Acids Res. 14:6115-6128.
Jakoby, W.B. and Pastan, I.H. (eds.) (1979). Cell Culture. Methods in Enzymology, Vol. 58 (Academic Press, Inc., Harcourt Brace Jovanovich (NY).
Jeffreys, et al. (1985). Nature 314:67-73.
Johnson, et al. (1993). "Peptide Turn Mimetics" In: Biotechnology and Pharmacy, Pezzuto et al., eds., Chapman and Hall, NY.
Kamb, A. et al. (1994). Science 264:436-440. Kandpal, et al. (1990). Nucl Acids Res. 18:1789-1795.
Kanehisa (1984). Nucl. Acids Res. 12:203-213.
Kinszler, K.W., et al (1991). Science 251:1366-1370.
Kinzler, K.W. and Vogelstein, B. 1997. Nature 386: 761-763.
Knudson, A.G. (1993). Nature Genet. 5:103. Krain, L.S. (1974). Preventive Medicine 3: 154-159.
Kubo, T., et al. (1988). FEBS Letts. 241:119.
Kyte, J. and Doolittle, R.F. (1982). J. Mol. Biol. 157:105-132.
Landegren, et al. (1988). Science 242:229.
Lathrop, G.M., et al. (1985). Am JHum Genet. 37:482-489. Lee, J.E., et al. (1995). Science 268:836-844.
Lin, Y. Z., et al. (1995). J. Biol. Chem. 270:14255-14258.
Litt, et al. (1989). Am. J. Hum. Genet. 44:397-401.
Lobe and Nagy (1998). Bioessays 20:200-208.
Maniatis. T., et al. (1982). Molecular Cloning: A Laboratory Manual (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY).
Martin, R., et al. (1990). BioTechniques 9:762-768.
Matteucci, M.D. and Caruthers, M.H. (1981). J. Am. Chem. Soc. 103:3185.
Matthews and Kricka (1988). Anal. Biochem. 169:1.
Meikle, A.W., et al. (1985). Prostate 6:121. Merrifield (1963). J. Am. Chem. Soc. 85:2149-2156. Metzger, et al. (1988). Nature 334:31-36.
Mittlin (1989). Clinical Chem. 35:1819.
Modrich, P. (1991). Ann. Rev. Genet. 25:229-253.
Mombaerts, P., et al. (1992). Cell 68:869. Morganti, G., et al. (1956). Acta Geneticae Medicae et Gemellogogiae 6:304-305.
Nakamura, et al. (1987). Science 235:1616-1622.
Neuhausen, S.L. et al. (1997). Br. J. Urol 19 Suppl. 1:15-20.
Newton, C.R., et al. (1989). Nucl. Acids Res. 17:2503-2516.
Nguyen, Q., et al. (1992). BioTechniques 13:116-123. Novack, et al. (1986). Proc. Natl. Acad. Sci. USA 83:586.
O'Connell, J.R. and Weeks, D.E. (1995). Nature Genet. 11:402-408.
Orita, et al. (1989). Proc. Natl. Acad. Sci. USA 86:2776-2770.
Osterrieder and Wolf (1998). Rev. Sci. Tech. 17:351-364.
Parimoo, S., et al. (1991). Proc. Natl. Acad. Sci. USA 88:9623-9627. Philpott, K.L., et al. (1992). Science 256:1448.
Rano and Kidd (1989). Nucl. Acids Res. 17:8392.
Rigby, P.W.J., et al. (1977). J. Mol Biol. 113:237-251.
Rojas, M., Yao, S., and Lin, Y. Z. (1996). J. Biol. Chem. 271:27456-27461.
Rommens, J.M., et al. (1994). In: "Identification of transcribed sequences", (U. Hochgeschwender and K. Gardiner, Eds.), pp. 65-79. Plenum Press, NY.
Sambrook, J., et al. (1989). Molecular Cloning: A Laboratory Manual, 2nd Ed. (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY).
Schaffer, A.A., et al. (1994). Human Heredity 44:225-237.
Scharf(1986). Science 233:1076. Scopes, R. (1982). Protein Purification: Principles and Practice, (Springer- Verlag, NY).
Shastry et al. (1995). Experientia 51:1028-1039.
Shastry et al. (1998). Mol. Cell. Biochem. 181:163-179.
Sheffield, V.C., et al. (1989). Proc. Natl. Acad. Sci. USA 86:232-236.
Sheffield, V.C., et al. (1991). Am. J. Hum. Genet. 49:699-706. Shenk, et al. (1975). Proc. Natl. Acad. Sci. USA 72:989.
Shinkai, Y., et al. (1992). Cell 68:855.
Skolnick, M.H. and Wallace, B.R. (1988). Genomics 2:273-279.
Smith .R., et al. (1996). Science 274:1371.
Smith, S.A., et al. (1992). Nature Genetics 2:128-131. Smith, S.W., et al. (1994). CABIOS 10:671-675.
Smith, T.F. and Waterman, M.S. (1981). J. Mol. Biol. 147:195-197.
Snouwaert, J.N., et al. (1992). Science 257:1083.
Spargo, C.A., et al. (1996). Mol. Cell. Probes 10:247-256. Tavtigian, S., et al. (1996). Nature Genetics 12:333-337.
Thierry-Mieg, D., et al. (1995). Ace.mbly. A graphic interactive program to support shotgun and directed sequencing projects.
Valancius, V. and Smithies, O. (1991). Mol. Cell Biol. 11:1402.
Wartell, R.M., et al. (1990). Nucl. Acids Res. 18:2699-2705. Weber and May (1989). Am. J. Hum. Genet. 44:388-396.
Wells, J.A. (1991). Methods in Enzymol 202:390-411.
Wetmur and Davidson (1968). J. Mol. Biol. 31:349-370.
White, M.B., et al, (1992). Genomics 12:301-306.
White and Lalouel (1988). Ann. Rev. Genet. 22:259-279. Woolf, CM. (1960). Cancer 13:739-744.
Wu, et al. (1989a). Genomics 4:560-569.
List of Patents and Patent Applications:
U.S. Patent No. 3,817,837.
U.S. Patent No. 3,850,752. U.S. Patent No. 3,939,350.
U.S. Patent No. 3,996,345.
U.S. Patent No. 4,275,149.
U.S. Patent No. 4,277,437.
U.S. Patent No. 4,366,241. U.S. Patent No. 4,376,110.
U.S. Patent No. 4,486,530.
U.S. Patent No. 4,554,101.
U.S. Patent No. 4,683,195.
U.S. Patent No. 4,683,202. U.S. Patent No. 4,816,567.
U.S. Patent No. 4,868,105.
U.S. Patent No. 4,873,191.
U.S. Patent No. 5,550,050.
U.S. Patent No.5,742,282. U.S. Patent No. 5,885,970.
U.S. Patent No. 5,891,858.
Hitzeman et al, EP 73,675 A
EPO Publication No. 225,807 EP 425 31A.
European Patent Application Publication No. 0332435
Geysen, H., PCT published application WO 84/03564, published 13 September 1984
WO 90/07936.
WO 92/19195. WO 94/25503.
WO 95/01203.
WO 95/05452.
WO 96/02286.
WO 96/02646. WO 96/11698.
WO 96/40871.
WO 96/40959.
WO 97/12635.
PCT published application WO 93/07282

