US20110008393A1 - Nucleic acids and corresponding proteins entitled 58p1d12 useful in treatment and detection of cancer - Google Patents

Nucleic acids and corresponding proteins entitled 58p1d12 useful in treatment and detection of cancer Download PDF

Info

Publication number
US20110008393A1
US20110008393A1 US12/836,449 US83644910A US2011008393A1 US 20110008393 A1 US20110008393 A1 US 20110008393A1 US 83644910 A US83644910 A US 83644910A US 2011008393 A1 US2011008393 A1 US 2011008393A1
Authority
US
United States
Prior art keywords
protein
cell
cancer
amino acid
seq
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/836,449
Inventor
Steven B. Kanner
Aya Jakobovits
Pia M. Challita-Eid
Wangmao Ge
Arthur B. Raitano
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Agensys Inc
Original Assignee
Agensys Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/638,203 external-priority patent/US6602501B1/en
Application filed by Agensys Inc filed Critical Agensys Inc
Priority to US12/836,449 priority Critical patent/US20110008393A1/en
Publication of US20110008393A1 publication Critical patent/US20110008393A1/en
Assigned to AGENSYS, INC. reassignment AGENSYS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHALLITA-EID, PIA M., GE, WANGMAO, JAKOBOVITS, AYA, KANNER, STEVEN B., RAITANO, ARTHUR B.
Abandoned legal-status Critical Current

Links

Images

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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/7056Lectin superfamily, e.g. CD23, CD72
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4615Dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • 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
    • C12Q1/6881Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for tissue or cell typing, e.g. human leukocyte antigen [HLA] probes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57434Specifically defined cancers of prostate
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/026Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a baculovirus
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2799/00Uses of viruses
    • C12N2799/02Uses of viruses as vector
    • C12N2799/021Uses of viruses as vector for the expression of a heterologous nucleic acid
    • C12N2799/027Uses of viruses as vector for the expression of a heterologous nucleic acid where the vector is derived from a retrovirus
    • 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
    • C12Q2600/136Screening for pharmacological compounds
    • 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
    • C12Q2600/156Polymorphic or mutational markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value
    • G01N2500/04Screening involving studying the effect of compounds C directly on molecule A (e.g. C are potential ligands for a receptor A, or potential substrates for an enzyme A)

Definitions

  • the invention described herein relates to genes and their encoded proteins, termed 58P1D12 and variants thereof, expressed in certain cancers, and to diagnostic and therapeutic methods and compositions useful in the management of cancers that express 58P1D12.
  • Cancer is the second leading cause of human death next to coronary disease. Worldwide, millions of people die from cancer every year. In the United States alone, as reported by the American Cancer Society, cancer causes the death of well over a half-million people annually, with over 1.2 million new cases diagnosed per year. While deaths from heart disease have been declining significantly, those resulting from cancer generally are on the rise. In the early part of the next century, cancer is predicted to become the leading cause of death.
  • carcinomas of the lung, prostate, breast, colon, pancreas, and ovary represent the primary causes of cancer death. These and virtually all other carcinomas share a common lethal feature. With very few exceptions, metastatic disease from a carcinoma is fatal. Moreover, even for those cancer patients who initially survive their primary cancers, common experience has shown that their lives are dramatically altered. Many cancer patients experience strong anxieties driven by the awareness of the potential for recurrence or treatment failure. Many cancer patients experience physical debilitations following treatment. Furthermore, many cancer patients experience a recurrence.
  • prostate cancer is the fourth most prevalent cancer in men. In North America and Northern Europe, it is by far the most common cancer in males and is the second leading cause of cancer death in men. In the United States alone, well over 30,000 men die annually of this disease—second only to lung cancer. Despite the magnitude of these figures, there is still no effective treatment for metastatic prostate cancer. Surgical prostatectomy, radiation therapy, hormone ablation therapy, surgical castration and chemotherapy continue to be the main treatment modalities. Unfortunately, these treatments are ineffective for many and are often associated with undesirable consequences.
  • PSA serum prostate specific antigen
  • the LAPC Los Angeles Prostate Cancer
  • SCID severe combined immune deficient mice
  • More recently identified prostate cancer markers include PCTA-1 (Su et al., 1996, Proc. Natl. Acad. Sci.
  • PSM prostate-specific membrane
  • STEAP Human, et al., Proc Natl Acad Sci U S A. 1999 Dec. 7; 96(25): 14523-8
  • PSCA prostate stem cell antigen
  • Renal cell carcinoma accounts for approximately 3 percent of adult malignancies. Once adenomas reach a diameter of 2 to 3 cm, malignant potential exists. In the adult, the two principal malignant renal tumors are renal cell adenocarcinoma and transitional cell carcinoma of the renal pelvis or urethras. The incidence of renal cell adenocarcinoma is estimated at more than 29,000 cases in the United States, and more than 11,600 patients died of this disease in 1998. Transitional cell carcinoma is less frequent, with an incidence of approximately 500 cases per year in the United States.
  • bladder cancer represents approximately 5 percent in men (fifth most common neoplasm) and 3 percent in women (eighth most common neoplasm). The incidence is increasing slowly, concurrent with an increasing older population. In 1998, there was an estimated 54,500 cases, including 39,500 in men and 15,000 in women. The age-adjusted incidence in the United States is 32 per 100,000 for men and eight per 100,000 in women. The historic male/female ratio of 3:1 may be decreasing related to smoking patterns in women. There were an estimated 11,000 deaths from bladder cancer in 1998 (7,800 in men and 3,900 in women). Bladder cancer incidence and mortality strongly increase with age and will be an increasing problem as the population becomes more elderly.
  • bladder cancers recur in the bladder.
  • Bladder cancer is managed with a combination of transurethral resection of the bladder (TUR) and intravesical chemotherapy or immunotherapy.
  • TUR transurethral resection of the bladder
  • the multifocal and recurrent nature of bladder cancer points out the limitations of TUR.
  • Most muscle-invasive cancers are not cured by TUR alone. Radical cystectomy and urinary diversion is the most effective means to eliminate the cancer but carry an undeniable impact on urinary and sexual function. There continues to be a significant need for treatment modalities that are beneficial for bladder cancer patients.
  • Treatment options for lung and bronchial cancer are determined by the type and stage of the cancer and include surgery, radiation therapy, and chemotherapy. For many localized cancers, surgery is usually the treatment of choice. Because the disease has usually spread by the time it is discovered, radiation therapy and chemotherapy are often needed in combination with surgery. Chemotherapy alone or combined with radiation is the treatment of choice for small cell lung cancer; on this regimen, a large percentage of patients experience remission, which in some cases is long lasting. There is however, an ongoing need for effective treatment and diagnostic approaches for lung and bronchial cancers.
  • treatment of breast cancer may involve lumpectomy (local removal of the tumor) and removal of the lymph nodes under the arm; mastectomy (surgical removal of the breast) and removal of the lymph nodes under the arm; radiation therapy; chemotherapy; or hormone therapy.
  • lumpectomy local removal of the tumor
  • mastectomy surgical removal of the breast
  • radiation therapy chemotherapy
  • hormone therapy chemotherapy
  • two or more methods are used in combination.
  • Numerous studies have shown that, for early stage disease, long-term survival rates after lumpectomy plus radiotherapy are similar to survival rates after modified radical mastectomy.
  • Significant advances in reconstruction techniques provide several options for breast reconstruction after mastectomy. Recently, such reconstruction has been done at the same time as the mastectomy.
  • DCIS ductal carcinoma in situ
  • Surgery, radiation therapy, and chemotherapy are treatment options for ovarian cancer.
  • Surgery usually includes the removal of one or both ovaries, the fallopian tubes (salpingo-oophorectomy), and the uterus (hysterectomy).
  • the fallopian tubes salivary-oophorectomy
  • the uterus hematoma-oophorectomy
  • pancreatic cancer There were an estimated 28,300 new cases of pancreatic cancer in the United States in 2000. Over the past 20 years, rates of pancreatic cancer have declined in men. Rates among women have remained approximately constant but may be beginning to decline. Pancreatic cancer caused an estimated 28,200 deaths in 2000 in the United States. Over the past 20 years, there has been a slight but significant decrease in mortality rates among men (about ⁇ 0.9% per year) while rates have increased slightly among women.
  • pancreatic cancer Surgery, radiation therapy, and chemotherapy are treatment options for pancreatic cancer. These treatment options can extend survival and/or relieve symptoms in many patients but are not likely to produce a cure for most. There is a significant need for additional therapeutic and diagnostic options for pancreatic cancer.
  • the present invention relates to a gene, designated 58P1D12, that has now been found to be over-expressed in the cancer(s) listed in Table I.
  • Northern blot expression analysis of 58P1D12 gene expression in normal tissues shows a restricted expression pattern in adult tissues.
  • the nucleotide ( FIG. 2 ) and amino acid ( FIG. 2 , and FIG. 3 ) sequences of 58P1D12 are provided.
  • tissue-related profile of 58P1D12 in normal adult tissues combined with the over-expression observed in the tissues listed in Table I, shows that 58P1D12 is aberrantly over-expressed in at least some cancers, and thus serves as a useful diagnostic, prophylactic, prognostic, and/or therapeutic target for cancers of the tissue(s) such as those listed in Table I.
  • the invention provides polynucleotides corresponding or complementary to all or part of the 58P1D12 genes, mRNAs, and/or coding sequences, preferably in isolated form, including polynucleotides encoding 58P1D12-related proteins and fragments of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or more than 25 contiguous amino acids; at least 30, 35, 40, 45, 50, 55, 60, 65, 70, 80, 85, 90, 95, 100 or more than 100 contiguous amino acids of a 58P1D12-related protein, as well as the peptides/proteins themselves; DNA, RNA, DNA/RNA hybrids, and related molecules, polynucleotides or oligonucleotides complementary or having at least a 90% homology to the 58P1D12 genes or mRNA sequences or parts thereof, and polynucleotides or oligonucleotides that hybridize to the 58P1D12
  • the invention further provides antibodies that bind to 58P1D12 proteins and polypeptide fragments thereof, including polyclonal and monoclonal antibodies, murine and other mammalian antibodies, chimeric antibodies, humanized and fully human antibodies, and antibodies labeled with a detectable marker or therapeutic agent.
  • 58P1D12 proteins and polypeptide fragments thereof including polyclonal and monoclonal antibodies, murine and other mammalian antibodies, chimeric antibodies, humanized and fully human antibodies, and antibodies labeled with a detectable marker or therapeutic agent.
  • the entire nucleic acid sequence of FIG. 2 is encoded and/or the entire amino acid sequence of FIG. 2 is prepared, either of which are in respective human unit dose forms.
  • the invention further provides methods for detecting the presence and status of 58P1D12 polynucleotides and proteins in various biological samples, as well as methods for identifying cells that express 58P1D12.
  • a typical embodiment of this invention provides methods for monitoring 58P1D12 gene products in a tissue or hematology sample having or suspected of having some form of growth dysregulation such as cancer.
  • the invention further provides various immunogenic or therapeutic compositions and strategies for treating cancers that express 58P1D12 such as cancers of tissues listed in Table I, including therapies aimed at inhibiting the transcription, translation, processing or function of 58P1D12 as well as cancer vaccines.
  • the invention provides compositions, and methods comprising them, for treating a cancer that expresses 58P1D12 in a human subject wherein the composition comprises a carrier suitable for human use and a human unit dose of one or more than one agent that inhibits the production or function of 58P1D12.
  • the carrier is a uniquely human carrier.
  • the agent is a moiety that is immunoreactive with 58P1D12 protein.
  • Non-limiting examples of such moieties include, but are not limited to, antibodies (such as single chain, monoclonal, polyclonal, humanized, chimeric, or human antibodies), functional equivalents thereof (whether naturally occurring or synthetic), and combinations thereof.
  • the antibodies can be conjugated to a diagnostic or therapeutic moiety.
  • the agent is a small molecule as defined herein.
  • the agent comprises one or more than one peptide which comprises a cytotoxic T lymphocyte (CTL) epitope that binds an HLA class I molecule in a human to elicit a CTL response to 58P1D12 and/or one or more than one peptide which comprises a helper T lymphocyte (HTL) epitope which binds an HLA class II molecule in a human to elicit an HTL response.
  • CTL cytotoxic T lymphocyte
  • HTL helper T lymphocyte
  • the peptides of the invention may be on the same or on one or more separate polypeptide molecules.
  • the agent comprises one or more than one nucleic acid molecule that expresses one or more than one of the CTL or HTL response stimulating peptides as described above.
  • the one or more than one nucleic acid molecule may express a moiety that is immunologically reactive with 58P1D12 as described above.
  • the one or more than one nucleic acid molecule may also be, or encodes, a molecule that inhibits production of 58P1D12.
  • Non-limiting examples of such molecules include, but are not limited to, those complementary to a nucleotide sequence essential for production of 58P1D12 (e.g. antisense sequences or molecules that form a triple helix with a nucleotide double helix essential for 58P1D12 production) or a ribozyme effective to lyse 58P1D12 mRNA.
  • HLA Peptide Tables respective to its parental protein, e.g., variant 1, variant 2, etc.
  • HLA Peptide Tables respective to its parental protein, e.g., variant 1, variant 2, etc.
  • search Peptides in Table VII Generally, a unique Search Peptide is used to obtain HLA peptides of a particular for a particular variant. The position of each Search Peptide relative to its respective parent molecule is listed in Table VII.
  • a Search Peptide begins at position “X”, one must add the value “X ⁇ 1” to each position in Tables VIII-XXI and XXII to XLIX to obtain the actual position of the HLA peptides in their parental molecule. For example, if a particular Search Peptide begins at position 150 of its parental molecule, one must add 150 ⁇ 1, i.e., 149 to each HLA peptide amino acid position to calculate the position of that amino acid in the parent molecule.
  • One embodiment of the invention comprises an HLA peptide, that occurs at least twice in Tables VIII-XXI and XXII to XLIX collectively, or an oligonucleotide that encodes the HLA peptide.
  • Another embodiment of the invention comprises an HLA peptide that occurs at least once in Tables VIII-XXI and at least once in tables XXII to XLIX, or an oligonucleotide that encodes the HLA peptide.
  • antibody epitopes which comprise a peptide regions, or an oligonucleotide encoding the peptide region, that has one two, three, four, or five of the following characteristics:
  • a peptide region of at least 5 amino acids of a particular peptide of FIG. 3 in any whole number increment up to the full length of that protein in FIG. 3 , that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Hydrophilicity profile of FIG. 5 ;
  • a peptide region of at least 5 amino acids of a particular peptide of FIG. 3 in any whole number increment up to the full length of that protein in FIG. 3 , that includes an amino acid position having a value equal to or less than 0.5, 0.4, 0.3, 0.2, 0.1, or having a value equal to 0.0, in the Hydropathicity profile of FIG. 6 ;
  • a peptide region of at least 5 amino acids of a particular peptide of FIG. 3 in any whole number increment up to the full length of that protein in FIG. 3 , that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Percent Accessible Residues profile of FIG. 7 ;
  • a peptide region of at least 5 amino acids of a particular peptide of FIG. 3 in any whole number increment up to the full length of that protein in FIG. 3 , that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Average Flexibility profile of FIG. 8 ; or
  • FIG. 1 The 58P1D12 SSH sequence of 427 nucleotides.
  • FIG. 2A The cDNA and amino acid sequence of 58P1D12 variant 1 (also called “58P1D12 v.1” or “58P1D12 variant 1”) is shown in FIG. 2A .
  • the start methionine is underlined.
  • the open reading frame extends from nucleic acid 380-1201 including the stop codon.
  • FIG. 2B The cDNA and amino acid sequence of 58P1D12 variant 2 (also called “58P1D12 v.2”) is shown in FIG. 2B .
  • the codon for the start methionine is underlined.
  • the open reading frame extends from nucleic acid 388-1086 including the stop codon.
  • FIG. 2C The cDNA and amino acid sequence of 58P1D12 variant 3 (also called “58P1D12 v.3”) is shown in FIG. 2C .
  • the codon for the start methionine is underlined.
  • the open reading frame extends from nucleic acid 206-904 including the stop codon.
  • FIG. 2D The cDNA and amino acid sequence of 58P1D12 variant 4 (also called “58P1D12 v.4”) is shown in FIG. 2D .
  • the codon for the start methionine is underlined.
  • the open reading frame extends from nucleic acid 206-916 including the stop codon.
  • FIG. 2E The cDNA and amino acid sequence of 58P1D12 variant 5 (also called “58P1D12 v.5”) is shown in FIG. 2E .
  • the codon for the start methionine is underlined.
  • the open reading frame extends from nucleic acid 106-816 including the stop codon.
  • FIG. 2F Variants 58P1D12 v.6 through v.15 are variants with single nucleotide difference from 58P1D12 v.1. Though these SNP variants are shown separately, they can also occur in any combinations and in any of the transcript variants listed above in FIGS. 2A-2F .
  • FIG. 3A The amino acid sequence of 58P1D12 v.1 is shown in FIG. 3A ; it has 273 amino acids.
  • FIG. 3B The amino acid sequence of 58P1D12 v.2 is shown in FIG. 3B ; it has 232 amino acids.
  • FIG. 3C The amino acid sequence of 58P1D12 v.3 is shown in FIG. 3C ; it has 236 amino acids.
  • 58P1D12 includes all variants thereof, including those shown in FIGS. 2 , 3 , 10 , 11 , and 12 unless the context clearly indicates otherwise.
  • FIG. 4 Intratibial tumor growth of 3T3-58P1D12 cells.
  • 3T3 fibroblasts were infected with empty vector control amphotropic virus (Neo) or virus containing the 58P1D12 gene. Stable transfectants were selected in G-418 (geneticin) and then grown in 10% FBS.
  • G-418 amphotropic virus
  • 3T3-Neo and 3T3-58P1D12 1 ⁇ 10 6 cells were mixed with Matrigel® and injected into the flanks of 7 male SCID mice. After 31 days, the mice were sacrified, and the tumors were extracted with the hind quarters. The data are representative of the 7 mice treated with the two cell types. The data indicate that the expression of 58P1D12 in the non-tumor forming 3T3 line has increased tumor growth potential in vivo.
  • FIG. 10 Structures of transcript variants of 58P1D12. Variant 58P1D12 v.2 through v.5 were transcript variants of 58P1D12 v.1. Variants 58P1D12 v.2 and v.3, each had two different exons in the 5′ end, shared five exons in the 3′ end with v.1. Variants v.4 and v.5 were similar to v.3, with deletions of one or two exons from v.3. All the exons in different transcript variants were aligned in relative order as on the genome. Poly A tails and SNP are not shown here. Numbers in “( )” underneath the boxes correspond to those of 58P1D12 v.1. Lengths of introns and exons are not proportional.
  • FIG. 11 Schematic alignment of protein variants of 58P1D12. Protein variants corresponded to nucleotide variants. Nucleotide variants 58P1D12 v.3 coded for the same protein as variant v.2, and variant v.5 coded for the same protein as v.4. SNP variants v.6 through v.15 coded for the same protein as v.1. Nucleotide variants 58P1D12 v.6 through v.15 each had one alternative allele of a SNP as compared with variant v.1, as shown in FIG. 12 .
  • Proteins of v.2 and v.3 were portion of v.1 protein, while variants v.4 and v.5 had a portion that was different from any port of v.1. Numbers underneath the box correspond to 58P1D12 v.1. Black boxes represent the same sequence as 58P1D12 v.1.
  • FIG. 12 Schematic alignment of SNP variants of 58P1D12. Variants 58P1D12 v.6 through v.12 were variants with single nucleotide differences as compared to variant v.1. Though these SNP variants were shown separately, they could also occur in any combinations (called haplotype), and in any transcript variants that contained the base pairs, such as v.2 shown in FIG. 10 . Numbers correspond to those of 58P1D12 v.1. Black box shows the same sequence as 58P1D12 v.1. SNPs are indicated above the box.
  • FIG. 13 Secondary structure and transmembrane domains prediction for 58P1D12 protein variants.
  • FIG. 13A-13C The secondary structures of 58P1D12 protein variants 1 (SEQ ID NO:15), variants 2 (SEQ ID NO:16), variant 3 (SEQ ID NO:17), respectively, were predicted using the HNN—Hierarchical Neural Network method (NPS@: Network Protein Sequence Analysis TIBS 2000 March Vol. 25, No 3 [291]:147-150 Combet C., Blanchet C., Geourjon C.
  • NPS@ Network Protein Sequence Analysis TIBS 2000 March Vol. 25, No 3 [291]:147-150 Combet C., Blanchet C., Geourjon C.
  • FIG. 13D , FIG. 13F , FIG. 13H Schematic representation of the probability of existence of transmembrane regions of 58P1D12 protein variants 1, 2, 3, respectively, based on the TMpred algorithm of Hofmann and Stoffel which utilizes TMBASE (K. Hofmann, W. Stoffel. TMBASE—A database of membrane spanning protein segments Biol. Chem. Hoppe-Seyler 374:166, 1993).
  • FIG. 13E , FIG. 13G , FIG. 13I Schematic representation of the probability of the existence of transmembrane regions of 58P1D12 variants 1, 2, 3, respectively, based on the TMHMM algorithm of Sonnhammer, von Heijne, and Krogh (Erik L.L. Sonnhammer, Gunnar von Heijne, and Anders Krogh: A hidden Markov model for predicting transmembrane helices in protein sequences. In Proc. of Sixth Int. Conf. on Intelligent Systems for Molecular Biology, p 175-182 Ed J. Glasgow, T. Littlejohn, F. Major, R. Lathrop, D. Sankoff, and C. Sensen Menlo Park, Calif.: AAAI Press, 1998).
  • the TMpred and TMHMM algorithms are accessed from the ExPasy molecular biology server located on the World Wide Web at (.expasy.ch/tools/).
  • FIG. 14 58P1D12 Expression in Normal and Patient Cancer Tissues.
  • First strand cDNA was prepared from normal tissues (bladder, brain, heart, kidney, liver, lung, prostate, spleen, skeletal muscle, testis, pancreas, colon and stomach), and from pools of patient cancer specimens (prostate cancer pool, bladder cancer pool, lung cancer pool, ovary cancer pool, prostate cancer xenograft pool, bone and melanoma cancer pool, and cervical cancer pool). Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using primers to 58P1D12, was performed at 30 cycles of amplification. Results expression of 58P1D12 predominantly in testis amongst the normal tissues tested. 58P1D12 is also expressed in prostate cancer, bladder cancer, lung cancer, ovary cancer, prostate cancer xenograft, bone/melanoma cancer pool, and cervical cancer.
  • FIG. 15 Expression of 58P1D12 in normal tissues. Two multiple tissue northern blots (Clontech) both with 2 ug of mRNA/lane were probed with the 58P1D12 sequence. Size standards in kilobases (kb) are indicated on the side. Results show predominant expression of 58P1D12 transcript in normal testis. A significantly weaker signal is detected in spleen, lung, kidney and pancreas.
  • FIG. 16 58P1D12 Expression in Prostate Cancer Xenograft tissues.
  • RNA was extracted from LAPC prostate cancer xenografts, LAPC-4AD, LAPC-4AI, LAPC-9AD and LAPC-9AI, as well as from normal prostate.
  • Northern blots with 10 ug of total RNA were probed with the 58P1D12 sequence. Size standards in kilobases are on the side. Results show expression of 58P1D12 in LAPC-9AD but not in the other tissues tested.
  • FIG. 17 58P1D12 Expression in Normal and Patient Cancer Specimens.
  • RNA was extracted from a pool of 3 ovary tumor patient specimens, normal prostate (NP), normal bladder (NB), normal kidney (NK), and normal colon (NC).
  • Northern blots with 10 ug of total RNA were probed with the 58P1D12 sequence. Size standards in kilobases are on the side. Results show expression of 58P1D12 in ovary tumor patient specimens but not in the normal tissues tested.
  • FIG. 18 58P1D12 Expression in Ovarian Cancer Patient Specimens. RNA was extracted from normal ovary, LAPC-9AD prostate cancer xenograft, and a panel of ovary tumor patient specimens (Ovary T). Northern blots with 10 ug of total RNA were probed with the 58P1D12 sequence. Size standards in kilobases are on the side. Results show expression of 58P1D12 in most of the patient specimens tested as well as in the prostate xenograft.
  • FIG. 19 58P1D12 Expression in a Large Panel of Ovarian Cancer Patient Specimens.
  • First strand cDNA was prepared from a panel of ovary patient cancer specimens as well as two different normal ovary tissues. Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using primers to 58P1D12, was performed at 26 and 30 cycles of amplification. Samples were run on an agarose gel, and PCR products were quantitated using the AlphaImager software. Expression was recorded as negative, weak, medium or strong. Results show expression of 58P1D12 in the majority of patient cancer specimens tested (75%), but not in normal ovary.
  • FIG. 20 Specific cell surface staining of Rat1-58P1D12 cells with serum from mice immunized with recombinant 58P1D12 protein.
  • Rat1-58P1D12 and control Rat1-neo cells ( ⁇ 100,000 cells) were incubated with various dilutions of serum obtained from 2 different mice immunized with recombinant Tag5-58P1D12 protein. Cells were washed and then incubated with goat anti-mouse IgG-PE secondary Ab (1:100 dilution in PBS+2% FBS) on ice for 1 hour. Cells were washed again, fixed in 2% paraformaldehyde in PBS, and subjected to FACS analysis. The Rat1-58P1D12 cells, but not the Rat1-neo cells, show a shift in fluorescent intensity over a large dilution range demonstrating specific binding of anti-58P1D12 Abs present in the serum from the 2 mice.
  • FIG. 21 Specific cell surface staining of Rat1-58P1D12 cells with monoclonal antibody hybridoma supernatants.
  • Rat1-58P1D12 and control Rat1-neo cells (100,000 cells) were incubated with supernatants derived from hybridomas M8-143(5).10.1 and M8-143(5).45.1. on ice for 1 hour. Cells were washed and then incubated with goat anti-mouse IgG-PE secondary Ab (1:100 dilution in PBS+2% FBS) on ice for 1 hour. Cells were washed again, fixed in 2% paraformaldehyde in PBS, and subjected to FACS analysis. The Rat1-58P1D12 cells, but not the Rat1-neo cells, show a shift in fluorescent intensity demonstrating specific binding of the MAb to 58P1D12 protein on the cell surface.
  • FIG. 22 Affinity determination of MAb M8-143(5).10.1 by FACS-based equilibrium binding analysis. 3T3-58P1D12 cells were incubated at 4 C for 2 hours with various concentrations of MAb M8-143(5).10.1 in PBS+2% FBS+0.04% NaN3. Cells were washed, and then incubated at 4 C for 1 hour with a 1:100 dilution of goat anti-mouse IgG-PE secondary Ab. After washing, cells were fixed in 2% paraformaldehyde in PBS and subjected to FACS analysis. Shown on the left are the histograms of cells stained with various concentrations of MAb.
  • MAb M8-143(5).10.1 is a high affinity MAb with an apparent KD of 0.086 nM on 3T3-58P1D12 cells.
  • FIG. 23 58P1D12 induces angiogenesis (tube formation) in umbilical vein endothelial cells (HUVEC).
  • UUVEC umbilical vein endothelial cells
  • Primary umbilical vein endothelial cells were cultured in 10% FBS, then removed with EDTA and mixed with media containing either 10% FBS, 0.1% FBS, 0.1% FBS+50 ng/ml VEGF or 0.1% FBS+5 ⁇ g/ml 58P1D12 ECD protein.
  • the mixtures were plated onto a 200 ⁇ l layer of Matrigel® and then incubated at 37° C. for 16 hours. The plates were then analyzed for the formation of tubes by microscopy. Images of representative fields are shown.
  • HUVEC cells form tubes in 10% FBS but not in 0.1% FBS, thereby allowing the assessment of 58P1D12-induced angiogenesis in low serum conditions.
  • 58P1D12 ECD exhibited the capacity to induce the formation of endothelial tubes similarly as the positive control VEGF.
  • FIG. 24 58P1D12 induces angiogenesis (tube formation) in lung microvascular endothelial cells (HMVEC).
  • HMVEC lung microvascular endothelial cells
  • Primary lung endothelial cells were cultured in 10% FBS, then removed with EDTA and mixed with media containing either 0.1% FBS, 0.1% FBS+50 ng/ml VEGF or 0.1% FBS+5 ⁇ g/ml 58P1D12 ECD protein. The mixtures were plated onto a 200 ⁇ l layer of Matrigel® and then incubated at 37° C. for 16 hours. The plates were then analyzed for the formation of tubes by microscopy. Images of representative fields are shown.
  • HMVEC cells do not form tubes in 0.1% FBS, thereby allowing the assessment of 58P1D12-induced angiogenesis in low serum conditions.
  • 58P1D12 ECD exhibited the capacity to induce the formation of endothelial tubes similarly as the positive control VEGF.
  • FIG. 25 Monoclonal antibody to 58P1D12 inhibits angiogenesis (tube formation) in umbilical vein endothelial cells (HUVEC).
  • Primary umbilical vein endothelial cells were cultured in 10% FBS, then removed with EDTA and mixed with media containing either 0.1% FBS, 5% FBS, 0.1% FBS+3 ⁇ g/ml 58P1D12 ECD protein, 0.1% FBS+3 ⁇ g/ml 58P1D12 ECD protein+30 ⁇ g/ml MAb M8-143(5)10 or 0.1% FBS+3 ⁇ g/ml 58P1D12 ECD protein+30 ⁇ g/ml MAb M3-47.24.
  • the mixtures were plated onto a 200 ⁇ l layer of Matrigel® and then incubated at 37° C. for 16 hours. The plates were then analyzed for the formation of tubes by microscopy. The total tube counts are shown. The data show that HUVEC cells form tubes in 5% FBS but not in 0.1% FBS, thereby allowing the assessment of 58P1D12-induced angiogenesis in low serum conditions. At 3 ⁇ g/ml, 58P1D12 ECD exhibited the capacity to induce the formation of endothelial tubes, and that incubation with MAb M8-143(5)10, but not MAb M3-47.24, inhibited the tube formation by 45%.
  • FIG. 26 58P1D12 ECD enhances survival of HUVEC.
  • Primary umbilical vein endothelial cells were cultured in 10% FBS. Cells were trypsinized and 3000 cells per well were plated onto 96-well dishes for 72 hours. Cell viability was assayed by Alamar blue staining for 3 hours, followed by fluorescence measurement. The assay was performed in triplicate with standard deviations represented. In low serum conditions (0.1% FBS), cell viability was low, but was enhanced by increasing concentrations of 58P1D12 ECD. For positive control, the cells were treated with 50 ng/ml VEGF. Cell viability at 10 ⁇ g/ml 58P1D12 ECD was similar to that observed using 50 ng/ml VEGF (a known survival signal for endothelium).
  • FIG. 27 58P1D12 ECD enhances proliferation of HMVEC.
  • Primary lung microvascular endothelial cells HMVEC
  • HMVEC Primary lung microvascular endothelial cells
  • Cells were trypsinized and 3000 cells per well were plated onto 96-well dishes in 2% FBS.
  • the cells were either left untreated, or were incubated with 50 ng/ml VEGF, 5 ⁇ g/ml 58P1D12 ECD or 10 ⁇ g/ml 58P1D12 ECD. After 48 hours, the cells were incubated with 3H-thymidine and allowed to grow overnight. Incorporation of 3H-thymidine into cell DNA was assayed by standard protocols. The assay was performed in triplicate with standard deviations represented.
  • prostate cancer locally advanced prostate cancer
  • advanced disease and “locally advanced disease” mean prostate cancers that have extended through the prostate capsule, and are meant to include stage C disease under the American Urological Association (AUA) system, stage C1-C2 disease under the Whitmore-Jewett system, and stage T3-T4 and N+ disease under the TNM (tumor, node, metastasis) system.
  • AUA American Urological Association
  • stage C1-C2 disease under the Whitmore-Jewett system
  • T3-T4 and N+ disease under the TNM (tumor, node, metastasis) system.
  • surgery is not recommended for patients with locally advanced disease, and these patients have substantially less favorable outcomes compared to patients having clinically localized (organ-confined) prostate cancer.
  • Locally advanced disease is clinically identified by palpable evidence of induration beyond the lateral border of the prostate, or asymmetry or induration above the prostate base.
  • Locally advanced prostate cancer is presently diagnosed pathologically following radical prostatectomy
  • “Altering the native glycosylation pattern” is intended for purposes herein to mean deleting one or more carbohydrate moieties found in native sequence 58P1D12 (either by removing the underlying glycosylation site or by deleting the glycosylation by chemical and/or enzymatic means), and/or adding one or more glycosylation sites that are not present in the native sequence 58P1D12.
  • the phrase includes qualitative changes in the glycosylation of the native proteins, involving a change in the nature and proportions of the various carbohydrate moieties present.
  • an analog refers to a molecule which is structurally similar or shares similar or corresponding attributes with another molecule (e.g. a 58P1D12-related protein).
  • a 58P1D12-related protein e.g. an analog of a 58P1D12 protein can be specifically bound by an antibody or T cell that specifically binds to 58P1D12.
  • Antibody is used in the broadest sense. Therefore, an “antibody” can be naturally occurring or man-made such as monoclonal antibodies produced by conventional hybridoma technology.
  • Anti-58P1D12 antibodies comprise monoclonal and polyclonal antibodies as well as fragments containing the antigen-binding domain and/or one or more complementarity determining regions of these antibodies.
  • an “antibody fragment” is defined as at least a portion of the variable region of the immunoglobulin molecule that binds to its target, i.e., the antigen-binding region. In one embodiment it specifically covers single anti-58P1D12 antibodies and clones thereof (including agonist, antagonist and neutralizing antibodies) and anti-58P1D12 antibody compositions with polyepitopic specificity.
  • codon optimized sequences refers to nucleotide sequences that have been optimized for a particular host species by replacing any codons having a usage frequency of less than about 20%. Nucleotide sequences that have been optimized for expression in a given host species by elimination of spurious polyadenylation sequences, elimination of exon/intron splicing signals, elimination of transposon-like repeats and/or optimization of GC content in addition to codon optimization are referred to herein as an “expression enhanced sequences.”
  • a “combinatorial library” is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis by combining a number of chemical “building blocks” such as reagents.
  • a linear combinatorial chemical library such as a polypeptide (e.g., mutein) library, is formed by combining a set of chemical building blocks called amino acids in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound).
  • Numerous chemical compounds are synthesized through such combinatorial mixing of chemical building blocks (Gallop et al., J. Med. Chem. 37(9): 1233-1251 (1994)).
  • combinatorial chemical libraries include, but are not limited to, peptide libraries (see, e.g., U.S. Pat. No. 5,010,175, Furka, Pept. Prot. Res. 37:487-493 (1991), Houghton et al., Nature, 354:84-88 (1991)), peptoids (PCT Publication No WO 91/19735), encoded peptides (PCT Publication WO 93/20242), random bio-oligomers (PCT Publication WO 92/00091), benzodiazepines (U.S. Pat. No.
  • cytotoxic agent refers to a substance that inhibits or prevents the expression activity of cells, function of cells and/or causes destruction of cells.
  • the term is intended to include radioactive isotopes chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof.
  • cytotoxic agents include, but are not limited to auristatins, auromycins, maytansinoids, yttrium, bismuth, ricin, ricin A-chain, combrestatin, duocarmycins, dolostatins, doxorubicin, daunorubicin, taxol, cisplatin, cc1065, ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, dihydroxy anthracin dione, actinomycin, diphtheria toxin, Pseudomonas exotoxin (PE) A, PE40, abrin, abrin A chain, modeccin A chain, alpha-sarcin, gelonin, mitogellin, retstrictocin, phenomycin, enomycin, curicin, crotin, calicheamicin, Sapaonaria officinalis inhibitor
  • the “gene product” is sometimes referred to herein as a protein or mRNA.
  • a “gene product of the invention” is sometimes referred to herein as a “cancer amino acid sequence”, “cancer protein”, “protein of a cancer listed in Table I”, a “cancer mRNA”, “mRNA of a cancer listed in Table I”, etc.
  • the cancer protein is encoded by a nucleic acid of FIG. 2 .
  • the cancer protein can be a fragment, or alternatively, be the full-length protein to the fragment encoded by the nucleic acids of FIG. 2 .
  • a cancer amino acid sequence is used to determine sequence identity or similarity.
  • the sequences are naturally occurring allelic variants of a protein encoded by a nucleic acid of FIG. 2 .
  • the sequences are sequence variants as further described herein.
  • “High throughput screening” assays for the presence, absence, quantification, or other properties of particular nucleic acids or protein products are well known to those of skill in the art.
  • binding assays and reporter gene assays are similarly well known.
  • U.S. Pat. No. 5,559,410 discloses high throughput screening methods for proteins
  • U.S. Pat. No. 5,585,639 discloses high throughput screening methods for nucleic acid binding (i.e., in arrays)
  • U.S. Pat. Nos. 5,576,220 and 5,541,061 disclose high throughput methods of screening for ligand/antibody binding.
  • high throughput screening systems are commercially available (see, e.g., Amersham Biosciences, Piscataway, N.J.; Zymark Corp., Hopkinton, Mass.; Air Technical Industries, Mentor, Ohio; Beckman Instruments, Inc. Fullerton, Calif.; Precision Systems, Inc., Natick, Mass.; etc.). These systems typically automate entire procedures, including all sample and reagent pipetting, liquid dispensing, timed incubations, and final readings of the microplate in detector(s) appropriate for the assay. These configurable systems provide high throughput and rapid start up as well as a high degree of flexibility and customization. The manufacturers of such systems provide detailed protocols for various high throughput systems. Thus, e.g., Zymark Corp. provides technical bulletins describing screening systems for detecting the modulation of gene transcription, ligand binding, and the like.
  • homolog refers to a molecule which exhibits homology to another molecule, by for example, having sequences of chemical residues that are the same or similar at corresponding positions.
  • HLA Human Leukocyte Antigen
  • MHC Major Histocompatibility Complex
  • hybridize used in the context of polynucleotides, are meant to refer to conventional hybridization conditions, preferably such as hybridization in 50% formamide/6 ⁇ SSC/0.1% SDS/100 ⁇ g/ml ssDNA, in which temperatures for hybridization are above 37 degrees C. and temperatures for washing in 0.1 ⁇ SSC/0.1% SDS are above 55 degrees C.
  • isolated or “biologically pure” refer to material which is substantially or essentially free from components which normally accompany the material as it is found in its native state.
  • isolated peptides in accordance with the invention preferably do not contain materials normally associated with the peptides in their in situ environment.
  • a polynucleotide is said to be “isolated” when it is substantially separated from contaminant polynucleotides that correspond or are complementary to genes other than the 58P1D12 genes or that encode polypeptides other than 58P1D12 gene product or fragments thereof.
  • a skilled artisan can readily employ nucleic acid isolation procedures to obtain an isolated 58P1D12 polynucleotide.
  • a protein is said to be “isolated,” for example, when physical, mechanical or chemical methods are employed to remove the 58P1D12 proteins from cellular constituents that are normally associated with the protein.
  • a skilled artisan can readily employ standard purification methods to obtain an isolated 58P1D12 protein.
  • an isolated protein can be prepared by chemical means.
  • mammal refers to any organism classified as a mammal, including mice, rats, rabbits, dogs, cats, cows, horses and humans. In one embodiment of the invention, the mammal is a mouse. In another embodiment of the invention, the mammal is a human.
  • metastatic prostate cancer and “metastatic disease” mean prostate cancers that have spread to regional lymph nodes or to distant sites, and are meant to include stage D disease under the AUA system and stage T ⁇ N ⁇ M+ under the TNM system.
  • surgery is generally not indicated for patients with metastatic disease, and hormonal (androgen ablation) therapy is a preferred treatment modality.
  • Patients with metastatic prostate cancer eventually develop an androgen-refractory state within 12 to 18 months of treatment initiation. Approximately half of these androgen-refractory patients die within 6 months after developing that status.
  • the most common site for prostate cancer metastasis is bone. Prostate cancer bone metastases are often osteoblastic rather than osteolytic (i.e., resulting in net bone formation).
  • Bone metastases are found most frequently in the spine, followed by the femur, pelvis, rib cage, skull and humerus. Other common sites for metastasis include lymph nodes, lung, liver and brain. Metastatic prostate cancer is typically diagnosed by open or laparoscopic pelvic lymphadenectomy, whole body radionuclide scans, skeletal radiography, and/or bone lesion biopsy.
  • modulator or “test compound” or “drug candidate” or grammatical equivalents as used herein describe any molecule, e.g., protein, oligopeptide, small organic molecule, polysaccharide, polynucleotide, etc., to be tested for the capacity to directly or indirectly alter the cancer phenotype or the expression of a cancer sequence, e.g., a nucleic acid or protein sequences, or effects of cancer sequences (e.g., signaling, gene expression, protein interaction, etc.)
  • a modulator will neutralize the effect of a cancer protein of the invention.
  • neutralize is meant that an activity of a protein is inhibited or blocked, along with the consequent effect on the cell.
  • a modulator will neutralize the effect of a gene, and its corresponding protein, of the invention by normalizing levels of said protein.