Claims

WHAT IS CLAIMED IS:
1. An isolated nucleic acid coding for an 07CG27 polypeptide, said polypeptide having the amino acid sequence set forth in SEQ ID NO:2 or at least 95% identity to said amino acid sequence.
2. The isolated nucleic acid of claim 1 wherein said DNA has the nucleotide sequence (a) set forth in SEQ ID NO:l, (b) its complement, (c) a corresponding RNA, (d) a nucleotide sequence which hybridizes under stringent conditions to the nucleotide sequence of (a), (b) or (c), or (e) a nucleotide sequence which has at least 95% identity with the nucleotide sequence of (a), (b) or (c).
3. The isolated nucleic acid of claim 1 which is a DNA comprising an allelic variant of the nucleotide sequence set forth in SEQ ID NO:l, its complement or a corresponding RNA.
4. The isolated nucleic acid of claim 1 coding for a mutated form of the 07CG27 polypeptide set forth in SEQ ID NO:2.
5. The isolated nucleic acid of claim 4 which is a DNA comprising a mutated form of the nucleotide sequence set forth in SEQ ID NO:l, its complement or a corresponding RNA.
6. The isolated nucleic acid of claim 5 wherein the mutation is selected from the group consisting of a deletion mutation, a nonsense mutation, an insertion mutation, a frameshift mutation and a missense mutation.
7. An isolated nucleic acid having a nucleotide sequence selected from the group consisting of (a) exons 1-33, (b) complements thereof, (c) RNAs corresponding thereto and (d) nucleic acids which hybridize under stringent conditions to the nucleotide sequences of (a), (b) or (c).
8. An isolated nucleic acid having at least 15 contiguous nucleotides of a nucleic acid as claimed in any one of claims 1 to 7 wherein the nucleic acid sequence suitable for use as a hybridization probe to detect in a sample (i) a DNA having a nucleotide sequence selected from the nucleotide sequence set forth in SEQ ID NO:l, allelic variants thereof and mutated forms thereof, (ii) an RNA corresponding to said DNA or (iii) a nucleotide sequence which hybridizes under stringent conditions to the nucleotide sequence of (i) or (ii).
9. A set of nucleic acid probes for use in a microchip assay wherein each of said nucleic acid probes comprises at least 8 contiguous nucleotides of a nucleic acid as claimed in any one of claims 1 to 7 and said set encompasses part or all of said nucleic acid.
10. A vector which comprises an isolated nucleic acid as claimed in any one of claims 1 to 9.
11. An expression vector which comprises an isolated nucleic acid of any one of claims 1 to 9 wherein the coding sequence for the 07CG27 polypeptide or a peptide having at least 95% identity to said polypeptide is operably linked to suitable control sequences capable of directing expression of said coding sequence in host cells for said vector.
12. Host cells transformed with a vector as claimed in claim 10 or 11.
13. A method of producing a polypeptide which is the 07CG27 polypeptide having the amino acid sequence set forth in SEQ ID NO: 2 or a peptide having at least 95% identity to said polypeptide as defined in claim 1 which comprises (i) culturing the host cells of claim 12 containing an expression vector encoding said polypeptide under conditions suitable for the production of said 07CG27 polypeptide and (ii) recovering said polypeptide.
14. A method as claimed in claim 13 which further comprises labeling the recovered polypeptide.
15. A preparation of human 07CG27 polypeptide substantially free of other human proteins, said polypeptide having the amino acid sequence set forth in SEQ ID NO:2.
16. A preparation of human 07CG27 polypeptide substantially free of other human proteins, the amino acid sequence of said polypeptide having substantial sequence homology with the wild-type 07CG27 polypeptide having the amino acid sequence set forth in SEQ ID NO:2.
17. A preparation of a polypeptide substantially free of other proteins, said polypeptide being a mutated human 07CG27 polypeptide obtainable by expression of a mutated form of the nucleic acid sequence set forth in SEQ ID NO:2.
18. A preparation as claimed in any one of claims 15 to 17 wherein said polypeptide is labeled.
19. An antibody capable of specifically binding one or more polypeptides as claimed in any one of claims 15 to 17.
20. An antigenic fragment of a polypeptide as defined in any one of claims 15 to 17 suitable for use as an immunogen to obtain an antibody as claimed in claim 19.
21. A polypeptide as defined in any one of claims 15 to 17 and 20 in the form of a fusion protein.
22. Use of a polypeptide as defined in any one of claims 15 to 17, 20 and 21 as an immunogen for antibody production.
23. A use as claimed in claim 22 wherein one or more antibodies products are subsequently labeled or bound to a solid support.
24. A pair of single-stranded oligonucleotide primers for determination of a nucleotide sequence of a 07CG27 gene by a nucleic acid amplification reaction, the sequence of said primers being derived from genomic clones for 07CG27, and the use of said primers in a nucleic acid amplification reaction resulting in the synthesis of DNA or RNA corresponding to all or part of the sequence of the 07CG27 gene.
25. A pair of primers as claimed in claim 24 for determination of all or part of the sequence of the 07CG27 gene having the nucleotide sequence set forth in SEQ ID NO:l, allelic variant or a mutated form thereof.
26. A method for identifying a mutant 07CG27 nucleotide sequence in a suspected mutant 07CG27 allele which comprises comparing the nucleotide sequence of the suspected mutant 07CG27 allele with a wild-type 07CG27 nucleotide sequence, wherein a difference between the suspected mutant and the wild-type sequence identifies a mutant 07CG27 nucleotide sequence.
27. A method for detecting an alteration in 07CG27 wherein said alteration is associated with disease in a human which comprises analyzing an 07CG27 gene or an 07CG27 gene expression product from a tissue of said human.
28. A method as claimed in claim 27 wherein the sequence of the 07CG27 gene in said sample is compared with the sequence of one or more wild-type 07CG27 gene sequences selected from the sequence set forth in SEQ ID NO:l and wild-type allelic variants thereof.
29. The method of claim 27 wherein said expression product is selected from the group consisting of mRNA of the 07CG27 gene and a 07CG27 polypeptide encoded by the 07CG27 gene.
30. The method of any one of claims 27 to 29 wherein one or more of the following procedures is carried out:
(a) observing shifts in elecfrophoretic mobility of single-stranded DNA from said sample on non-denaturing polyacrylamide gels;
(b) hybridizing a 07CG27 gene probe to genomic DNA isolated from said sample under conditions suitable for hybridization of said probe to said gene; (c) determining hybridization of an allele-specific probe to genomic DNA from said sample; (d) amplifying all or part of the 07CG27 gene from said sample to produce an amplified sequence and sequencing the amplified sequence;
(e) determining by nucleic acid amplification the presence of a specific 07CG27 mutant allele in said sample; (f) molecularly cloning all or part of the 07CG27 gene from said sample to produce a cloned sequence and sequencing the cloned sequence;
(g) determining whether there is a mismatch between molecules (1) 07CG27 gene genomic DNA or 07CG27 mRNA isolated from said sample, and (2) a nucleic acid probe complementary to the human wild-type 07CG27 gene DNA, when molecules (1) and (2) are hybridized to each other to form a duplex;
(h) amplification of 07CG27 gene sequences in said sample and hybridization of the amplified sequences to nucleic acid probes which comprise wild-type 07CG27 gene sequences;
(i) amplification of 07CG27 gene sequences in said tissue and hybridization of the amplified sequences to nucleic acid probes which comprise mutant 07CG27 gene sequences;
(j) screening for a deletion mutation;
(k) screening for a point mutation;
(1) screening for an insertion mutation; (m) determining in situ hybridization of the 07CG27 gene in said sample with one or more nucleic acid probes which comprise the 07CG27 gene sequence or a mutant 07CG27 gene sequence;
(n) immunoblotting;
(o) immunocytochemistry; (p) assaying for binding interactions between 07CG27 protein isolated from said tissue and a binding partner capable of specifically binding the polypeptide expression product of a 07CG27 mutant allele and/or a binding partner for the 07CG27 polypeptide having the amino acid sequence set forth in SEQ ID NO:2; and
(q) assaying for the inhibition of biochemical activity of said binding partner.
31. A non-human animal which carries an altered 07CG27 allele in its genome.
32. A non-human animal in which its native 07CG27 alleles have been replaced by 07CG27 alleles from a second animal.
33. A non-human animal in which its native 07CG27 alleles have been disrupted or deleted.