  • modulators alter expression profiles, or expression profile nucleic acids or proteins provided herein, or downstream effector pathways.
  • the modulator suppresses a cancer phenotype, e.g. to a normal tissue fingerprint.
  • a modulator induced a cancer phenotype.
  • a plurality of assay mixtures is run in parallel with different agent concentrations to obtain a differential response to the various concentrations. Typically, one of these concentrations serves as a negative control, i.e., at zero concentration or below the level of detection.
  • Modulators, drug candidates or test compounds encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 100 and less than about 2,500 Daltons. Preferred small molecules are less than 2000, or less than 1500 or less than 1000 or less than 500 D.
  • Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups.
  • the candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups.
  • Modulators also comprise biomolecules such as peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof. Particularly preferred are peptides.
  • One class of modulators are peptides, for example of from about five to about 35 amino acids, with from about five to about 20 amino acids being preferred, and from about 7 to about 15 being particularly preferred.
  • the cancer modulatory protein is soluble, includes a non-transmembrane region, and/or, has an N-terminal Cys to aid in solubility.
  • the C-terminus of the fragment is kept as a free acid and the N-terminus is a free amine to aid in coupling, i.e., to cysteine.
  • a cancer protein of the invention is conjugated to an immunogenic agent as discussed herein.
  • the cancer protein is conjugated to BSA.
  • the peptides of the invention e.g., of preferred lengths, can be linked to each other or to other amino acids to create a longer peptide/protein.
  • the modulatory peptides can be digests of naturally occurring proteins as is outlined above, random peptides, or “biased” random peptides.
  • peptide/protein-based modulators are antibodies, and fragments thereof, as defined herein.
  • Modulators of cancer can also be nucleic acids.
  • Nucleic acid modulating agents can be naturally occurring nucleic acids, random nucleic acids, or “biased” random nucleic acids. For example, digests of prokaryotic or eukaryotic genomes can be used in an approach analogous to that outlined above for proteins.
  • the term “monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the antibodies comprising the population are identical except for possible naturally occurring mutations that are present in minor amounts.
  • a motif can be either contiguous or capable of being aligned to certain positions that are generally correlated with a certain function or property.
  • motif refers to the pattern of residues in a peptide of defined length, usually a peptide of from about 8 to about 13 amino acids for a class I HLA motif and from about 6 to about 25 amino acids for a class II HLA motif, which is recognized by a particular HLA molecule.
  • Peptide motifs for HLA binding are typically different for each protein encoded by each human HLA allele and differ in the pattern of the primary and secondary anchor residues.
  • a “pharmaceutical excipient” comprises a material such as an adjuvant, a carrier, pH-adjusting and buffering agents, tonicity adjusting agents, wetting agents, preservative, and the like.
  • “Pharmaceutically acceptable” refers to a non-toxic, inert, and/or composition that is physiologically compatible with humans or other mammals.
  • polynucleotide means a polymeric form of nucleotides of at least 10 bases or base pairs in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide, and is meant to include single and double stranded forms of DNA and/or RNA. In the art, this term if often used interchangeably with “oligonucleotide”.
  • a polynucleotide can comprise a nucleotide sequence disclosed herein wherein thymidine (T), as shown for example in FIG. 2 , can also be uracil (U); this definition pertains to the differences between the chemical structures of DNA and RNA, in particular the observation that one of the four major bases in RNA is uracil (U) instead of thymidine (T).
  • polypeptide means a polymer of at least about 4, 5, 6, 7, or 8 amino acids. Throughout the specification, standard three letter or single letter designations for amino acids are used. In the art, this term is often used interchangeably with “peptide” or “protein”.
  • HLA “primary anchor residue” is an amino acid at a specific position along a peptide sequence which is understood to provide a contact point between the immunogenic peptide and the HLA molecule.
  • One to three, usually two, primary anchor residues within a peptide of defined length generally defines a “motif” for an immunogenic peptide. These residues are understood to fit in close contact with peptide binding groove of an HLA molecule, with their side chains buried in specific pockets of the binding groove.
  • the primary anchor residues for an HLA class I molecule are located at position 2 (from the amino terminal position) and at the carboxyl terminal position of a 8, 9, 10, 11, or 12 residue peptide epitope in accordance with the invention.
  • the primary anchor residues of a peptide binds an HLA class II molecule are spaced relative to each other, rather than to the termini of a peptide, where the peptide is generally of at least 9 amino acids in length.
  • the primary anchor positions for each motif and supermotif are set forth in Table IV.
  • analog peptides can be created by altering the presence or absence of particular residues in the primary and/or secondary anchor positions shown in Table IV. Such analogs are used to modulate the binding affinity and/or population coverage of a peptide comprising a particular HLA motif or supermotif.
  • Radioisotopes include, but are not limited to the following (non-limiting exemplary uses are also set forth):
  • Actinium-225 See Thorium-229 (Th-229) (AC-225) Actinium-227 Parent of Radium-223 (Ra-223) which is an alpha emitter used to treat (AC-227) metastases in the skeleton resulting from cancer (i.e., breast and prostate cancers), and cancer radioimmunotherapy Bismuth-212 See Thorium-228 (Th-228) (Bi-212) Bismuth-213 See Thorium-229 (Th-229) (Bi-213) Cadmium-109 Cancer detection (Cd-109) Cobalt-60 Radiation source for radiotherapy of cancer, for food irradiators, and for (Co-60) sterilization of medical supplies Copper-64 A positron emitter used for cancer therapy and SPECT imaging (Cu-64) Copper-67 Beta/gamma emitter used in cancer radioimmunotherapy and diagnostic (Cu-67) studies (i.e., breast and colon cancers, and lymphoma) Dysprosium-166 Cancer radioimmunotherapy (Dy-16)
  • Tc-99m is the most widely used radioisotope used for diagnostic imaging of various cancers and diseases involving the brain, heart, liver, lungs; also used in detection of deep vein thrombosis of the legs Osmium-194 Cancer radioimmunotherapy (Os-194) Palladium-103 Prostate cancer treatment (Pd-103) Platinum-195m
  • a chemotherapeutic (Pt-195m) drug Phosphorus-32 Polycythemia rubra vera (blood cell disease) and leukemia treatment, bone (P-32) cancer diagnosis/treatment; colon, pancreatic, and liver cancer treatment; radiolabeling nucleic acids for in vitro research, diagnosis of superficial tumors, treatment of blocked arteries (i.e., arteriosclerosis and restenosis), and intracavity therapy Phosphorus-33 Leukemia treatment, bone disease diagnosis/treatment, radiolabeling, and (P-33) treatment of blocked arteries (i.e.,
  • each nucleic acid and peptide consists of essentially random nucleotides and amino acids, respectively. These random peptides (or nucleic acids, discussed herein) can incorporate any nucleotide or amino acid at any position.
  • the synthetic process can be designed to generate randomized proteins or nucleic acids, to allow the formation of all or most of the possible combinations over the length of the sequence, thus forming a library of randomized candidate bioactive proteinaceous agents.
  • a library is “fully randomized,” with no sequence preferences or constants at any position.
  • the library is a “biased random” library. That is, some positions within the sequence either are held constant, or are selected from a limited number of possibilities.
  • the nucleotides or amino acid residues are randomized within a defined class, e.g., of hydrophobic amino acids, hydrophilic residues, sterically biased (either small or large) residues, towards the creation of nucleic acid binding domains, the creation of cysteines, for cross-linking, prolines for SH-3 domains, serines, threonines, tyrosines or histidines for phosphorylation sites, etc., or to purines, etc.
  • a “recombinant” DNA or RNA molecule is a DNA or RNA molecule that has been subjected to molecular manipulation in vitro.
  • Non-limiting examples of small molecules include compounds that bind or interact with 58P1D12, ligands including hormones, neuropeptides, chemokines, odorants, phospholipids, and functional equivalents thereof that bind and preferably inhibit 58P1D12 protein function.
  • Such non-limiting small molecules preferably have a molecular weight of less than about 10 kDa, more preferably below about 9, about 8, about 7, about 6, about 5 or about 4 kDa.
  • small molecules physically associate with, or bind, 58P1D12 protein; are not found in naturally occurring metabolic pathways; and/or are more soluble in aqueous than non-aqueous solutions
  • “Stringency” of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization generally depends on the ability of denatured nucleic acid sequences to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature that can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel et al., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995).
  • “Stringent conditions” or “high stringency conditions”, as defined herein, are identified by, but not limited to, those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C.; or (3) employ 50% formamide, 5 ⁇ SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5 ⁇ Denhardt's solution, sonicated salmon sperm DNA (50 ⁇ g/ml), 0.1% SDS, and
  • Modely stringent conditions are described by, but not limited to, those in Sambrook et al., Molecular Cloning: A Laboratory Manual, New York: Cold Spring Harbor Press, 1989, and include the use of washing solution and hybridization conditions (e.g., temperature, ionic strength and % SDS) less stringent than those described above.
  • washing solution and hybridization conditions e.g., temperature, ionic strength and % SDS
  • An example of moderately stringent conditions is overnight incubation at 37° C.
  • HLA “supermotif” is a peptide binding specificity shared by HLA molecules encoded by two or more HLA alleles. Overall phenotypic frequencies of HLA-supertypes in different ethnic populations are set forth in Table IV (F). The non-limiting constituents of various supetypes are as follows:
  • A2 A*0201, A*0202, A*0203, A*0204, A*0205, A*0206, A*6802, A*6901, A*0207
  • A3 A3, A11, A31, A*3301, A*6801, A*0301, A*1101, A*3101
  • B7 B7, B*3501-03, B*51, B*5301, B*5401, B*5501, B*5502, B*5601, B*6701, B*7801, B*0702, B*5101, B*5602
  • B44 B*3701, B*4402, B*4403, B*60 (B*4001), B61 (B*4006)
  • A1 A*0102, A*2604, A*3601, A*4301, A*8001
  • A24 A*24, A*30, A*2403, A*2404, A*3002, A*3003
  • B27 B*1401-02, B*1503, B*1509, B*1510, B*1518, B*3801-02, B*3901, B*3902, B*3903-04, B*4801-02, B*7301, B*2701-08
  • B58 B*1516, B*1517, B*5701, B*5702, B58
  • B62 B*4601, B52, B*1501 (B62), B*1502 (B75), B*1513 (B77)
  • to treat or “therapeutic” and grammatically related terms, refer to any improvement of any consequence of disease, such as prolonged survival, less morbidity, and/or a lessening of side effects which are the byproducts of an alternative therapeutic modality; full eradication of disease is not required.
  • a “transgenic animal” (e.g., a mouse or rat) is an animal having cells that contain a transgene, which transgene was introduced into the animal or an ancestor of the animal at a prenatal, e.g., an embryonic stage.
  • a “transgene” is a DNA that is integrated into the genome of a cell from which a transgenic animal develops.
  • an HLA or cellular immune response “vaccine” is a composition that contains or encodes one or more peptides of the invention.
  • vaccines such as a cocktail of one or more individual peptides; one or more peptides of the invention comprised by a polyepitopic peptide; or nucleic acids that encode such individual peptides or polypeptides, e.g., a minigene that encodes a polyepitopic peptide.
  • the “one or more peptides” can include any whole unit integer from 1-150 or more, e.g., at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, or 150 or more peptides of the invention.
  • the peptides or polypeptides can optionally be modified, such as by lipidation, addition of targeting or other sequences.
  • HLA class I peptides of the invention can be admixed with, or linked to, HLA class II peptides, to facilitate activation of both cytotoxic T lymphocytes and helper T lymphocytes.
  • HLA vaccines can also comprise peptide-pulsed antigen presenting cells, e.g., dendritic cells.
  • variant refers to a molecule that exhibits a variation from a described type or norm, such as a protein that has one or more different amino acid residues in the corresponding position(s) of a specifically described protein (e.g. the 58P1D12 protein shown in FIG. 2 or FIG. 3 .
  • An analog is an example of a variant protein.
  • Splice isoforms and single nucleotides polymorphisms (SNPs) are further examples of variants.
  • the “58P1D12-related proteins” of the invention include those specifically identified herein, as well as allelic variants, conservative substitution variants, analogs and homologs that can be isolated/generated and characterized without undue experimentation following the methods outlined herein or readily available in the art. Fusion proteins that combine parts of different 58P1D12 proteins or fragments thereof, as well as fusion proteins of a 58P1D12 protein and a heterologous polypeptide are also included. Such 58P1D12 proteins are collectively referred to as the 58P1D12-related proteins, the proteins of the invention, or 58P1D12.
  • 58P1D12-related protein refers to a polypeptide fragment or a 58P1D12 protein sequence of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or more than 25 amino acids; or, at least 30, 35, 40, 45, 50, 55, 60, 65, 70, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, or 576 or more amino acids.
  • One aspect of the invention provides polynucleotides corresponding or complementary to all or part of a 58P1D12 gene, mRNA, and/or coding sequence, preferably in isolated form, including polynucleotides encoding a 58P1D12-related protein and fragments thereof, DNA, RNA, DNA/RNA hybrid, and related molecules, polynucleotides or oligonucleotides complementary to a 58P1D12 gene or mRNA sequence or a part thereof, and polynucleotides or oligonucleotides that hybridize to a 58P1D12 gene, mRNA, or to a 58P1D12 encoding polynucleotide (collectively, “58P1D12 polynucleotides”).
  • T can also be U in FIG. 2 .
  • Embodiments of a 58P1D12 polynucleotide include: a 58P1D12 polynucleotide having the sequence shown in FIG. 2 , the nucleotide sequence of 58P1D12 as shown in FIG. 2 wherein T is U; at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in FIG. 2 ; or, at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in FIG. 2 where T is U.
  • embodiments of 58P1D12 nucleotides comprise, without limitation:
  • V a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2D , from nucleotide residue number 206 through nucleotide residue number 916, including the stop codon, wherein T can also be U;
  • VI a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2E , from nucleotide residue number 106 through nucleotide residue number 816, including the stop codon, wherein T can also be U;
  • VII a polynucleotide comprising, consisting essentially of, or consisting of the sequence as set forth in FIG. 2F , wherein T can also be U;
  • VIII a polynucleotide that encodes a 58P1D12-related protein that is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% homologous to an entire amino acid sequence shown in FIG. 2A-F ;
  • (IX) a polynucleotide that encodes a 58P1D12-related protein that is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% identical to an entire amino acid sequence shown in FIG. 2A-F ;
  • (X) a polynucleotide that encodes at least one peptide set forth in Tables VIII-XXI and XXII-XLIX;
  • (XI) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3A in any whole number increment up to 273 that includes at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of FIG. 5 ;
  • (XII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3A in any whole number increment up to 273 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of FIG. 6 ;
  • (XIII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3A in any whole number increment up to 273 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of FIG. 7 ;
  • XIV a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3A in any whole number increment up to 273 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of FIG. 8 ;
  • (XV) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3A in any whole number increment up to 273 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of FIG. 9 ;
  • XVI a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3B , and/or 3 C in any whole number increment up to 232 and/or 236 respectively that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of FIG. 5 ;
  • XVII a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3B and/or 3 C in any whole number increment up to 232 and/or 236 respectively that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of FIG. 6 ;
  • (XVIII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3B and/or 3 C in any whole number increment up to 232 and/or 236 respectively that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of FIG. 7 ;
  • (XIX) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3B and/or 3 C in any whole number increment up to 232 and/or 236 respectively that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of FIG. 8 ;
  • (XX) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3B and/or 3 C in any whole number increment up to 232 and/or 236 respectively that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of FIG. 9 ;
  • XXI a polynucleotide that is fully complementary to a polynucleotide of any one of (I)-(XX);
  • (XXII) a polynucleotide that is fully complementary to a polynucleotide of any one of (I)-(XXI);
  • (XXIV) a composition comprising a polynucleotide of any of (I)-(XXII) or peptide of (XXIII) together with a pharmaceutical excipient and/or in a human unit dose form;
  • (XXV) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to modulate a cell expressing 58P1D12;
  • (XXVI) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing 58P1D12;
  • (XXVII) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing 58P1D12, said cell from a cancer of a tissue listed in Table I;
  • (XXVIII) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to diagnose, prophylax, prognose, or treat a a cancer;
  • (XXIX) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to diagnose, prophylax, prognose, or treat a a cancer of a tissue listed in Table I; and;
  • (XXX) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to identify or characterize a modulator of a cell expressing 58P1D12.
  • Typical embodiments of the invention disclosed herein include 58P1D12 polynucleotides that encode specific portions of 58P1D12 mRNA sequences (and those which are complementary to such sequences) such as those that encode the proteins and/or fragments thereof, for example: [!!! revise to include up to the appropriate length].
  • representative embodiments of the invention disclosed herein include: polynucleotides and their encoded peptides themselves encoding about amino acid 1 to about amino acid 10 of the 58P1D12 protein shown in FIG. 2 or FIG. 3 , polynucleotides encoding about amino acid 10 to about amino acid 20 of the 58P1D12 protein shown in FIG. 2 or FIG. 3 , polynucleotides encoding about amino acid 20 to about amino acid 30 of the 58P1D12 protein shown in FIG. 2 or FIG. 3 , polynucleotides encoding about amino acid 30 to about amino acid 40 of the 58P1D12 protein shown in FIG. 2 or FIG.
  • polynucleotides encoding about amino acid 40 to about amino acid 50 of the 58P1D12 protein shown in FIG. 2 or FIG. 3 polynucleotides encoding about amino acid 50 to about amino acid 60 of the 58P1D12 protein shown in FIG. 2 or FIG. 3
  • polynucleotides encoding about amino acid 60 to about amino acid 70 of the 58P1D12 protein shown in FIG. 2 or FIG. 3 polynucleotides encoding about amino acid 70 to about amino acid 80 of the 58P1D12 protein shown in FIG. 2 or FIG. 3
  • polynucleotides encoding portions of the amino acid sequence (of about 10 amino acids), of amino acids, 100 through the carboxyl terminal amino acid of the 58P1D12 protein are embodiments of the invention. Wherein it is understood that each particular amino acid position discloses that position plus or minus five amino acid residues.
  • Polynucleotides encoding relatively long portions of a 58P1D12 protein are also within the scope of the invention.
  • polynucleotides encoding from about amino acid 1 (or 20 or 30 or 40 etc.) to about amino acid 20, (or 30, or 40 or 50 etc.) of the 58P1D12 protein “or variant” shown in FIG. 2 or FIG. 3 can be generated by a variety of techniques well known in the art. These polynucleotide fragments can include any portion of the 58P1D12 sequence as shown in FIG. 2 .
  • Additional illustrative embodiments of the invention disclosed herein include 58P1D12 polynucleotide fragments encoding one or more of the biological motifs contained within a 58P1D12 protein “or variant” sequence, including one or more of the motif-bearing subsequences of a 58P1D12 protein “or variant” set forth in Tables VIII-XXI and XXII-XLIX.
  • typical polynucleotide fragments of the invention encode one or more of the regions of 58P1D12 protein or variant that exhibit homology to a known molecule.
  • typical polynucleotide fragments can encode one or more of the 58P1D12 protein or variant N-glycosylation sites, cAMP and cGMP-dependent protein kinase phosphorylation sites, casein kinase II phosphorylation sites or N-myristoylation site and amidation sites.
  • HLA Peptide Tables e.g., variant 1, variant 2, etc.
  • search Peptides listed in Table VII.
  • a unique Search Peptide is used to obtain HLA peptides for a particular variant. The position of each Search Peptide relative to its respective parent molecule is listed in Table VII.
  • a Search Peptide begins at position “X”, one must add the value “X minus 1” to each position in Tables VIII-XXI and Tables XXII-IL to obtain the actual position of the HLA peptides in their parental molecule. For example if a particular Search Peptide begins at position 150 of its parental molecule, one must add 150-1, i.e., 149 to each HLA peptide amino acid position to calculate the position of that amino acid in the parent molecule.
  • the polynucleotides of the preceding paragraphs have a number of different specific uses.
  • the human 58P1D12 gene maps to the chromosomal location set forth in the Example entitled “Chromosomal Mapping of 58P1D12.”
  • polynucleotides that encode different regions of the 58P1D12 proteins are used to characterize cytogenetic abnormalities of this chromosomal locale, such as abnormalities that are identified as being associated with various cancers.
  • cytogenetic abnormalities of this chromosomal locale such as abnormalities that are identified as being associated with various cancers.
  • a variety of chromosomal abnormalities including rearrangements have been identified as frequent cytogenetic abnormalities in a number of different cancers (see e.g.
  • polynucleotides encoding specific regions of the 58P1D12 proteins provide new tools that can be used to delineate, with greater precision than previously possible, cytogenetic abnormalities in the chromosomal region that encodes 58P1D12 that may contribute to the malignant phenotype.