34. A non-human animal in which one or both of its native 07CG27 alleles have been (a) modified to contain a DNA system to conditionally knockout said alleles or (b) replaced by 07CG27 alleles from a second animal modified to contain a DNA system to conditionally knockout said alleles.
35. A cell line isolated from the non-human animal of any one of claims 32 to 35.
36. An isolated mutant 07CG27 which is capable of activating ras.
37. Use of the nucleic acid of claim 1 or 7 or fragments thereof as a marker for prostate cancer.
38. A method of screening for drug candidates useful in treating a disease resulting from a mutation in 07CG27, wherein said method involves mixing a mutant 07CG27 with ras in both the presence of a drug and the absence of said drug and measuring the amount of activation of ras, wherein if the amount of said activation is less in the presence of said drug than in the absence of said drug then said drug is a drug candidate for treating said disease.
39. The method of claim 38 wherein said mutant 07CG27 is a fusion protein.
40. A method of screening for drug candidates useful in treating a disease resulting from a mutation in 07CG27, wherein said method comprises treating an animal which is homozygous for 07CG27 containing said mutation with a drug wherein if said animal does not develop said disease, said drug is a drug candidate for treating said disease.
41. A method of screening for drug candidates useful in treating a disease resulting from a mutation in 07CG27, wherein said method comprises treating an animal which has a disease and which is homozygous for 07CG27 containing said mutation with a drug wherein if said disease regresses said drug is a drug candidate for treating said disease.
42. The method of claim 40 or 41 wherein said animal is transgenic for 07CG27 with said mutation.
43. A method of screening for drug candidates useful in treating a disease resulting from a mutation in 07CG27, wherein said method comprises the steps of:
(a) growing a cell culture of cells which are homozygous for 07CG27 containing said mutation in the presence of a drug,
(b) growing a cell culture of cells which contain a wild-type 07CG27 gene, and
(c) growing a cell culture of cells which are homozygous for 07CG27 containing said mutation in the absence of said drug, wherein if the cells in step (a) behave more like the cells in step (b) than like the cells in step (c) then said drug is a drug candidate for treating said disease.
44. A method of screening for drug candidates useful in treating a disease resulting from an overexpressin of 07CG27, wherein said method involves mixing a wild-type 07CG27 with ras in both the presence of a drug and the absence of said drug and measuring the amount of activation of ras, wherein if the amount of said activation is less in the presence of said drug than in the absence of said drug then said drug is a drug candidate for treating said disease.
45. The method of claim 44 wherein said mutant 07CG27 is a fusion protein.
46. A method of screening for drug candidates useful in treating a disease resulting from an overexpression of 07CG27, wherein said method comprises treating an animal which overexpresses 07CG27 with a drug, wherein if said animal does not develop said disease, said drug is a drug candidate for treating said disease.
47. A method of screening for drug candidates useful in treating a disease resulting from an overexpression O 07CG27, wherein said method comprises treating an animal which has a disease and which overexpresses 07CG27 with a drug, wherein if said disease regresses said drug is a drug candidate for treating said disease.
48. A method of screening for drug candidates useful in treating a disease resulting from an overexpression O 07CG27, wherein said method comprises the steps of:
(a) growing a cell culture of cells which overexpress 07CG27 in the presence of a drug, (b) growing a cell culture of cells which have a normal expression of 07CG27, and
(c) growing a cell culture of cells which overexpress 07CG27 in the absence of said drug, wherein if the cells in step (a) behave more like the cells in step (b) than like the cells in step (c) then said drug is a drug candidate for treating said disease.
PCT/US2000/023291 1999-08-27 2000-08-25 07cg27 gene WO2001016291A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU70709/00A AU7070900A (en) 1999-08-27 2000-08-25 07cg27 gene