  • these polynucleotides satisfy a need in the art for expanding the sensitivity of chromosomal screening in order to identify more subtle and less common chromosomal abnormalities (see e.g. Evans et al., Am. J. Obstet. Gynecol 171(4): 1055-1057 (1994)).
  • 58P1D12 was shown to be highly expressed in prostate and other cancers
  • 58P1D12 polynucleotides are used in methods assessing the status of 58P1D12 gene products in normal versus cancerous tissues.
  • polynucleotides that encode specific regions of the 58P1D12 proteins are used to assess the presence of perturbations (such as deletions, insertions, point mutations, or alterations resulting in a loss of an antigen etc.) in specific regions of the 58P1D12 gene, such as regions containing one or more motifs.
  • Exemplary assays include both RT-PCR assays as well as single-strand conformation polymorphism (SSCP) analysis (see, e.g., Marrogi et al., J. Cutan. Pathol. 26(8): 369-378 (1999), both of which utilize polynucleotides encoding specific regions of a protein to examine these regions within the protein.
  • SSCP single-strand conformation polymorphism
  • nucleic acid related embodiments of the invention disclosed herein are genomic DNA, cDNAs, ribozymes, and antisense molecules, as well as nucleic acid molecules based on an alternative backbone, or including alternative bases, whether derived from natural sources or synthesized, and include molecules capable of inhibiting the RNA or protein expression of 58P1D12.
  • antisense molecules can be RNAs or other molecules, including peptide nucleic acids (PNAs) or non-nucleic acid molecules such as phosphorothioate derivatives that specifically bind DNA or RNA in a base pair-dependent manner.
  • PNAs peptide nucleic acids
  • non-nucleic acid molecules such as phosphorothioate derivatives that specifically bind DNA or RNA in a base pair-dependent manner.
  • a skilled artisan can readily obtain these classes of nucleic acid molecules using the 58P1D12 polynucleotides and polynucleotide sequences disclosed herein.
  • Antisense technology entails the administration of exogenous oligonucleotides that bind to a target polynucleotide located within the cells.
  • the term “antisense” refers to the fact that such oligonucleotides are complementary to their intracellular targets, e.g., 58P1D12. See for example, Jack Cohen, Oligodeoxynucleotides, Antisense Inhibitors of Gene Expression, CRC Press, 1989; and Synthesis 1:1-5 (1988).
  • the 58P1D12 antisense oligonucleotides of the present invention include derivatives such as S-oligonucleotides (phosphorothioate derivatives or S-oligos, see, Jack Cohen, supra), which exhibit enhanced cancer cell growth inhibitory action.
  • S-oligos are isoelectronic analogs of an oligonucleotide (O-oligo) in which a nonbridging oxygen atom of the phosphate group is replaced by a sulfur atom.
  • the S-oligos of the present invention can be prepared by treatment of the corresponding O-oligos with 3H-1,2-benzodithiol-3-one-1,1-dioxide, which is a sulfur transfer reagent. See, e.g., Iyer, R. P. et al., J. Org. Chem. 55:4693-4698 (1990); and Iyer, R. P. et al., J. Am. Chem. Soc. 112:1253-1254 (1990).
  • Additional 58P1D12 antisense oligonucleotides of the present invention include morpholino antisense oligonucleotides known in the art (see, e.g., Partridge et al., 1996, Antisense & Nucleic Acid Drug Development 6: 169-175).
  • the 58P1D12 antisense oligonucleotides of the present invention typically can be RNA or DNA that is complementary to and stably hybridizes with the first 100 5′ codons or last 100 3′ codons of a 58P1D12 genomic sequence or the corresponding mRNA. Absolute complementarity is not required, although high degrees of complementarity are preferred. Use of an oligonucleotide complementary to this region allows for the selective hybridization to 58P1D12 mRNA and not to mRNA specifying other regulatory subunits of protein kinase.
  • 58P1D12 antisense oligonucleotides of the present invention are 15 to 30-mer fragments of the antisense DNA molecule that have a sequence that hybridizes to 58P1D12 mRNA.
  • 58P1D12 antisense oligonucleotide is a 30-mer oligonucleotide that is complementary to a region in the first 10 5′ codons or last 10 3′ codons of 58P1D12.
  • the antisense molecules are modified to employ ribozymes in the inhibition of 58P1D12 expression, see, e.g., L. A. Couture & D. T. Stinchcomb; Trends Genet. 12: 510-515 (1996).
  • nucleotides of the invention include primers and primer pairs, which allow the specific amplification of polynucleotides of the invention or of any specific parts thereof, and probes that selectively or specifically hybridize to nucleic acid molecules of the invention or to any part thereof.
  • Probes can be labeled with a detectable marker, such as, for example, a radioisotope, fluorescent compound, bioluminescent compound, a chemiluminescent compound, metal chelator or enzyme.
  • a detectable marker such as, for example, a radioisotope, fluorescent compound, bioluminescent compound, a chemiluminescent compound, metal chelator or enzyme.
  • Such probes and primers are used to detect the presence of a 58P1D12 polynucleotide in a sample and as a means for detecting a cell expressing a 58P1D12 protein.
  • probes include polypeptides comprising all or part of the human 58P1D12 cDNA sequence shown in FIG. 2 .
  • primer pairs capable of specifically amplifying 58P1D12 mRNAs are also described in the Examples.
  • a great many different primers and probes can be prepared based on the sequences provided herein and used effectively to amplify and/or detect a 58P1D12 mRNA.
  • the 58P1D12 polynucleotides of the invention are useful for a variety of purposes, including but not limited to their use as probes and primers for the amplification and/or detection of the 58P1D12 gene(s), mRNA(s), or fragments thereof; as reagents for the diagnosis and/or prognosis of prostate cancer and other cancers; as coding sequences capable of directing the expression of 58P1D12 polypeptides; as tools for modulating or inhibiting the expression of the 58P1D12 gene(s) and/or translation of the 58P1D12 transcript(s); and as therapeutic agents.
  • the present invention includes the use of any probe as described herein to identify and isolate a 58P1D12 or 58P1D12 related nucleic acid sequence from a naturally occurring source, such as humans or other mammals, as well as the isolated nucleic acid sequence per se, which would comprise all or most of the sequences found in the probe used.
  • the 58P1D12 cDNA sequences described herein enable the isolation of other polynucleotides encoding 58P1D12 gene product(s), as well as the isolation of polynucleotides encoding 58P1D12 gene product homologs, alternatively spliced isoforms, allelic variants, and mutant forms of a 58P1D12 gene product as well as polynucleotides that encode analogs of 58P1D12-related proteins.
  • Various molecular cloning methods that can be employed to isolate full length cDNAs encoding a 58P1D12 gene are well known (see, for example, Sambrook, J.
  • 58P1D12 gene cDNAs can be identified by probing with a labeled 58P1D12 cDNA or a fragment thereof.
  • a 58P1D12 cDNA e.g., FIG.
  • a 58P1D12 gene itself can be isolated by screening genomic DNA libraries, bacterial artificial chromosome libraries (BACs), yeast artificial chromosome libraries (YACs), and the like, with 58P1D12 DNA probes or primers.
  • BACs bacterial artificial chromosome libraries
  • YACs yeast artificial chromosome libraries
  • the invention also provides recombinant DNA or RNA molecules containing a 58P1D12 polynucleotide, a fragment, analog or homologue thereof, including but not limited to phages, plasmids, phagemids, cosmids, YACs, BACs, as well as various viral and non-viral vectors well known in the art, and cells transformed or transfected with such recombinant DNA or RNA molecules. Methods for generating such molecules are well known (see, for example, Sambrook et al., 1989, supra).
  • the invention further provides a host-vector system comprising a recombinant DNA molecule containing a 58P1D12 polynucleotide, fragment, analog or homologue thereof within a suitable prokaryotic or eukaryotic host cell.
  • suitable eukaryotic host cells include a yeast cell, a plant cell, or an animal cell, such as a mammalian cell or an insect cell (e.g., a baculovirus-infectible cell such as an Sf9 or HighFive cell).
  • suitable mammalian cells include various prostate cancer cell lines such as DU145 and TsuPrl, other transfectable or transducible prostate cancer cell lines, primary cells (PrEC), as well as a number of mammalian cells routinely used for the expression of recombinant proteins (e.g., COS, CHO, 293, 293T cells). More particularly, a polynucleotide comprising the coding sequence of 58P1D12 or a fragment, analog or homolog thereof can be used to generate 58P1D12 proteins or fragments thereof using any number of host-vector systems routinely used and widely known in the art.
  • 58P1D12 A wide range of host-vector systems suitable for the expression of 58P1D12 proteins or fragments thereof are available, see for example, Sambrook et al., 1989, supra; Current Protocols in Molecular Biology, 1995, supra).
  • Preferred vectors for mammalian expression include but are not limited to pcDNA 3.1 myc-His-tag (Invitrogen) and the retroviral vector pSRatkneo (Muller et al., 1991, MCB 11:1785).
  • 58P1D12 can be expressed in several prostate cancer and non-prostate cell lines, including for example 293, 293T, rat-1, NIH 3T3 and TsuPr1.
  • the host-vector systems of the invention are useful for the production of a 58P1D12 protein or fragment thereof. Such host-vector systems can be employed to study the functional properties of 58P1D12 and 58P1D12 mutations or analogs.
  • Recombinant human 58P1D12 protein or an analog or homolog or fragment thereof can be produced by mammalian cells transfected with a construct encoding a 58P1D12-related nucleotide.
  • 293T cells can be transfected with an expression plasmid encoding 58P1D12 or fragment, analog or homolog thereof, a 58P1D12-related protein is expressed in the 293T cells, and the recombinant 58P1D12 protein is isolated using standard purification methods (e.g., affinity purification using anti-58P1D12 antibodies).
  • a 58P1D12 coding sequence is subcloned into the retroviral vector pSR ⁇ MSVtkneo and used to infect various mammalian cell lines, such as NIH 3T3, TsuPr1, 293 and rat-1 in order to establish 58P1D12 expressing cell lines.
  • mammalian cell lines such as NIH 3T3, TsuPr1, 293 and rat-1
  • Various other expression systems well known in the art can also be employed.
  • Expression constructs encoding a leader peptide joined in frame to a 58P1D12 coding sequence can be used for the generation of a secreted form of recombinant 58P1D12 protein.
  • codon preferences for a specific species are calculated, for example, by utilizing codon usage tables available on the INTERNET such as at URL dna.affrc.go.jp/ ⁇ nakamura/codon.html.
  • Additional sequence modifications are known to enhance protein expression in a cellular host. These include elimination of sequences encoding spurious polyadenylation signals, exon/intron splice site signals, transposon-like repeats, and/or other such well-characterized sequences that are deleterious to gene expression.
  • the GC content of the sequence is adjusted to levels average for a given cellular host, as calculated by reference to known genes expressed in the host cell. Where possible, the sequence is modified to avoid predicted hairpin secondary mRNA structures.
  • Other useful modifications include the addition of a translational initiation consensus sequence at the start of the open reading frame, as described in Kozak, Mol. Cell Biol., 9:5073-5080 (1989).
  • 58P1D12-related proteins Another aspect of the present invention provides 58P1D12-related proteins.
  • Specific embodiments of 58P1D12 proteins comprise a polypeptide having all or part of the amino acid sequence of human 58P1D12 as shown in FIG. 2 or FIG. 3 .
  • embodiments of 58P1D12 proteins comprise variant, homolog or analog polypeptides that have alterations in the amino acid sequence of 58P1D12 shown in FIG. 2 or FIG. 3 .
  • Embodiments of a 58P1D12 polypeptide include: a 58P1D12 polypeptide having a sequence shown in FIG. 2 , a peptide sequence of a 58P1D12 as shown in FIG. 2 wherein T is U; at least 10 contiguous nucleotides of a polypeptide having the sequence as shown in FIG. 2 ; or, at least 10 contiguous peptides of a polypeptide having the sequence as shown in FIG. 2 where T is U.
  • embodiments of 58P1D12 peptides comprise, without limitation:
  • (I) a protein comprising, consisting essentially of, or consisting of an amino acid sequence as shown in FIG. 2A-F or FIG. 3A-C ;
  • (V) a protein that comprises at least one peptide set forth in Tables VIII-XXI, collectively, which peptide is also set forth in Tables XXII to XLIX, collectively, optionally with a proviso that it is not an entire protein of FIG. 2 ;
  • (VI) a protein that comprises at least two peptides selected from the peptides set forth in Tables VIII-XLIX, optionally with a proviso that it is not an entire protein of FIG. 2 ;
  • VII a protein that comprises at least two peptides selected from the peptides set forth in Tables VIII to XLIX collectively, with a proviso that the protein is not a contiguous sequence from an amino acid sequence of FIG. 2 ;
  • (VIII) a protein that comprises at least one peptide selected from the peptides set forth in Tables VIII-XXI; and at least one peptide selected from the peptides set forth in Tables XXII to XLIX, with a proviso that the protein is not a contiguous sequence from an amino acid sequence of FIG. 2 ;
  • (IX) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of FIG. 3A , 3 B, and/or 3 C in any whole number increment up to 273, 232, and/or 236 respectively that includes at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of FIG. 5 ;
  • (X) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of FIG. 3A , 3 B, and/or 3 C, in any whole number increment up to 273, 236, and/or 232 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of FIG. 6 ;
  • (XI) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of FIG. 3A , 3 B and/or 3 C, in any whole number increment up to 273, 232 and/or 236 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of FIG. 7 ;
  • (XII) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of FIG. 3A , 3 B and/or 3 C, in any whole number increment up to 273, 232 and/or 236 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of FIG. 8 ;
  • (XIII) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, amino acids of a protein of FIG. 3A , 3 B, and/or 3 C in any whole number increment up to 272, 232 and/or 236 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of FIG. 9 ;
  • XXI a peptide that occurs at least twice in Tables VIII-XXI, and at least twice in tables XXII to XLIX;
  • XXII a peptide which comprises one two, three, four, or five of the following characteristics, or an oligonucleotide encoding such peptide:
  • a region of at least 5 amino acids of a particular peptide of FIG. 3 in any whole number increment up to the full length of that protein in FIG. 3 , that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Hydrophilicity profile of FIG. 5 ;
  • a region of at least 5 amino acids of a particular peptide of FIG. 3 in any whole number increment up to the full length of that protein in FIG. 3 , that includes an amino acid position having a value equal to or less than 0.5, 0.4, 0.3, 0.2, 0.1, or having a value equal to 0.0, in the Hydropathicity profile of FIG. 6 ;
  • a region of at least 5 amino acids of a particular peptide of FIG. 3 in any whole number increment up to the full length of that protein in FIG. 3 , that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Percent Accessible Residues profile of FIG. 7 ;
  • a region of at least 5 amino acids of a particular peptide of FIG. 3 in any whole number increment up to the full length of that protein in FIG. 3 , that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Average Flexibility profile of FIG. 8 ; or,
  • (XXIII) a composition comprising a peptide of (I)-(XXII) or an antibody or binding region thereof together with a pharmaceutical excipient and/or in a human unit dose form;
  • (XXIV) a method of using a peptide of (I)-(XXII), or an antibody or binding region thereof or a composition of (XXIII) in a method to modulate a cell expressing 58P1D12;
  • (XXV) a method of using a peptide of (I)-(XXII) or an antibody or binding region thereof or a composition of (XXIII) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing 58P1D12;
  • (XXVI) a method of using a peptide of (I)-(XXII) or an antibody or binding region thereof or a composition (XXIII) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing 58P1D12, said cell from a cancer of a tissue listed in Table I;
  • (XXVII) a method of using a peptide of (I)-(XXII) or an antibody or binding region thereof or a composition of (XXIII) in a method to diagnose, prophylax, prognose, or treat a cancer;
  • (XXVIII) a method of using a peptide of (I)-(XXII) or an antibody or binding region thereof or a composition of (XXIII) in a method to diagnose, prophylax, prognose, or treat a a cancer of a tissue listed in Table I;
  • (XXIX) a method of using a a peptide of (I)-(XXII) or an antibody or binding region thereof or a composition and;
  • Typical embodiments of the invention disclosed herein include 58P1D12 polynucleotides that encode specific portions of 58P1D12 mRNA sequences (and those which are complementary to such sequences) such as those that encode the proteins and/or fragments thereof, for example:
  • allelic variants of human 58P1D12 share a high degree of structural identity and homology (e.g., 90% or more homology).
  • allelic variants of a 58P1D12 protein contain conservative amino acid substitutions within the 58P1D12 sequences described herein or contain a substitution of an amino acid from a corresponding position in a homologue of 58P1D12.
  • One class of 58P1D12 allelic variants are proteins that share a high degree of homology with at least a small region of a particular 58P1D12 amino acid sequence, but further contain a radical departure from the sequence, such as a non-conservative substitution, truncation, insertion or frame shift.
  • Proteins of the invention can comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 conservative substitutions. Such changes include substituting any of isoleucine (I), valine (V), and leucine (L) for any other of these hydrophobic amino acids; aspartic acid (D) for glutamic acid (E) and vice versa; glutamine (Q) for asparagine (N) and vice versa; and serine (S) for threonine (T) and vice versa.
  • isoleucine I
  • V valine
  • L leucine
  • Such changes include substituting any of isoleucine (I), valine (V), and leucine (L) for any other of these hydrophobic amino acids; aspartic acid (D) for glutamic acid (E) and vice versa; glutamine (Q) for asparagine (N) and vice versa; and serine (S) for threonine (T) and vice versa.
  • substitutions can also be considered conservative, depending on the environment of the particular amino acid and its role in the three-dimensional structure of the protein.
  • G glycine
  • A alanine
  • V valine
  • M Methionine
  • L Lysine
  • K arginine
  • R arginine
  • Embodiments of the invention disclosed herein include a wide variety of art-accepted variants or analogs of 58P1D12 proteins such as polypeptides having amino acid insertions, deletions and substitutions.
  • 58P1D12 variants can be made using methods known in the art such as site-directed mutagenesis, alanine scanning, and PCR mutagenesis. Site-directed mutagenesis (Carter et al., Nucl. Acids Res., 13:4331 (1986); Zoller et al., Nucl.
  • Scanning amino acid analysis can also be employed to identify one or more amino acids along a contiguous sequence that is involved in a specific biological activity such as a protein-protein interaction.
  • preferred scanning amino acids are relatively small, neutral amino acids.
  • amino acids include alanine, glycine, serine, and cysteine.
  • Alanine is typically a preferred scanning amino acid among this group because it eliminates the side-chain beyond the beta-carbon and is less likely to alter the main-chain conformation of the variant. Alanine is also typically preferred because it is the most common amino acid. Further, it is frequently found in both buried and exposed positions (Creighton, The Proteins, (W.H. Freeman & Co., N.Y.); Chothia, J. Mol. Biol., 150:1 (1976)). If alanine substitution does not yield adequate amounts of variant, an isosteric amino acid can be used.
  • 58P1D12 variants, analogs or homologs have the distinguishing attribute of having at least one epitope that is “cross reactive” with a 58P1D12 protein having an amino acid sequence of FIG. 3 .
  • cross reactive means that an antibody or T cell that specifically binds to a 58P1D12 variant also specifically binds to a 58P1D12 protein having an amino acid sequence set forth in FIG. 3 .
  • a polypeptide ceases to be a variant of a protein shown in FIG. 3 , when it no longer contains any epitope capable of being recognized by an antibody or T cell that specifically binds to the starting 58P1D12 protein.
  • 58P1D12-related protein variants share 70%, 75%, 80%, 85% or 90% or more similarity with an amino acid sequence of FIG. 3 , or a fragment thereof.
  • Another specific class of 58P1D12 protein variants or analogs comprises one or more of the 58P1D12 biological motifs described herein or presently known in the art.
  • analogs of 58P1D12 fragments that have altered functional (e.g. immunogenic) properties relative to the starting fragment. It is to be appreciated that motifs now or which become part of the art are to be applied to the nucleic or amino acid sequences of FIG. 2 or FIG. 3 .
  • embodiments of the claimed invention include polypeptides containing less than the full amino acid sequence of a 58P1D12 protein shown in FIG. 2 or FIG. 3 .
  • representative embodiments of the invention comprise peptides/proteins having any 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more contiguous amino acids of a 58P1D12 protein shown in FIG. 2 or FIG. 3 .
  • representative embodiments of the invention disclosed herein include polypeptides consisting of about amino acid 1 to about amino acid 10 of a 58P1D12 protein shown in FIG. 2 or FIG. 3 , polypeptides consisting of about amino acid 10 to about amino acid 20 of a 58P1D12 protein shown in FIG. 2 or FIG. 3 , polypeptides consisting of about amino acid 20 to about amino acid 30 of a 58P1D12 protein shown in FIG. 2 or FIG. 3 , polypeptides consisting of about amino acid 30 to about amino acid 40 of a 58P1D12 protein shown in FIG. 2 or FIG. 3 , polypeptides consisting of about amino acid 40 to about amino acid 50 of a 58P1D12 protein shown in FIG.
  • polypeptides consisting of about amino acid 50 to about amino acid 60 of a 58P1D12 protein shown in FIG. 2 or FIG. 3 polypeptides consisting of about amino acid 60 to about amino acid 70 of a 58P1D12 protein shown in FIG. 2 or FIG. 3
  • polypeptides consisting of about amino acid 70 to about amino acid 80 of a 58P1D12 protein shown in FIG. 2 or FIG. 3 polypeptides consisting of about amino acid 80 to about amino acid 90 of a 58P1D12 protein shown in FIG. 2 or FIG. 3
  • polypeptides consisting of about amino acid 1 (or 20 or 30 or 40 etc.) to about amino acid 20, (or 130, or 140 or 150 etc.) of a 58P1D12 protein shown in FIG. 2 or FIG. 3 are embodiments of the invention. It is to be appreciated that the starting and stopping positions in this paragraph refer to the specified position as well as that position plus or minus 5 residues.
  • 58P1D12-related proteins are generated using standard peptide synthesis technology or using chemical cleavage methods well known in the art. Alternatively, recombinant methods can be used to generate nucleic acid molecules that encode a 58P1D12-related protein. In one embodiment, nucleic acid molecules provide a means to generate defined fragments of a 58P1D12 protein (or variants, homologs or analogs thereof).
  • Additional illustrative embodiments of the invention disclosed herein include 58P1D12 polypeptides comprising the amino acid residues of one or more of the biological motifs contained within a 58P1D12 polypeptide sequence set forth in FIG. 2 or FIG. 3 .
  • motifs are known in the art, and a protein can be evaluated for the presence of such motifs by a number of publicly available Internet sites (see, e.g., URL addresses: pfam.wustl.edu/; searchlauncher.bcm.tmc.edu/seq-search/struc-predict.html; psort.ims.u-tokyo.ac.jp/; cbs.dtu.dk/; ebi.ac.uk/interpro/scan.html; expasy.ch/tools/scnpsitl.html; EpimatrixTM and EpimerTM, Brown University, brown.edu/Research/TB-HIV_Lab/epimatrix/epimatrix.html; and BIMAS, bimas.dcrt.nih.gov/.).
  • Motif bearing subsequences of all 58P1D12 variant proteins are set forth and identified in Tables VIII-XXI and XXII-XLIX.
  • Table V sets forth several frequently occurring motifs based on pfam searches (see URL address pfam.wustl.edu/). The columns of Table V list (1) motif name abbreviation, (2) percent identity found amongst the different member of the motif family, (3) motif name or description and (4) most common function; location information is included if the motif is relevant for location.
  • Polypeptides comprising one or more of the 58P1D12 motifs discussed above are useful in elucidating the specific characteristics of a malignant phenotype in view of the observation that the 58P1D12 motifs discussed above are associated with growth dysregulation and because 58P1D12 is overexpressed in certain cancers (See, e.g., Table I).
  • Casein kinase II, cAMP and camp-dependent protein kinase, and Protein Kinase C are enzymes known to be associated with the development of the malignant phenotype (see e.g.
  • Amidation is another protein modification also associated with cancer and cancer progression (see e.g. Treston et al., J. Natl. Cancer Inst. Monogr. (13): 169-175 (1992)).
  • proteins of the invention comprise one or more of the immunoreactive epitopes identified in accordance with art-accepted methods, such as the peptides set forth in Tables VIII-XXI and XXII-XLIX.
  • CTL epitopes can be determined using specific algorithms to identify peptides within a 58P1D12 protein that are capable of optimally binding to specified HLA alleles (e.g., Table IV; EpimatrixTM and EpimerTM, Brown University, URL brown.edu/Research/TB-HIV_Lab/epimatrix/epimatrix.html; and BIMAS, URL bimas.dcrt nih gov/.)
  • processes for identifying peptides that have sufficient binding affinity for HLA molecules and which are correlated with being immunogenic epitopes are well known in the art, and are carried out without undue experimentation.
  • processes for identifying peptides that are immunogenic epitopes are well known in
  • epitopes in order to modulate immunogenicity. For example, one begins with an epitope that bears a CTL or HTL motif (see, e.g., the HLA Class I and HLA Class II motifs/supermotifs of Table IV).
  • the epitope is analoged by substituting out an amino acid at one of the specified positions, and replacing it with another amino acid specified for that position.
  • residues defined in Table IV one can substitute out a deleterious residue in favor of any other residue, such as a preferred residue; substitute a less-preferred residue with a preferred residue; or substitute an originally-occurring preferred residue with another preferred residue. Substitutions can occur at primary anchor positions or at other positions in a peptide; see, e.g., Table IV.
  • polypeptides comprising combinations of the different motifs set forth in Table VI, and/or, one or more of the predicted CTL epitopes of Tables VIII-XXI and XXII-XLIX, and/or, one or more of the predicted HTL epitopes of Tables XLVI-XLIX, and/or, one or more of the T cell binding motifs known in the art.
  • Preferred embodiments contain no insertions, deletions or substitutions either within the motifs or within the intervening sequences of the polypeptides.
  • embodiments which include a number of either N-terminal and/or C-terminal amino acid residues on either side of these motifs may be desirable (to, for example, include a greater portion of the polypeptide architecture in which the motif is located).
  • the number of N-terminal and/or C-terminal amino acid residues on either side of a motif is between about 1 to about 100 amino acid residues, preferably 5 to about 50 amino acid residues.
  • 58P1D12-related proteins are embodied in many forms, preferably in isolated form.
  • a purified 58P1D12 protein molecule will be substantially free of other proteins or molecules that impair the binding of 58P1D12 to antibody, T cell or other ligand.
  • the nature and degree of isolation and purification will depend on the intended use.
  • Embodiments of a 58P1D12-related proteins include purified 58P1D12-related proteins and functional, soluble 58P1D12-related proteins.
  • a functional, soluble 58P1D12 protein or fragment thereof retains the ability to be bound by antibody, T cell or other ligand.
  • the invention also provides 58P1D12 proteins comprising biologically active fragments of a 58P1D12 amino acid sequence shown in FIG. 2 or FIG. 3 .
  • Such proteins exhibit properties of the starting 58P1D12 protein, such as the ability to elicit the generation of antibodies that specifically bind an epitope associated with the starting 58P1D12 protein; to be bound by such antibodies; to elicit the activation of HTL or CTL; and/or, to be recognized by HTL or CTL that also specifically bind to the starting protein.
  • 58P1D12-related polypeptides that contain particularly interesting structures can be predicted and/or identified using various analytical techniques well known in the art, including, for example, the methods of Chou-Fasman, Garnier-Robson, Kyte-Doolittle, Eisenberg, Karplus-Schultz or Jameson-Wolf analysis, or based on immunogenicity. Fragments that contain such structures are particularly useful in generating subunit-specific anti-58P1D12 antibodies or T cells or in identifying cellular factors that bind to 58P1D12. For example, hydrophilicity profiles can be generated, and immunogenic peptide fragments identified, using the method of Hopp, T. P. and Woods, K. R., 1981, Proc. Natl. Acad.
  • Hydropathicity profiles can be generated, and immunogenic peptide fragments identified, using the method of Kyte, J. and Doolittle, R. F., 1982, J. Mol. Biol. 157:105-132. Percent (%) Accessible Residues profiles can be generated, and immunogenic peptide fragments identified, using the method of Janin J., 1979, Nature 277:491-492. Average Flexibility profiles can be generated, and immunogenic peptide fragments identified, using the method of Bhaskaran R., Ponnuswamy P.K., 1988, Int. J. Pept. Protein Res. 32:242-255. Beta-turn profiles can be generated, and immunogenic peptide fragments identified, using the method of Deleage, G., Roux B., 1987, Protein Engineering 1:289-294.
  • CTL epitopes can be determined using specific algorithms to identify peptides within a 58P1D12 protein that are capable of optimally binding to specified HLA alleles (e.g., by using the SYFPEITHI site at World Wide Web URL syfpeithi.bmi-heidelberg.com/; the listings in Table IV(A)-(E); EpimatrixTM and EpimerTM, Brown University, URL (brown.edu/Research/TB-HIV_Lab/epimatrix/epimatrix.html); and BIMAS, URL bimas.dcrt.nih gov/).
  • peptide epitopes from 58P1D12 that are presented in the context of human MHC Class I molecules, e.g., HLA-A1, A2, A3, A11, A24, B7 and B35 were predicted (see, e.g., Tables VIII-XXI, XXII-XLIX).
  • the complete amino acid sequence of the 58P1D12 protein and relevant portions of other variants i.e., for HLA Class I predictions 9 flanking residues on either side of a point mutation or exon junction, and for HLA Class II predictions 14 flanking residues on either side of a point mutation or exon junction corresponding to that variant, were entered into the HLA Peptide Motif Search algorithm found in the Bioinformatics and Molecular Analysis Section (BIMAS) web site listed above; in addition to the site SYFPEITHI, at URL syfpeithi.bmi-heidelberg.com/.
  • BIMAS Bioinformatics and Molecular Analysis Section
  • HLA peptide motif search algorithm was developed by Dr. Ken Parker based on binding of specific peptide sequences in the groove of HLA Class I molecules, in particular HLA-A2 (see, e.g., Falk et al., Nature 351: 290-6 (1991); Hunt et al., Science 255:1261-3 (1992); Parker et al., J. Immunol. 149:3580-7 (1992); Parker et al., J. Immunol. 152:163-75 (1994)).
  • This algorithm allows location and ranking of 8-mer, 9-mer, and 10-mer peptides from a complete protein sequence for predicted binding to HLA-A2 as well as numerous other HLA Class I molecules.
  • HLA class I binding peptides are 8-, 9-, 10 or 11-mers.
  • the epitopes preferably contain a leucine (L) or methionine (M) at position 2 and a valine (V) or leucine (L) at the C-terminus (see, e.g., Parker et al., J. Immunol. 149:3580-7 (1992)).
  • Selected results of 58P1D12 predicted binding peptides are shown in Tables VIII-XXI and XXII-XLIX herein.
  • Tables VIII-XXI and XXII-XLVII selected candidates, 9-mers and 10-mers, for each family member are shown along with their location, the amino acid sequence of each specific peptide, and an estimated binding score.
  • Tables XLVI-XLIX selected candidates, 15-mers, for each family member are shown along with their location, the amino acid sequence of each specific peptide, and an estimated binding score.
  • the binding score corresponds to the estimated half time of dissociation of complexes containing the peptide at 37° C. at pH 6.5. Peptides with the highest binding score are predicted to be the most tightly bound to HLA Class I on the cell surface for the greatest period of time and thus represent the best immunogenic targets for T-cell recognition.
  • every epitope predicted by the BIMAS site, EpimerTM and EpimatrixTM sites, or specified by the HLA class I or class II motifs available in the art or which become part of the art such as set forth in Table IV (or determined using World Wide Web site URL syfpeithi.bmi-heidelberg.com/, or BIMAS, bimas.dcrt.nih.gov/) are to be “applied” to a 58P1D12 protein in accordance with the invention.
  • “applied” means that a 58P1D12 protein is evaluated, e.g., visually or by computer-based patterns finding methods, as appreciated by those of skill in the relevant art.
  • Every subsequence of a 58P1D12 protein of 8, 9, 10, or 11 amino acid residues that bears an HLA Class I motif, or a subsequence of 9 or more amino acid residues that bear an HLA Class II motif are within the scope of the invention.
  • 58P1D12 can be conveniently expressed in cells (such as 293T cells) transfected with a commercially available expression vector such as a CMV-driven expression vector encoding 58P1D12 with a C-terminal 6 ⁇ His and MYC tag (pcDNA3.1/mycHIS, Invitrogen or Tag5, GenHunter Corporation, Nashville Tenn.).
  • the Tag5 vector provides an IgGK secretion signal that can be used to facilitate the production of a secreted 58P1D12 protein in transfected cells.
  • the secreted HIS-tagged 58P1D12 in the culture media can be purified, e.g., using a nickel column using standard techniques.
  • Modifications of 58P1D12-related proteins such as covalent modifications are included within the scope of this invention.
  • One type of covalent modification includes reacting targeted amino acid residues of a 58P1D12 polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C-terminal residues of a 58P1D12 protein.
  • Another type of covalent modification of a 58P1D12 polypeptide included within the scope of this invention comprises altering the native glycosylation pattern of a protein of the invention.
  • Another type of covalent modification of 58P1D12 comprises linking a 58P1D12 polypeptide to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol (PEG), polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Pat. Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192 or 4,179,337.
  • PEG polyethylene glycol
  • polypropylene glycol polypropylene glycol
  • polyoxyalkylenes polyoxyalkylenes
  • the 58P1D12-related proteins of the present invention can also be modified to form a chimeric molecule comprising 58P1D12 fused to another, heterologous polypeptide or amino acid sequence.
  • a chimeric molecule can be synthesized chemically or recombinantly.
  • a chimeric molecule can have a protein of the invention fused to another tumor-associated antigen or fragment thereof.
  • a protein in accordance with the invention can comprise a fusion of fragments of a 58P1D12 sequence (amino or nucleic acid) such that a molecule is created that is not, through its length, directly homologous to the amino or nucleic acid sequences shown in FIG. 2 or FIG. 3 .
  • Such a chimeric molecule can comprise multiples of the same subsequence of 58P1D12.
  • a chimeric molecule can comprise a fusion of a 58P1D12-related protein with a polyhistidine epitope tag, which provides an epitope to which immobilized nickel can selectively bind, with cytokines or with growth factors.
  • the epitope tag is generally placed at the amino- or carboxyl-terminus of a 58P1D12 protein.
  • the chimeric molecule can comprise a fusion of a 58P1D12-related protein with an immunoglobulin or a particular region of an immunoglobulin.
  • the chimeric molecule also referred to as an “immunoadhesin”
  • a fusion could be to the Fc region of an IgG molecule.
  • the Ig fusions preferably include the substitution of a soluble (transmembrane domain deleted or inactivated) form of a 58P1D12 polypeptide in place of at least one variable region within an Ig molecule.
  • the immunoglobulin fusion includes the hinge, CH2 and CH3, or the hinge, CH1, CH2 and CH3 regions of an IgGI molecule.
  • the proteins of the invention have a number of different specific uses. As 58P1D12 is highly expressed in prostate and other cancers, 58P1D12-related proteins are used in methods that assess the status of 58P1D12 gene products in normal versus cancerous tissues, thereby elucidating the malignant phenotype. Typically, polypeptides from specific regions of a 58P1D12 protein are used to assess the presence of perturbations (such as deletions, insertions, point mutations etc.) in those regions (such as regions containing one or more motifs).
  • perturbations such as deletions, insertions, point mutations etc.
  • Exemplary assays utilize antibodies or T cells targeting 58P1D12-related proteins comprising the amino acid residues of one or more of the biological motifs contained within a 58P1D12 polypeptide sequence in order to evaluate the characteristics of this region in normal versus cancerous tissues or to elicit an immune response to the epitope.
  • 58P1D12-related proteins that contain the amino acid residues of one or more of the biological motifs in a 58P1D12 protein are used to screen for factors that interact with that region of 58P1D12.
  • 58P1D12 protein fragments/subsequences are particularly useful in generating and characterizing domain-specific antibodies (e.g., antibodies recognizing an extracellular or intracellular epitope of a 58P1D12 protein), for identifying agents or cellular factors that bind to 58P1D12 or a particular structural domain thereof, and in various therapeutic and diagnostic contexts, including but not limited to diagnostic assays, cancer vaccines and methods of preparing such vaccines.
  • domain-specific antibodies e.g., antibodies recognizing an extracellular or intracellular epitope of a 58P1D12 protein
  • Proteins encoded by the 58P1D12 genes, or by analogs, homologs or fragments thereof, have a variety of uses, including but not limited to generating antibodies and in methods for identifying ligands and other agents and cellular constituents that bind to a 58P1D12 gene product.
  • Antibodies raised against a 58P1D12 protein or fragment thereof are useful in diagnostic and prognostic assays, and imaging methodologies in the management of human cancers characterized by expression of 58P1D12 protein, such as those listed in Table I. Such antibodies can be expressed intracellularly and used in methods of treating patients with such cancers.
  • 58P1D12-related nucleic acids or proteins are also used in generating HTL or CTL responses.
  • immunological assays useful for the detection of 58P1D12 proteins are used, including but not limited to various types of radioimmunoassays, enzyme-linked immunosorbent assays (ELISA), enzyme-linked immunofluorescent assays (ELIFA), immunocytochemical methods, and the like.
  • Antibodies can be labeled and used as immunological imaging reagents capable of detecting 58P1D12-expressing cells (e.g., in radioscintigraphic imaging methods).
  • 58P1D12 proteins are also particularly useful in generating cancer vaccines, as further described herein.
  • Another aspect of the invention provides antibodies that bind to 58P1D12-related proteins.
  • Preferred antibodies specifically bind to a 58P1D12-related protein and do not bind (or bind weakly) to peptides or proteins that are not 58P1D12-related proteins under physiological conditions.
  • physiological conditions include: 1) phosphate buffered saline; 2) Tris-buffered saline containing 25 mM Tris and 150 mM NaCl; or normal saline (0.9% NaCl); 4) animal serum such as human serum; or, 5) a combination of any of 1) through 4); these reactions preferably taking place at pH 7.5, alternatively in a range of pH 7.0 to 8.0, or alternatively in a range of pH 6.5 to 8.5; also, these reactions taking place at a temperature between 4° C. to 37° C.
  • antibodies that bind 58P1D12 can bind 58P1D12-related proteins such as the homologs or analogs thereof.
  • 58P1D12 antibodies of the invention are particularly useful in cancer (see, e.g., Table I) diagnostic and prognostic assays, and imaging methodologies. Similarly, such antibodies are useful in the treatment, diagnosis, and/or prognosis of other cancers, to the extent 58P1D12 is also expressed or overexpressed in these other cancers. Moreover, intracellularly expressed antibodies (e.g., single chain antibodies) are therapeutically useful in treating cancers in which the expression of 58P1D12 is involved, such as advanced or metastatic prostate cancers.
  • the invention also provides various immunological assays useful for the detection and quantification of 58P1D12 and mutant 58P1D12-related proteins.
  • Such assays can comprise one or more 58P1D12 antibodies capable of recognizing and binding a 58P1D12-related protein, as appropriate.
  • These assays are performed within various immunological assay formats well known in the art, including but not limited to various types of radioimmunoassays, enzyme-linked immunosorbent assays (ELISA), enzyme-linked immunofluorescent assays (ELIFA), and the like.
  • Immunological non-antibody assays of the invention also comprise T cell immunogenicity assays (inhibitory or stimulatory) as well as major histocompatibility complex (MHC) binding assays.
  • T cell immunogenicity assays inhibitory or stimulatory
  • MHC major histocompatibility complex
  • immunological imaging methods capable of detecting prostate cancer and other cancers expressing 58P1D12 are also provided by the invention, including but not limited to radioscintigraphic imaging methods using labeled 58P1D12 antibodies.
  • assays are clinically useful in the detection, monitoring, and prognosis of 58P1D12 expressing cancers such as prostate cancer.
  • 58P1D12 antibodies are also used in methods for purifying a 58P1D12-related protein and for isolating 58P1D12 homologues and related molecules.
  • a method of purifying a 58P1D12-related protein comprises incubating a 58P1D12 antibody, which has been coupled to a solid matrix, with a lysate or other solution containing a 58P1D12-related protein under conditions that permit the 58P1D12 antibody to bind to the 58P1D12-related protein; washing the solid matrix to eliminate impurities; and eluting the 58P1D12-related protein from the coupled antibody.
  • Other uses of 58P1D12 antibodies in accordance with the invention include generating anti-idiotypic antibodies that mimic a 58P1D12 protein.
  • antibodies can be prepared by immunizing a suitable mammalian host using a 58P1D12-related protein, peptide, or fragment, in isolated or immunoconjugated form (Antibodies: A Laboratory Manual, CSH Press, Eds., Harlow, and Lane (1988); Harlow, Antibodies, Cold Spring Harbor Press, NY (1989)).
  • fusion proteins of 58P1D12 can also be used, such as a 58P1D12 GST-fusion protein.
  • a GST fusion protein comprising all or most of the amino acid sequence of FIG. 2 or FIG. 3 is produced, then used as an immunogen to generate appropriate antibodies.