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US15104999P 1999-08-27 1999-08-27
US60/151,049 1999-08-27

Publications (2)

Publication Number Publication Date
WO2001016291A2 true WO2001016291A2 (en) 2001-03-08
WO2001016291A3 WO2001016291A3 (en) 2001-07-26

Family

ID=22537122

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/023291 WO2001016291A2 (en) 1999-08-27 2000-08-25 07cg27 gene

Country Status (2)

Country Link
AU (1) AU7070900A (en)
WO (1) WO2001016291A2 (en)

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: 'ESP yeast protein expression and purification system', 1997, STRATAGENE 1997/1998 CATALOG XP002935290 * page 45 * *
CAPECCHI M.R.: 'Targeted gene replacement' SCIENTIFIC AMERICAN March 1994, pages 34 - 41, XP002935291 *
DATABASE GENBANK [Online] 19 July 1999 RHODES ET AL.: 'Novel human mRNA from chromosome 1, which has similarities to BAT2 genes', XP002935289 Database accession no. AL096857 *
THEILER KARL: 'The house mouse', 1989, SPRINGER-VERLAG, NEW YORK XP002935292 * page 148 - page 149 * *

Also Published As

Publication number Publication date
AU7070900A (en) 2001-03-26
WO2001016291A3 (en) 2001-07-26

Similar Documents

Publication Publication Date Title
EP0699754B1 (en) Method for diagnosing a predisposition for breast and ovarian cancer
EP0705903B1 (en) Mutations in the 17q-linked breast and ovarian cancer susceptibility gene
US5747282A (en) 17Q-linked breast and ovarian cancer susceptibility gene
US5709999A (en) Linked breast and ovarian cancer susceptibility gene
US5710001A (en) 17q-linked breast and ovarian cancer susceptibility gene
US5693473A (en) Linked breast and ovarian cancer susceptibility gene
EP0785216A1 (en) Chomosome 13-linked breast cancer susceptibility gene BRCA2
US5753441A (en) 170-linked breast and ovarian cancer susceptibility gene
EP1260520A2 (en) Chromosome 13-linked breast cancer susceptibility gene
US6440699B1 (en) Prostate cancer susceptible CA7 CG04 gene
US6465629B1 (en) BRG1 is a tumor suppressor that is mutated in prostate and other cancer types
US20040265882A1 (en) Diabetes gene
US6844189B1 (en) Chromosome 17P-linked prostate cancer susceptibility gene
WO2001016291A2 (en) 07cg27 gene
US6337192B1 (en) MMSC1-an MMAC1 interacting protein
WO2000012694A1 (en) Chromosome 1-linked prostate cancer susceptibility gene and multisite tumor suppressor
JP3241736B2 (en) 17q-linked breast and ovarian cancer susceptibility genes
US6291173B1 (en) MMSC2—an MMAC1 interacting protein
US6395488B1 (en) Cholecystokinin (CCK) gene as a risk factor for smoking in women

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 CR CU CZ DE DK DM DZ 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 PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA 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 ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ 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 PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

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

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

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

Ref country code: JP