  • a 58P1D12-related protein is synthesized and used as an immunogen.
  • naked DNA immunization techniques known in the art are used (with or without purified 58P1D12-related protein or 58P1D12 expressing cells) to generate an immune response to the encoded immunogen (for review, see Donnelly et al., 1997, Ann. Rev. Immunol. 15: 617-648).
  • the amino acid sequence of a 58P1D12 protein as shown in FIG. 2 or FIG. 3 can be analyzed to select specific regions of the 58P1D12 protein for generating antibodies.
  • hydrophobicity and hydrophilicity analyses of a 58P1D12 amino acid sequence are used to identify hydrophilic regions in the 58P1D12 structure. Regions of a 58P1D12 protein that show immunogenic structure, as well as other regions and domains, can readily be identified using various other methods known in the art, such as Chou-Fasman, Garnier-Robson, Kyte-Doolittle, Eisenberg, Karplus-Schultz or Jameson-Wolf analysis. Hydrophilicity profiles can be generated using the method of Hopp, T. P.
  • Hydropathicity profiles can be generated using the method of Kyte, J. and Doolittle, R.F., 1982, J. Mol. Biol. 157:105-132. Percent (%) Accessible Residues profiles can be generated using the method of Janin J., 1979, Nature 277:491-492. Average Flexibility profiles can be generated using the method of Bhaskaran R., Ponnuswamy P. K., 1988, Int. J. Pept. Protein Res. 32:242-255.
  • Beta-turn profiles can be generated using the method of Deleage, G., Roux B., 1987, Protein Engineering 1:289-294. Thus, each region identified by any of these programs or methods is within the scope of the present invention. Methods for the generation of 58P1D12 antibodies are further illustrated by way of the examples provided herein. Methods for preparing a protein or polypeptide for use as an immunogen are well known in the art. Also well known in the art are methods for preparing immunogenic conjugates of a protein with a carrier, such as BSA, KLH or other carrier protein. In some circumstances, direct conjugation using, for example, carbodiimide reagents are used; in other instances linking reagents such as those supplied by Pierce Chemical Co., Rockford, Ill., are effective.
  • 58P1D12 immunogen is often conducted by injection over a suitable time period and with use of a suitable adjuvant, as is understood in the art.
  • titers of antibodies can be taken to determine adequacy of antibody formation.
  • 58P1D12 monoclonal antibodies can be produced by various means well known in the art.
  • immortalized cell lines that secrete a desired monoclonal antibody are prepared using the standard hybridoma technology of Kohler and Milstein or modifications that immortalize antibody-producing B cells, as is generally known.
  • Immortalized cell lines that secrete the desired antibodies are screened by immunoassay in which the antigen is a 58P1D12-related protein.
  • the cells can be expanded and antibodies produced either from in vitro cultures or from ascites fluid.
  • the antibodies or fragments of the invention can also be produced, by recombinant means. Regions that bind specifically to the desired regions of a 58P1D12 protein can also be produced in the context of chimeric or complementarity-determining region (CDR) grafted antibodies of multiple species origin. Humanized or human 58P1D12 antibodies can also be produced, and are preferred for use in therapeutic contexts.
  • CDR complementarity-determining region
  • Fully human 58P1D12 monoclonal antibodies can be generated using cloning technologies employing large human Ig gene combinatorial libraries (i.e., phage display) (Griffiths and Hoogenboom, Building an in vitro immune system: human antibodies from phage display libraries. In: Protein Engineering of Antibody Molecules for Prophylactic and Therapeutic Applications in Man, Clark, M. (Ed.), Nottingham Academic, pp 45-64 (1993); Burton and Barbas, Human Antibodies from combinatorial libraries. Id., pp 65-82).
  • Fully human 58P1D12 monoclonal antibodies can also be produced using transgenic mice engineered to contain human immunoglobulin gene loci as described in PCT Patent Application WO98/24893, Kucherlapati and Jakobovits et al., published Dec. 3, 1997 (see also, Jakobovits, 1998, Exp. Opin. Invest. Drugs 7(4): 607-614; U.S. Pat. Nos. 6,162,963 issued 19 Dec. 2000; 6,150,584 issued 12 Nov. 2000; and, 6,114,598 issued 5 Sep. 2000). This method avoids the in vitro manipulation required with phage display technology and efficiently produces high affinity authentic human antibodies.
  • Reactivity of 58P1D12 antibodies with a 58P1D12-related protein can be established by a number of well known means, including Western blot, immunoprecipitation, ELISA, and FACS analyses using, as appropriate, 58P1D12-related proteins, 58P1D12-expressing cells or extracts thereof.
  • a 58P1D12 antibody or fragment thereof can be labeled with a detectable marker or conjugated to a second molecule. Suitable detectable markers include, but are not limited to, a radioisotope, a fluorescent compound, a bioluminescent compound, chemiluminescent compound, a metal chelator or an enzyme.
  • bi-specific antibodies specific for two or more 58P1D12 epitopes are generated using methods generally known in the art.
  • Homodimeric antibodies can also be generated by cross-linking techniques known in the art (e.g., Wolff et al., Cancer Res. 53: 2560-2565).
  • compositions of the invention induce a therapeutic or prophylactic immune responses in very broad segments of the world-wide population.
  • immunology-related technology For an understanding of the value and efficacy of compositions of the invention that induce cellular immune responses, a brief review of immunology-related technology is provided.
  • a complex of an HLA molecule and a peptidic antigen acts as the ligand recognized by HLA-restricted T cells (Buus, S. et al., Cell 47:1071, 1986; Babbitt, B. P. et al., Nature 317:359, 1985; Townsend, A. and Bodmer, H., Annu. Rev. Immunol. 7:601, 1989; Germain, R. N., Annu. Rev. Immunol. 11:403, 1993).
  • class I and class II allele-specific HLA binding motifs allows identification of regions within a protein that are correlated with binding to particular HLA antigen(s).
  • candidates for epitope-based vaccines have been identified; such candidates can be further evaluated by HLA-peptide binding assays to determine binding affinity and/or the time period of association of the epitope and its corresponding HLA molecule. Additional confirmatory work can be performed to select, amongst these vaccine candidates, epitopes with preferred characteristics in terms of population coverage, and/or immunogenicity.
  • HLA transgenic mice see, e.g., Wentworth, P. A. et al., J. Immunol. 26:97, 1996; Wentworth, P. A. et al., Int. Immunol. 8:651, 1996; Alexander, J. et al., J. Immunol. 159:4753, 1997).
  • peptides in incomplete Freund's adjuvant are administered subcutaneously to HLA transgenic mice.
  • splenocytes are removed and cultured in vitro in the presence of test peptide for approximately one week.
  • Peptide-specific T cells are detected using, e.g., a 51Cr-release assay involving peptide sensitized target cells and target cells expressing endogenously generated antigen.
  • recall responses are detected by culturing PBL from subjects that have been exposed to the antigen due to disease and thus have generated an immune response “naturally”, or from patients who were vaccinated against the antigen.
  • PBL from subjects are cultured in vitro for 1-2 weeks in the presence of test peptide plus antigen presenting cells (APC) to allow activation of “memory” T cells, as compared to “naive” T cells.
  • APC antigen presenting cells
  • T cell activity is detected using assays including 51Cr release involving peptide-sensitized targets, T cell proliferation, or lymphokine release.
  • Nucleic acids that encode a 58P1D12-related protein can also be used to generate either transgenic animals or “knock out” animals that, in turn, are useful in the development and screening of therapeutically useful reagents.
  • cDNA encoding 58P1D12 can be used to clone genomic DNA that encodes 58P1D12. The cloned genomic sequences can then be used to generate transgenic animals containing cells that express DNA that encode 58P1D12.
  • Methods for generating transgenic animals, particularly animals such as mice or rats have become conventional in the art and are described, for example, in U.S. Pat. Nos. 4,736,866 issued 12 Apr. 1988, and 4,870,009 issued 26 Sep. 1989.
  • particular cells would be targeted for 58P1D12 transgene incorporation with tissue-specific enhancers.
  • Transgenic animals that include a copy of a transgene encoding 58P1D12 can be used to examine the effect of increased expression of DNA that encodes 58P1D12. Such animals can be used as tester animals for reagents thought to confer protection from, for example, pathological conditions associated with its overexpression.
  • an animal is treated with a reagent and a reduced incidence of a pathological condition, compared to untreated animals that bear the transgene, would indicate a potential therapeutic intervention for the pathological condition.
  • non-human homologues of 58P1D12 can be used to construct a 58P1D12 “knock out” animal that has a defective or altered gene encoding 58P1D12 as a result of homologous recombination between the endogenous gene encoding 58P1D12 and altered genomic DNA encoding 58P1D12 introduced into an embryonic cell of the animal.
  • cDNA that encodes 58P1D12 can be used to clone genomic DNA encoding 58P1D12 in accordance with established techniques.
  • a portion of the genomic DNA encoding 58P1D12 can be deleted or replaced with another gene, such as a gene encoding a selectable marker that can be used to monitor integration.
  • flanking DNA typically, several kilobases of unaltered flanking DNA (both at the 5′ and 3′ ends) are included in the vector (see, e.g., Thomas and Capecchi, Cell, 51:503 (1987) for a description of homologous recombination vectors).
  • the vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced DNA has homologously recombined with the endogenous DNA are selected (see, e.g., Li et al., Cell, 69:915 (1992)).
  • the selected cells are then injected into a blastocyst of an animal (e.g., a mouse or rat) to form aggregation chimeras (see, e.g., Bradley, in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E. J. Robertson, ed. (IRL, Oxford, 1987), pp. 113-152).
  • a chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal, and the embryo brought to term to create a “knock out” animal
  • Progeny harboring the homologously recombined DNA in their germ cells can be identified by standard techniques and used to breed animals in which all cells of the animal contain the homologously recombined DNA. Knock out animals can be characterized, for example, for their ability to defend against certain pathological conditions or for their development of pathological conditions due to absence of a 58P1D12 polypeptide.
  • Another aspect of the present invention relates to methods for detecting 58P1D12 polynucleotides and 58P1D12-related proteins, as well as methods for identifying a cell that expresses 58P1D12.
  • the expression profile of 58P1D12 makes it a diagnostic marker for metastasized disease. Accordingly, the status of 58P1D12 gene products provides information useful for predicting a variety of factors including susceptibility to advanced stage disease, rate of progression, and/or tumor aggressiveness.
  • the status of 58P1D12 gene products in patient samples can be analyzed by a variety protocols that are well known in the art including immunohistochemical analysis, the variety of Northern blotting techniques including in situ hybridization, RT-PCR analysis (for example on laser capture micro-dissected samples), Western blot analysis and tissue array analysis.
  • the invention provides assays for the detection of 58P1D12 polynucleotides in a biological sample, such as serum, bone, prostate, and other tissues, urine, semen, cell preparations, and the like.
  • Detectable 58P1D12 polynucleotides include, for example, a 58P1D12 gene or fragment thereof, 58P1D12 mRNA, alternative splice variant 58P1D12 mRNAs, and recombinant DNA or RNA molecules that contain a 58P1D12 polynucleotide.
  • a number of methods for amplifying and/or detecting the presence of 58P1D12 polynucleotides are well known in the art and can be employed in the practice of this aspect of the invention.
  • a method for detecting a 58P1D12 mRNA in a biological sample comprises producing cDNA from the sample by reverse transcription using at least one primer; amplifying the cDNA so produced using a 58P1D12 polynucleotides as sense and antisense primers to amplify 58P1D12 cDNAs therein; and detecting the presence of the amplified 58P1D12 cDNA.
  • the sequence of the amplified 58P1D12 cDNA can be determined.
  • a method of detecting a 58P1D12 gene in a biological sample comprises first isolating genomic DNA from the sample; amplifying the isolated genomic DNA using 58P1D12 polynucleotides as sense and antisense primers; and detecting the presence of the amplified 58P1D12 gene.
  • Any number of appropriate sense and antisense probe combinations can be designed from a 58P1D12 nucleotide sequence (see, e.g., FIG. 2 ) and used for this purpose.
  • the invention also provides assays for detecting the presence of a 58P1D12 protein in a tissue or other biological sample such as serum, semen, bone, prostate, urine, cell preparations, and the like.
  • Methods for detecting a 58P1D12-related protein are also well known and include, for example, immunoprecipitation, immunohistochemical analysis, Western blot analysis, molecular binding assays, ELISA, ELIFA and the like.
  • a method of detecting the presence of a 58P1D12-related protein in a biological sample comprises first contacting the sample with a 58P1D12 antibody, a 58P1D12-reactive fragment thereof, or a recombinant protein containing an antigen-binding region of a 58P1D12 antibody; and then detecting the binding of 58P1D12-related protein in the sample.
  • an assay for identifying a cell that expresses a 58P1D12 gene comprises detecting the presence of 58P1D12 mRNA in the cell.
  • Methods for the detection of particular mRNAs in cells include, for example, hybridization assays using complementary DNA probes (such as in situ hybridization using labeled 58P1D12 riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers specific for 58P1D12, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like).
  • an assay for identifying a cell that expresses a 58P1D12 gene comprises detecting the presence of 58P1D12-related protein in the cell or secreted by the cell.
  • Various methods for the detection of proteins are well known in the art and are employed for the detection of 58P1D12-related proteins and cells that express 58P1D12-related proteins.
  • 58P1D12 expression analysis is also useful as a tool for identifying and evaluating agents that modulate 58P1D12 gene expression.
  • 58P1D12 expression is significantly upregulated in prostate cancer, and is expressed in cancers of the tissues listed in Table I.
  • Identification of a molecule or biological agent that inhibits 58P1D12 expression or over-expression in cancer cells is of therapeutic value.
  • such an agent can be identified by using a screen that quantifies 58P1D12 expression by RT-PCR, nucleic acid hybridization or antibody binding.
  • Oncogenesis is known to be a multistep process where cellular growth becomes progressively dysregulated and cells progress from a normal physiological state to precancerous and then cancerous states (see, e.g., Alers et al., Lab Invest. 77(5): 437-438 (1997) and Isaacs et al., Cancer Surv. 23: 19-32 (1995)).
  • examining a biological sample for evidence of dysregulated cell growth allows for early detection of such aberrant physiology, before a pathologic state such as cancer has progressed to a stage that therapeutic options are more limited and or the prognosis is worse.
  • the status of 58P1D12 in a biological sample of interest can be compared, for example, to the status of 58P1D12 in a corresponding normal sample (e.g. a sample from that individual or alternatively another individual that is not affected by a pathology).
  • a corresponding normal sample e.g. a sample from that individual or alternatively another individual that is not affected by a pathology.
  • An alteration in the status of 58P1D12 in the biological sample provides evidence of dysregulated cellular growth.
  • a predetermined normative value such as a predetermined normal level of mRNA expression (see, e.g., Greyer et al., J. Comp. Neurol. 1996 Dec. 9; 376(2): 306-14 and U.S. Pat. No. 5,837,501) to compare 58P1D12 status in a sample.
  • status in this context is used according to its art accepted meaning and refers to the condition or state of a gene and its products.
  • skilled artisans use a number of parameters to evaluate the condition or state of a gene and its products. These include, but are not limited to the location of expressed gene products (including the location of 58P1D12 expressing cells) as well as the level, and biological activity of expressed gene products (such as 58P1D12 mRNA, polynucleotides and polypeptides).
  • an alteration in the status of 58P1D12 comprises a change in the location of 58P1D12 and/or 58P1D12 expressing cells and/or an increase in 58P1D12 mRNA and/or protein expression.
  • 58P1D12 status in a sample can be analyzed by a number of means well known in the art, including without limitation, immunohistochemical analysis, in situ hybridization, RT-PCR analysis on laser capture micro-dissected samples, Western blot analysis, and tissue array analysis.
  • Typical protocols for evaluating the status of a 58P1D12 gene and gene products are found, for example in Ausubel et al. eds., 1995, Current Protocols In Molecular Biology, Units 2 (Northern Blotting), 4 (Southern Blotting), 15 (Immunoblotting) and 18 (PCR Analysis).
  • 58P1D12 in a biological sample is evaluated by various methods utilized by skilled artisans including, but not limited to genomic Southern analysis (to examine, for example perturbations in a 58P1D12 gene), Northern analysis and/or PCR analysis of 58P1D12 mRNA (to examine, for example alterations in the polynucleotide sequences or expression levels of 58P1D12 mRNAs), and, Western and/or immunohistochemical analysis (to examine, for example alterations in polypeptide sequences, alterations in polypeptide localization within a sample, alterations in expression levels of 58P1D12 proteins and/or associations of 58P1D12 proteins with polypeptide binding partners).
  • genomic Southern analysis to examine, for example perturbations in a 58P1D12 gene
  • Northern analysis and/or PCR analysis of 58P1D12 mRNA to examine, for example alterations in the polynucleotide sequences or expression levels of 58P1D12 mRNAs
  • Detectable 58P1D12 polynucleotides include, for example, a 58P1D12 gene or fragment thereof, 58P1D12 mRNA, alternative splice variants, 58P1D12 mRNAs, and recombinant DNA or RNA molecules containing a 58P1D12 polynucleotide.
  • the expression profile of 58P1D12 makes it a diagnostic marker for local and/or metastasized disease, and provides information on the growth or oncogenic potential of a biological sample.
  • the status of 58P1D12 provides information useful for predicting susceptibility to particular disease stages, progression, and/or tumor aggressiveness.
  • the invention provides methods and assays for determining 58P1D12 status and diagnosing cancers that express 58P1D12, such as cancers of the tissues listed in Table I.
  • assays that evaluate the levels of 58P1D12 mRNA transcripts or proteins in a biological sample can be used to diagnose a disease associated with 58P1D12 dysregulation, and can provide prognostic information useful in defining appropriate therapeutic options.
  • the expression status of 58P1D12 provides information including the presence, stage and location of dysplastic, precancerous and cancerous cells, predicting susceptibility to various stages of disease, and/or for gauging tumor aggressiveness. Moreover, the expression profile makes it useful as an imaging reagent for metastasized disease. Consequently, an aspect of the invention is directed to the various molecular prognostic and diagnostic methods for examining the status of 58P1D12 in biological samples such as those from individuals suffering from, or suspected of suffering from a pathology characterized by dysregulated cellular growth, such as cancer.
  • the status of 58P1D12 in a biological sample can be examined by a number of well-known procedures in the art.
  • the status of 58P1D12 in a biological sample taken from a specific location in the body can be examined by evaluating the sample for the presence or absence of 58P1D12 expressing cells (e.g. those that express 58P1D12 mRNAs or proteins).
  • This examination can provide evidence of dysregulated cellular growth, for example, when 58P1D12-expressing cells are found in a biological sample that does not normally contain such cells (such as a lymph node), because such alterations in the status of 58P1D12 in a biological sample are often associated with dysregulated cellular growth.
  • one indicator of dysregulated cellular growth is the metastases of cancer cells from an organ of origin (such as the prostate) to a different area of the body (such as a lymph node).
  • evidence of dysregulated cellular growth is important for example because occult lymph node metastases can be detected in a substantial proportion of patients with prostate cancer, and such metastases are associated with known predictors of disease progression (see, e.g., Murphy et al., Prostate 42(4): 315-317 (2000);Su et al., Semin Surg. Oncol. 18(1): 17-28 (2000) and Freeman et al., J Urol 1995 August 154(2 Pt 1):474-8).
  • the invention provides methods for monitoring 58P1D12 gene products by determining the status of 58P1D12 gene products expressed by cells from an individual suspected of having a disease associated with dysregulated cell growth (such as hyperplasia or cancer) and then comparing the status so determined to the status of 58P1D12 gene products in a corresponding normal sample.
  • the presence of aberrant 58P1D12 gene products in the test sample relative to the normal sample provides an indication of the presence of dysregulated cell growth within the cells of the individual.
  • the invention provides assays useful in determining the presence of cancer in an individual, comprising detecting a significant increase in 58P1D12 mRNA or protein expression in a test cell or tissue sample relative to expression levels in the corresponding normal cell or tissue.
  • the presence of 58P1D12 mRNA can, for example, be evaluated in tissues including but not limited to those listed in Table I.
  • the presence of significant 58P1D12 expression in any of these tissues is useful to indicate the emergence, presence and/or severity of a cancer, since the corresponding normal tissues do not express 58P1D12 mRNA or express it at lower levels.
  • 58P1D12 status is determined at the protein level rather than at the nucleic acid level.
  • a method comprises determining the level of 58P1D12 protein expressed by cells in a test tissue sample and comparing the level so determined to the level of 58P1D12 expressed in a corresponding normal sample.
  • the presence of 58P1D12 protein is evaluated, for example, using immunohistochemical methods.
  • 58P1D12 antibodies or binding partners capable of detecting 58P1D12 protein expression are used in a variety of assay formats well known in the art for this purpose.
  • These perturbations can include insertions, deletions, substitutions and the like.
  • Such evaluations are useful because perturbations in the nucleotide and amino acid sequences are observed in a large number of proteins associated with a growth dysregulated phenotype (see, e.g., Marrogi et al., 1999, J. Cutan. Pathol. 26(8):369-378).
  • a mutation in the sequence of 58P1D12 may be indicative of the presence or promotion of a tumor.
  • Such assays therefore have diagnostic and predictive value where a mutation in 58P1D12 indicates a potential loss of function or increase in tumor growth.
  • nucleotide and amino acid sequences are well known in the art. For example, the size and structure of nucleic acid or amino acid sequences of 58P1D12 gene products are observed by the Northern, Southern, Western, PCR and DNA sequencing protocols discussed herein.
  • other methods for observing perturbations in nucleotide and amino acid sequences such as single strand conformation polymorphism analysis are well known in the art (see, e.g., U.S. Pat. Nos. 5,382,510 issued 7 Sep. 1999, and 5,952,170 issued 17 Jan. 1995).
  • a 58P1D12 gene Aberrant demethylation and/or hypermethylation of CpG islands in gene 5′ regulatory regions frequently occurs in immortalized and transformed cells, and can result in altered expression of various genes.
  • promoter hypermethylation of the pi-class glutathione S-transferase (a protein expressed in normal prostate but not expressed in >90% of prostate carcinomas) appears to permanently silence transcription of this gene and is the most frequently detected genomic alteration in prostate carcinomas (De Marzo et al., Am. J. Pathol. 155(6): 1985-1992 (1999)).
  • methylation-sensitive restriction enzymes that cannot cleave sequences that contain methylated CpG sites to assess the methylation status of CpG islands.
  • MSP methylation specific PCR
  • MSP methylation specific PCR
  • This procedure involves initial modification of DNA by sodium bisulfite (which will convert all unmethylated cytosines to uracil) followed by amplification using primers specific for methylated versus unmethylated DNA. Protocols involving methylation interference can also be found for example in Current Protocols In Molecular Biology, Unit 12, Frederick M. Ausubel et al. eds., 1995.
  • Gene amplification is an additional method for assessing the status of 58P1D12.
  • Gene amplification is measured in a sample directly, for example, by conventional Southern blotting or Northern blotting to quantitate the transcription of mRNA (Thomas, 1980, Proc. Natl. Acad. Sci. USA, 77:5201-5205), dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein.
  • antibodies are employed that recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. The antibodies in turn are labeled and the assay carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.
  • Biopsied tissue or peripheral blood can be conveniently assayed for the presence of cancer cells using for example, Northern, dot blot or RT-PCR analysis to detect 58P1D12 expression.
  • the presence of RT-PCR amplifiable 58P1D12 mRNA provides an indication of the presence of cancer.
  • RT-PCR assays are well known in the art.
  • RT-PCR detection assays for tumor cells in peripheral blood are currently being evaluated for use in the diagnosis and management of a number of human solid tumors. In the prostate cancer field, these include RT-PCR assays for the detection of cells expressing PSA and PSM (Verkaik et al., 1997, Urol. Res. 25:373-384; Ghossein et al., 1995, J. Clin. Oncol. 13:1195-2000; Heston et al., 1995, Clin. Chem. 41:1687-1688).
  • a further aspect of the invention is an assessment of the susceptibility that an individual has for developing cancer.
  • a method for predicting susceptibility to cancer comprises detecting 58P1D12 mRNA or 58P1D12 protein in a tissue sample, its presence indicating susceptibility to cancer, wherein the degree of 58P1D12 mRNA expression correlates to the degree of susceptibility.
  • the presence of 58P1D12 in prostate or other tissue is examined, with the presence of 58P1D12 in the sample providing an indication of prostate cancer susceptibility (or the emergence or existence of a prostate tumor).
  • 58P1D12 nucleotide and amino acid sequences in a biological sample, in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like.
  • the presence of one or more perturbations in 58P1D12 gene products in the sample is an indication of cancer susceptibility (or the emergence or existence of a tumor).
  • the invention also comprises methods for gauging tumor aggressiveness.
  • a method for gauging aggressiveness of a tumor comprises determining the level of 58P1D12 mRNA or 58P1D12 protein expressed by tumor cells, comparing the level so determined to the level of 58P1D12 mRNA or 58P1D12 protein expressed in a corresponding normal tissue taken from the same individual or a normal tissue reference sample, wherein the degree of 58P1D12 mRNA or 58P1D12 protein expression in the tumor sample relative to the normal sample indicates the degree of aggressiveness.
  • aggressiveness of a tumor is evaluated by determining the extent to which 58P1D12 is expressed in the tumor cells, with higher expression levels indicating more aggressive tumors.
  • Another embodiment is the evaluation of the integrity of 58P1D12 nucleotide and amino acid sequences in a biological sample, in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like. The presence of one or more perturbations indicates more aggressive tumors.
  • methods for observing the progression of a malignancy in an individual over time comprise determining the level of 58P1D12 mRNA or 58P1D12 protein expressed by cells in a sample of the tumor, comparing the level so determined to the level of 58P1D12 mRNA or 58P1D12 protein expressed in an equivalent tissue sample taken from the same individual at a different time, wherein the degree of 58P1D12 mRNA or 58P1D12 protein expression in the tumor sample over time provides information on the progression of the cancer.
  • the progression of a cancer is evaluated by determining 58P1D12 expression in the tumor cells over time, where increased expression over time indicates a progression of the cancer. Also, one can evaluate the integrity 58P1D12 nucleotide and amino acid sequences in a biological sample in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like, where the presence of one or more perturbations indicates a progression of the cancer.
  • Another embodiment of the invention is directed to methods for observing a coincidence between the expression of 58P1D12 gene and 58P1D12 gene products (or perturbations in 58P1D12 gene and 58P1D12 gene products) and a factor that is associated with malignancy, as a means for diagnosing and prognosticating the status of a tissue sample.
  • factors associated with malignancy can be utilized, such as the expression of genes associated with malignancy (e.g.
  • Methods for observing a coincidence between the expression of 58P1D12 gene and 58P1D12 gene products (or perturbations in 58P1D12 gene and 58P1D12 gene products) and another factor that is associated with malignancy are useful, for example, because the presence of a set of specific factors that coincide with disease provides information crucial for diagnosing and prognosticating the status of a tissue sample.
  • methods for observing a coincidence between the expression of 58P1D12 gene and 58P1D12 gene products (or perturbations in 58P1D12 gene and 58P1D12 gene products) and another factor associated with malignancy entails detecting the overexpression of 58P1D12 mRNA or protein in a tissue sample, detecting the overexpression of PSA mRNA or protein in a tissue sample (or PSCA or PSM expression), and observing a coincidence of 58P1D12 mRNA or protein and PSA mRNA or protein overexpression (or PSCA or PSM expression).
  • the expression of 58P1D12 and PSA mRNA in prostate tissue is examined, where the coincidence of 58P1D12 and PSA mRNA overexpression in the sample indicates the existence of prostate cancer, prostate cancer susceptibility or the emergence or status of a prostate tumor.
  • Standard methods for the detection and quantification of 58P1D12 mRNA include in situ hybridization using labeled 58P1D12 riboprobes, Northern blot and related techniques using 58P1D12 polynucleotide probes, RT-PCR analysis using primers specific for 58P1D12, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like.
  • semi-quantitative RT-PCR is used to detect and quantify 58P1D12 mRNA expression.
  • primers capable of amplifying 58P1D12 can be used for this purpose, including but not limited to the various primer sets specifically described herein.
  • polyclonal or monoclonal antibodies specifically reactive with the wild-type 58P1D12 protein can be used in an immunohistochemical assay of biopsied tissue.
  • 58P1D12 protein and nucleic acid sequences disclosed herein allow a skilled artisan to identify proteins, small molecules and other agents that interact with 58P1D12, as well as pathways activated by 58P1D12 via any one of a variety of art accepted protocols.
  • one can utilize one of the so-called interaction trap systems also referred to as the “two-hybrid assay”.
  • molecules interact and reconstitute a transcription factor which directs expression of a reporter gene, whereupon the expression of the reporter gene is assayed.
  • Other systems identify protein-protein interactions in vivo through reconstitution of a eukaryotic transcriptional activator, see, e.g., U.S. Pat. Nos. 5,955,280 issued 21 Sep.
  • peptide libraries can be screen peptide libraries to identify molecules that interact with 58P1D12 protein sequences.
  • peptides that bind to 58P1D12 are identified by screening libraries that encode a random or controlled collection of amino acids.
  • Peptides encoded by the libraries are expressed as fusion proteins of bacteriophage coat proteins, the bacteriophage particles are then screened against the 58P1D12 protein(s).
  • peptides having a wide variety of uses are thus identified without any prior information on the structure of the expected ligand or receptor molecule.
  • Typical peptide libraries and screening methods that can be used to identify molecules that interact with 58P1D12 protein sequences are disclosed for example in U.S. Pat. Nos. 5,723,286 issued 3 Mar. 1998 and 5,733,731 issued 31 Mar. 1998.
  • cell lines that express 58P1D12 are used to identify protein-protein interactions mediated by 58P1D12. Such interactions can be examined using immunoprecipitation techniques (see, e.g., Hamilton B. J., et al. Biochem. Biophys. Res. Commun 1999, 261:646-51).
  • 58P1D12 protein can be immunoprecipitated from 58P1D12-expressing cell lines using anti-58P1D12 antibodies.
  • antibodies against His-tag can be used in a cell line engineered to express fusions of 58P1D12 and a His-tag (vectors mentioned above).
  • the immunoprecipitated complex can be examined for protein association by procedures such as Western blotting, 35 S-methionine labeling of proteins, protein microsequencing, silver staining and two-dimensional gel electrophoresis.
  • Small molecules and ligands that interact with 58P1D12 can be identified through related embodiments of such screening assays. For example, small molecules can be identified that interfere with protein function, including molecules that interfere with 58P1D12's ability to mediate phosphorylation and de-phosphorylation, interaction with DNA or RNA molecules as an indication of regulation of cell cycles, second messenger signaling or tumorigenesis.
  • small molecules that modulate 58P1D12-related ion channel, protein pump, or cell communication functions are identified and used to treat patients that have a cancer that expresses 58P1D12 (see, e.g., Hille, B., Ionic Channels of Excitable Membranes 2nd Ed., Sinauer Assoc., Sunderland, Mass., 1992).
  • ligands that regulate 58P1D12 function can be identified based on their ability to bind 58P1D12 and activate a reporter construct. Typical methods are discussed for example in U.S. Pat. No. 5,928,868 issued 27 Jul. 1999, and include methods for forming hybrid ligands in which at least one ligand is a small molecule.
  • cells engineered to express a fusion protein of 58P1D12 and a DNA-binding protein are used to co-express a fusion protein of a hybrid ligand/small molecule and a cDNA library transcriptional activator protein.
  • the cells further contain a reporter gene, the expression of which is conditioned on the proximity of the first and second fusion proteins to each other, an event that occurs only if the hybrid ligand binds to target sites on both hybrid proteins.
  • Those cells that express the reporter gene are selected and the unknown small molecule or the unknown ligand is identified. This method provides a means of identifying modulators, which activate or inhibit 58P1D12.
  • An embodiment of this invention comprises a method of screening for a molecule that interacts with a 58P1D12 amino acid sequence shown in FIG. 2 or FIG. 3 , comprising the steps of contacting a population of molecules with a 58P1D12 amino acid sequence, allowing the population of molecules and the 58P1D12 amino acid sequence to interact under conditions that facilitate an interaction, determining the presence of a molecule that interacts with the 58P1D12 amino acid sequence, and then separating molecules that do not interact with the 58P1D12 amino acid sequence from molecules that do.
  • the method further comprises purifying, characterizing and identifying a molecule that interacts with the 58P1D12 amino acid sequence. The identified molecule can be used to modulate a function performed by 58P1D12.
  • the 58P1D12 amino acid sequence is contacted with a library of peptides.
  • 58P1D12 as a protein that is normally expressed in a restricted set of tissues, but which is also expressed in cancers such as those listed in Table I, opens a number of therapeutic approaches to the treatment of such cancers.
  • targeted antitumor therapies have been useful even when the targeted protein is expressed on normal tissues, even vital normal organ tissues.
  • a vital organ is one that is necessary to sustain life, such as the heart or colon.
  • a non-vital organ is one that can be removed whereupon the individual is still able to survive. Examples of non-vital organs are ovary, breast, and prostate.
  • Herceptin® is an FDA approved pharmaceutical that has as its active ingredient an antibody which is immunoreactive with the protein variously known as HER2, HER2/neu, and erb-b-2. It is marketed by Genentech and has been a commercially successful antitumor agent. Herceptin sales reached almost $400 million in 2002. Herceptin is a treatment for HER2 positive metastatic breast cancer. However, the expression of HER2 is not limited to such tumors. The same protein is expressed in a number of normal tissues. In particular, it is known that HER2/neu is present in normal kidney and heart, thus these tissues are present in all human recipients of Herceptin.
  • HER2/neu in normal kidney is also confirmed by Latif, Z., et al., B.J.U. International (2002) 89:5-9.
  • this article which evaluated whether renal cell carcinoma should be a preferred indication for anti-HER2 antibodies such as Herceptin
  • both protein and mRNA are produced in benign renal tissues.
  • HER2/neu protein was strongly overexpressed in benign renal tissue.
  • Herceptin is a very useful, FDA approved, and commercially successful drug.
  • the effect of Herceptin on cardiac tissue, i.e., “cardiotoxicity,” has merely been a side effect to treatment. When patients were treated with Herceptin alone, significant cardiotoxicity occurred in a very low percentage of patients.
  • kidney tissue is indicated to exhibit normal expression, possibly even higher expression than cardiac tissue, kidney has no appreciable Herceptin side effect whatsoever.
  • kidney tissue is indicated to exhibit normal expression, possibly even higher expression than cardiac tissue, kidney has no appreciable Herceptin side effect whatsoever.
  • cardiac tissue has manifested any appreciable side effect at all.
  • EGFR epidermal growth factor receptor
  • therapeutic approaches that inhibit the activity of a 58P1D12 protein are useful for patients suffering from a cancer that expresses 58P1D12.
  • These therapeutic approaches generally fall into two classes.
  • One class comprises various methods for inhibiting the binding or association of a 58P1D12 protein with its binding partner or with other proteins.
  • Another class comprises a variety of methods for inhibiting the transcription of a 58P1D12 gene or translation of 58P1D12 mRNA.
  • the invention provides cancer vaccines comprising a 58P1D12-related protein or 58P1D12-related nucleic acid.
  • cancer vaccines prevent and/or treat 58P1D12-expressing cancers with minimal or no effects on non-target tissues.
  • the use of a tumor antigen in a vaccine that generates humoral and/or cell-mediated immune responses as anti-cancer therapy is well known in the art and has been employed in prostate cancer using human PSMA and rodent PAP immunogens (Hodge et al., 1995, Int. J. Cancer 63:231-237; Fong et al., 1997, J. Immunol. 159:3113-3117).
  • Such methods can be readily practiced by employing a 58P1D12-related protein, or a 58P1D12-encoding nucleic acid molecule and recombinant vectors capable of expressing and presenting the 58P1D12 immunogen (which typically comprises a number of antibody or T cell epitopes).
  • Skilled artisans understand that a wide variety of vaccine systems for delivery of immunoreactive epitopes are known in the art (see, e.g., Heryln et al., Ann Med 1999 February 31(1):66-78; Maruyama et al., Cancer Immunol Immunother 2000 June 49(3):123-32) Briefly, such methods of generating an immune response (e.g.
  • a mammal's immune system comprises the steps of: exposing the mammal's immune system to an immunoreactive epitope (e.g. an epitope present in a 58P1D12 protein shown in FIG. 3 or analog or homolog thereof) so that the mammal generates an immune response that is specific for that epitope (e.g. generates antibodies that specifically recognize that epitope).
  • an immunoreactive epitope e.g. an epitope present in a 58P1D12 protein shown in FIG. 3 or analog or homolog thereof
  • an immunoreactive epitope e.g. an epitope present in a 58P1D12 protein shown in FIG. 3 or analog or homolog thereof
  • an immunoreactive epitope e.g. an epitope present in a 58P1D12 protein shown in FIG. 3 or analog or homolog thereof
  • an immune response e.g. generates antibodies that specifically recognize that epitope.
  • a 58P1D12 immunogen contains
  • Such vaccine compositions can include, for example, lipopeptides (e.g.,Vitiello, A. et al., J. Clin. Invest. 95:341, 1995), peptide compositions encapsulated in poly(DL-lactide-co-glycolide) (“PLG”) microspheres (see, e.g., Eldridge, et al., Molec. Immunol.
  • lipopeptides e.g.,Vitiello, A. et al., J. Clin. Invest. 95:341, 1995
  • PLG poly(DL-lactide-co-glycolide)
  • Toxin-targeted delivery technologies also known as receptor mediated targeting, such as those of Avant Immunotherapeutics, Inc. (Needham, Mass.) may also be used.
  • the vaccine compositions of the invention can also be used in conjunction with other treatments used for cancer, e.g., surgery, chemotherapy, drug therapies, radiation therapies, etc. including use in combination with immune adjuvants such as IL-2, IL-12, GM-CSF, and the like.
  • CTL epitopes can be determined using specific algorithms to identify peptides within 58P1D12 protein that bind corresponding HLA alleles (see e.g., Table IV; EpimerTM and EpimatrixTM, Brown University (URL brown.edu/Research/TB-HIV_Lab/epimatrix/epimatrix.html); and, BIMAS, (URL bimas.dcrt.nih gov/; SYFPEITHI at URL syfpeithi.bmi-heidelberg.com/).
  • a 58P1D12 immunogen contains one or more amino acid sequences identified using techniques well known in the art, such as the sequences shown in Tables VIII-XXI and XXII-XLIX or a peptide of 8, 9, 10 or 11 amino acids specified by an HLA Class I motif/supermotif (e.g., Table IV (A), Table IV (D), or Table IV (E)) and/or a peptide of at least 9 amino acids that comprises an HLA Class II motif/supermotif (e.g., Table IV (B) or Table IV (C)).
  • HLA Class I motif/supermotif e.g., Table IV (A), Table IV (D), or Table IV (E)
  • HLA Class II motif/supermotif e.g., Table IV (B) or Table IV (C)
  • the HLA Class I binding groove is essentially closed ended so that peptides of only a particular size range can fit into the groove and be bound, generally HLA Class I epitopes are 8, 9, 10, or 11 amino acids long.
  • the HLA Class II binding groove is essentially open ended; therefore a peptide of about 9 or more amino acids can be bound by an HLA Class II molecule. Due to the binding groove differences between HLA Class I and II, HLA Class I motifs are length specific, i.e., position two of a Class I motif is the second amino acid in an amino to carboxyl direction of the peptide.
  • HLA Class II epitopes are often 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acids long, or longer than 25 amino acids.
  • Methods of generating an immune response in a mammal comprise exposing the mammal's immune system to an immunogenic epitope on a protein (e.g. a 58P1D12 protein) so that an immune response is generated.
  • a protein e.g. a 58P1D12 protein
  • a typical embodiment consists of a method for generating an immune response to 58P1D12 in a host, by contacting the host with a sufficient amount of at least one 58P1D12 B cell or cytotoxic T-cell epitope or analog thereof; and at least one periodic interval thereafter re-contacting the host with the 58P1D12 B cell or cytotoxic T-cell epitope or analog thereof.
  • a specific embodiment consists of a method of generating an immune response against a 58P1D12-related protein or a man-made multiepitopic peptide comprising: administering 58P1D12 immunogen (e.g.
  • a 58P1D12 protein or a peptide fragment thereof, a 58P1D12 fusion protein or analog etc. in a vaccine preparation to a human or another mammal.
  • vaccine preparations further contain a suitable adjuvant (see, e.g., U.S. Pat. No. 6,146,635) or a universal helper epitope such as a PADRETMpeptide (Epimmune Inc., San Diego, Calif.; see, e.g., Alexander et al., J. Immunol. 2000 164(3); 164(3): 1625-1633; Alexander et al., Immunity 1994 1(9): 751-761 and Alexander et al., Immunol. Res.
  • An alternative method comprises generating an immune response in an individual against a 58P1D12 immunogen by: administering in vivo to muscle or skin of the individual's body a DNA molecule that comprises a DNA sequence that encodes a 58P1D12 immunogen, the DNA sequence operatively linked to regulatory sequences which control the expression of the DNA sequence; wherein the DNA molecule is taken up by cells, the DNA sequence is expressed in the cells and an immune response is generated against the immunogen (see, e.g., U.S. Pat. No. 5,962,428).
  • a genetic vaccine facilitator such as anionic lipids; saponins; lectins; estrogenic compounds; hydroxylated lower alkyls; dimethyl sulfoxide; and urea is also administered.
  • an antiidiotypic antibody can be administered that mimics 58P1D12, in order to generate a response to the target antigen.
  • Vaccine compositions of the invention include nucleic acid-mediated modalities.
  • DNA or RNA that encode protein(s) of the invention can be administered to a patient.
  • Genetic immunization methods can be employed to generate prophylactic or therapeutic humoral and cellular immune responses directed against cancer cells expressing 58P1D12.
  • Constructs comprising DNA encoding a 58P1D12-related protein/immunogen and appropriate regulatory sequences can be injected directly into muscle or skin of an individual, such that the cells of the muscle or skin take-up the construct and express the encoded 58P1D12 protein/immunogen.
  • a vaccine comprises a 58P1D12-related protein.
  • 58P1D12-related protein immunogen results in the generation of prophylactic or therapeutic humoral and cellular immunity against cells that bear a 58P1D12 protein.
  • Various prophylactic and therapeutic genetic immunization techniques known in the art can be used (for review, see information and references published at Internet address genweb.com). Nucleic acid-based delivery is described, for instance, in Wolff et. al., Science 247:1465 (1990) as well as U.S. Pat. Nos. 5,580,859; 5,589,466; 5,804,566; 5,739,118; 5,736,524; 5,679,647; WO 98/04720.
  • DNA-based delivery technologies include “naked DNA”, facilitated (bupivicaine, polymers, peptide-mediated) delivery, cationic lipid complexes, and particle-mediated (“gene gun”) or pressure-mediated delivery (see, e.g., U.S. Pat. No. 5,922,687).
  • proteins of the invention can be expressed via viral or bacterial vectors.
  • viral gene delivery systems that can be used in the practice of the invention include, but are not limited to, vaccinia, fowlpox, canarypox, adenovirus, influenza, poliovirus, adeno-associated virus, lentivirus, and Sindbis virus (see, e.g., Restifo, 1996, Curr. Opin. Immunol. 8:658-663; Tsang et al. J. Natl. Cancer Inst. 87:982-990 (1995)).
  • Non-viral delivery systems can also be employed by introducing naked DNA encoding a 58P1D12-related protein into the patient (e.g., intramuscularly or intradermally) to induce an anti-tumor response.
  • Vaccinia virus is used, for example, as a vector to express nucleotide sequences that encode the peptides of the invention. Upon introduction into a host, the recombinant vaccinia virus expresses the protein immunogenic peptide, and thereby elicits a host immune response.
  • Vaccinia vectors and methods useful in immunization protocols are described in, e.g., U.S. Pat. No. 4,722,848.
  • Another vector is BCG (Bacille Calmette Guerin). BCG vectors are described in Stover et al., Nature 351:456-460 (1991).
  • BCG vectors are described in Stover et al., Nature 351:456-460 (1991).
  • a wide variety of other vectors useful for therapeutic administration or immunization of the peptides of the invention e.g. adeno and adeno-associated virus vectors, retroviral vectors, Salmonella typhi vectors, detoxified an
  • gene delivery systems are used to deliver a 58P1D12-related nucleic acid molecule.
  • the full-length human 58P1D12 cDNA is employed.
  • 58P1D12 nucleic acid molecules encoding specific cytotoxic T lymphocyte (CTL) and/or antibody epitopes are employed.
  • APCs antigen presenting cells
  • DC dendritic cells
  • DRCs antigen presenting cells
  • DRCs dendritic cells
  • MHC class I and II molecules MHC class I and II molecules
  • B7 co-stimulator B7 co-stimulator
  • IL-12 IL-12
  • PSMA prostate-specific membrane antigen
  • dendritic cells can be used to present 58P1D12 peptides to T cells in the context of MHC class I or II molecules.
  • autologous dendritic cells are pulsed with 58P1D12 peptides capable of binding to MHC class I and/or class II molecules.
  • dendritic cells are pulsed with the complete 58P1D12 protein.
  • Yet another embodiment involves engineering the overexpression of a 58P1D12 gene in dendritic cells using various implementing vectors known in the art, such as adenovirus (Arthur et al., 1997, Cancer Gene Ther. 4:17-25), retrovirus (Henderson et al., 1996, Cancer Res.
  • lentivirus lentivirus
  • adeno-associated virus DNA transfection
  • DNA transfection Ribas et al., 1997, Cancer Res. 57:2865-2869
  • tumor-derived RNA transfection Ashley et al., 1997, J. Exp. Med. 186:1177-1182.
  • Cells that express 58P1D12 can also be engineered to express immune modulators, such as GM-CSF, and used as immunizing agents.
  • immune modulators such as GM-CSF
  • 58P1D12 is an attractive target for antibody-based therapeutic strategies.
  • a number of antibody strategies are known in the art for targeting both extracellular and intracellular molecules (see, e.g., complement and ADCC mediated killing as well as the use of intrabodies).
  • 58P1D12 is expressed by cancer cells of various lineages relative to corresponding normal cells, systemic administration of 58P1D12-immunoreactive compositions are prepared that exhibit excellent sensitivity without toxic, non-specific and/or non-target effects caused by binding of the immunoreactive composition to non-target organs and tissues.
  • Antibodies specifically reactive with domains of 58P1D12 are useful to treat 58P1D12-expressing cancers systemically, either as conjugates with a toxin or therapeutic agent, or as naked antibodies capable of inhibiting cell proliferation or function.
  • 58P1D12 antibodies can be introduced into a patient such that the antibody binds to 58P1D12 and modulates a function, such as an interaction with a binding partner, and consequently mediates destruction of the tumor cells and/or inhibits the growth of the tumor cells.
  • Mechanisms by which such antibodies exert a therapeutic effect can include complement-mediated cytolysis, antibody-dependent cellular cytotoxicity, modulation of the physiological function of 58P1D12, inhibition of ligand binding or signal transduction pathways, modulation of tumor cell differentiation, alteration of tumor angiogenesis factor profiles, and/or apoptosis.
  • antibodies can be used to specifically target and bind immunogenic molecules such as an immunogenic region of a 58P1D12 sequence shown in FIG. 2 or FIG. 3 .
  • skilled artisans understand that it is routine to conjugate antibodies to cytotoxic agents (see, e.g., Slevers et al. Blood 93:11 3678-3684 (Jun. 1, 1999)).
  • cytotoxic agents see, e.g., Slevers et al. Blood 93:11 3678-3684 (Jun. 1, 1999)
  • the cytotoxic agent When cytotoxic and/or therapeutic agents are delivered directly to cells, such as by conjugating them to antibodies specific for a molecule expressed by that cell (e.g. 58P1D12), the cytotoxic agent will exert its known biological effect (i.e. cytotoxicity) on those cells.
  • compositions and methods for using antibody-cytotoxic agent conjugates to kill cells are known in the art.
  • typical methods entail administering to an animal having a tumor a biologically effective amount of a conjugate comprising a selected cytotoxic and/or therapeutic agent linked to a targeting agent (e.g. an anti-58P1D12 antibody) that binds to a marker (e.g. 58P1D12) expressed, accessible to binding or localized on the cell surfaces.
  • a targeting agent e.g. an anti-58P1D12 antibody
  • a marker e.g. 58P1D12
  • a typical embodiment is a method of delivering a cytotoxic and/or therapeutic agent to a cell expressing 58P1D12, comprising conjugating the cytotoxic agent to an antibody that immunospecifically binds to a 58P1D12 epitope, and, exposing the cell to the antibody-agent conjugate.
  • Another illustrative embodiment is a method of treating an individual suspected of suffering from metastasized cancer, comprising a step of administering parenterally to said individual a pharmaceutical composition comprising a therapeutically effective amount of an antibody conjugated to a cytotoxic and/or therapeutic agent.
  • Cancer immunotherapy using anti-58P1D12 antibodies can be done in accordance with various approaches that have been successfully employed in the treatment of other types of cancer, including but not limited to colon cancer (Arlen et al., 1998, Crit. Rev. Immunol. 18:133-138), multiple myeloma (Ozaki et al., 1997, Blood 90:3179-3186, Tsunenari et al., 1997, Blood 90:2437-2444), gastric cancer (Kasprzyk et al., 1992, Cancer Res. 52:2771-2776), B-cell lymphoma (Funakoshi et al., 1996, J. Immunother. Emphasis Tumor Immunol.
  • leukemia Zhong et al., 1996, Leuk. Res. 20:581-589
  • colorectal cancer Moun et al., 1994, Cancer Res. 54:6160-6166; Velders et al., 1995, Cancer Res. 55:4398-4403)
  • breast cancer Shepard et al., 1991, J. Clin. Immunol. 11:117-127.
  • Some therapeutic approaches involve conjugation of naked antibody to a toxin or radioisotope, such as the conjugation of Y91 or I131 to anti-CD20 antibodies (e.g., ZevalinTM, IDEC Pharmaceuticals Corp.
  • paclitaxel Genetech, Inc.
  • the antibodies can be conjugated to a therapeutic agent.
  • 58P1D12 antibodies can be administered in conjunction with radiation, chemotherapy or hormone ablation.
  • antibodies can be conjugated to a toxin such as calicheamicin (e.g., MylotargTM, Wyeth-Ayerst, Madison, N J, a recombinant humanized IgG4 kappa antibody conjugated to antitumor antibiotic calicheamicin) or a maytansinoid (e.g., taxane-based Tumor-Activated Prodrug, TAP, platform, ImmunoGen, Cambridge, Mass., also see e.g., U.S. Pat. No. 5,416,064).
  • calicheamicin e.g., MylotargTM, Wyeth-Ayerst, Madison, N J
  • a maytansinoid e.g., taxane-based Tumor-Activated Prodrug, TAP, platform, ImmunoGen, Cambridge, Mass.
  • antibody therapy can be particularly appropriate in advanced or metastatic cancers.
  • Treatment with the antibody therapy of the invention is indicated for patients who have received one or more rounds of chemotherapy.
  • antibody therapy of the invention is combined with a chemotherapeutic or radiation regimen for patients who have not received chemotherapeutic treatment.
  • antibody therapy can enable the use of reduced dosages of concomitant chemotherapy, particularly for patients who do not tolerate the toxicity of the chemotherapeutic agent very well.
  • Fan et al. (Cancer Res. 53:4637-4642, 1993), Prewett et al. (International J. of Onco. 9:217-224, 1996), and Hancock et al. (Cancer Res. 51:4575-4580, 1991) describe the use of various antibodies together with chemotherapeutic agents.
  • antibody therapy can be particularly appropriate in advanced or metastatic cancers. Treatment with the antibody therapy of the invention is indicated for patients who have received one or more rounds of chemotherapy. Alternatively, antibody therapy of the invention is combined with a chemotherapeutic or radiation regimen for patients who have not received chemotherapeutic treatment. Additionally, antibody therapy can enable the use of reduced dosages of concomitant chemotherapy, particularly for patients who do not tolerate the toxicity of the chemotherapeutic agent very well.
  • Cancer patients can be evaluated for the presence and level of 58P1D12 expression, preferably using immunohistochemical assessments of tumor tissue, quantitative 58P1D12 imaging, or other techniques that reliably indicate the presence and degree of 58P1D12 expression. Immunohistochemical analysis of tumor biopsies or surgical specimens is preferred for this purpose. Methods for immunohistochemical analysis of tumor tissues are well known in the art.
  • Anti-58P1D12 monoclonal antibodies that treat prostate and other cancers include those that initiate a potent immune response against the tumor or those that are directly cytotoxic.
  • anti-58P1D12 monoclonal antibodies mAbs
  • ADCC antibody-dependent cell cytotoxicity
  • anti-58P1D12 mAbs that exert a direct biological effect on tumor growth are useful to treat cancers that express 58P1D12.
  • Mechanisms by which directly cytotoxic mAbs act include: inhibition of cell growth, modulation of cellular differentiation, modulation of tumor angiogenesis factor profiles, and the induction of apoptosis.
  • the mechanism(s) by which a particular anti-58P1D12 mAb exerts an anti-tumor effect is evaluated using any number of in vitro assays that evaluate cell death such as ADCC, ADMMC, complement-mediated cell lysis, and so forth, as is generally known in the art.
  • preferred monoclonal antibodies used in the therapeutic methods of the invention are those that are either fully human or humanized and that bind specifically to the target 58P1D12 antigen with high affinity but exhibit low or no antigenicity in the patient.
  • Therapeutic methods of the invention contemplate the administration of single anti-58P1D12 mAbs as well as combinations, or cocktails, of different mAbs.
  • Such mAb cocktails can have certain advantages inasmuch as they contain mAbs that target different epitopes, exploit different effector mechanisms or combine directly cytotoxic mAbs with mAbs that rely on immune effector functionality. Such mAbs in combination can exhibit synergistic therapeutic effects.
  • anti-58P1D12 mAbs can be administered concomitantly with other therapeutic modalities, including but not limited to various chemotherapeutic agents, androgen-blockers, immune modulators (e.g., IL-2, GM-CSF), surgery or radiation.
  • the anti-58P1D12 mAbs are administered in their “naked” or unconjugated form, or can have a therapeutic agent(s) conjugated to them.
  • Anti-58P1D12 antibody formulations are administered via any route capable of delivering the antibodies to a tumor cell.
  • Routes of administration include, but are not limited to, intravenous, intraperitoneal, intramuscular, intratumor, intradermal, and the like.
  • Treatment generally involves repeated administration of the anti-58P1D12 antibody preparation, via an acceptable route of administration such as intravenous injection (IV), typically at a dose in the range of about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 25 mg/kg body weight.
  • IV intravenous injection
  • doses in the range of 10-1000 mg mAb per week are effective and well tolerated.
  • an initial loading dose of approximately 4 mg/kg patient body weight IV, followed by weekly doses of about 2 mg/kg IV of the anti-58P1D12 mAb preparation represents an acceptable dosing regimen.
  • the initial loading dose is administered as a 90-minute or longer infusion.
  • the periodic maintenance dose is administered as a 30 minute or longer infusion, provided the initial dose was well tolerated.
  • various factors can influence the ideal dose regimen in a particular case.
  • Such factors include, for example, the binding affinity and half life of the Ab or mAbs used, the degree of 58P1D12 expression in the patient, the extent of circulating shed 58P1D12 antigen, the desired steady-state antibody concentration level, frequency of treatment, and the influence of chemotherapeutic or other agents used in combination with the treatment method of the invention, as well as the health status of a particular patient.
  • patients should be evaluated for the levels of 58P1D12 in a given sample (e.g. the levels of circulating 58P1D12 antigen and/or 58P1D12 expressing cells) in order to assist in the determination of the most effective dosing regimen, etc.
  • levels of 58P1D12 in a given sample e.g. the levels of circulating 58P1D12 antigen and/or 58P1D12 expressing cells
  • Such evaluations are also used for monitoring purposes throughout therapy, and are useful to gauge therapeutic success in combination with the evaluation of other parameters (for example, urine cytology and/or ImmunoCyt levels in bladder cancer therapy, or by analogy, serum PSA levels in prostate cancer therapy).
  • Anti-idiotypic anti-58P1D12 antibodies can also be used in anti-cancer therapy as a vaccine for inducing an immune response to cells expressing a 58P1D12-related protein.
  • the generation of anti-idiotypic antibodies is well known in the art; this methodology can readily be adapted to generate anti-idiotypic anti-58P1D12 antibodies that mimic an epitope on a 58P1D12-related protein (see, for example, Wagner et al., 1997, Hybridoma 16: 33-40; Foon et al., 1995, J. Clin. Invest. 96:334-342; Herlyn et al., 1996, Cancer Immunol. Immunother. 43:65-76).
  • Such an anti-idiotypic antibody can be used in cancer vaccine strategies.
  • Vaccines and methods of preparing vaccines that contain an immunogenically effective amount of one or more HLA-binding peptides as described herein are further embodiments of the invention.
  • vaccines in accordance with the invention encompass compositions of one or more of the claimed peptides.
  • a peptide can be present in a vaccine individually.
  • the peptide can exist as a homopolymer comprising multiple copies of the same peptide, or as a heteropolymer of various peptides.
  • Polymers have the advantage of increased immunological reaction and, where different peptide epitopes are used to make up the polymer, the additional ability to induce antibodies and/or CTLs that react with different antigenic determinants of the pathogenic organism or tumor-related peptide targeted for an immune response.
  • the composition can be a naturally occurring region of an antigen or can be prepared, e.g., recombinantly or by chemical synthesis.
  • Carriers that can be used with vaccines of the invention are well known in the art, and include, e.g., thyroglobulin, albumins such as human serum albumin, tetanus toxoid, polyamino acids such as poly 1-lysine, poly 1-glutamic acid, influenza, hepatitis B virus core protein, and the like.
  • the vaccines can contain a physiologically tolerable (i.e., acceptable) diluent such as water, or saline, preferably phosphate buffered saline.
  • the vaccines also typically include an adjuvant.
  • Adjuvants such as incomplete Freund's adjuvant, aluminum phosphate, aluminum hydroxide, or alum are examples of materials well known in the art.
  • CTL responses can be primed by conjugating peptides of the invention to lipids, such as tripalmitoyl-S-glycerylcysteinlyseryl-serine (P3CSS).
  • P3CSS tripalmitoyl-S-glycerylcysteinlyseryl-serine
  • an adjuvant such as a synthetic cytosine-phosphorothiolated-guanine-containing (CpG) oligonucleotides has been found to increase CTL responses 10- to 100-fold. (see, e.g. Davila and Celis, J. Immunol. 165:539-547 (2000)).
  • the immune system of the host Upon immunization with a peptide composition in accordance with the invention, via injection, aerosol, oral, transdermal, transmucosal, intrapleural, intrathecal, or other suitable routes, the immune system of the host responds to the vaccine by producing large amounts of CTLs and/or HTLs specific for the desired antigen. Consequently, the host becomes at least partially immune to later development of cells that express or overexpress 58P1D12 antigen, or derives at least some therapeutic benefit when the antigen was tumor-associated.
  • class I peptide components may be desirable to combine with components that induce or facilitate neutralizing antibody and or helper T cell responses directed to the target antigen.
  • a preferred embodiment of such a composition comprises class I and class II epitopes in accordance with the invention.
  • An alternative embodiment of such a composition comprises a class I and/or class II epitope in accordance with the invention, along with a cross reactive HTL epitope such as PADRETM(Epimmune, San Diego, Calif.) molecule (described e.g., in U.S. Pat. No. 5,736,142).
  • a vaccine of the invention can also include antigen-presenting cells (APC), such as dendritic cells (DC), as a vehicle to present peptides of the invention.
  • APC antigen-presenting cells
  • DC dendritic cells
  • Vaccine compositions can be created in vitro, following dendritic cell mobilization and harvesting, whereby loading of dendritic cells occurs in vitro.
  • dendritic cells are transfected, e.g., with a minigene in accordance with the invention, or are pulsed with peptides.
  • the dendritic cell can then be administered to a patient to elicit immune responses in vivo.
  • Vaccine compositions either DNA- or peptide-based, can also be administered in vivo in combination with dendritic cell mobilization whereby loading of dendritic cells occurs in vivo.
  • the following principles are utilized when selecting an array of epitopes for inclusion in a polyepitopic composition for use in a vaccine, or for selecting discrete epitopes to be included in a vaccine and/or to be encoded by nucleic acids such as a minigene. It is preferred that each of the following principles be balanced in order to make the selection.
  • the multiple epitopes to be incorporated in a given vaccine composition may be, but need not be, contiguous in sequence in the native antigen from which the epitopes are derived.
  • Epitopes are selected which, upon administration, mimic immune responses that have been observed to be correlated with tumor clearance.
  • this includes 3-4 epitopes that come from at least one tumor associated antigen (TAA).
  • TAA tumor associated antigen
  • HLA Class II a similar rationale is employed; again 3-4 epitopes are selected from at least one TAA (see, e.g., Rosenberg et al., Science 278:1447-1450).
  • Epitopes from one TAA may be used in combination with epitopes from one or more additional TAAs to produce a vaccine that targets tumors with varying expression patterns of frequently-expressed TAAs.
  • Epitopes are selected that have the requisite binding affinity established to be correlated with immunogenicity: for HLA Class I an IC50 of 500 nM or less, often 200 nM or less; and for Class II an IC50 of 1000 nM or less.
  • Sufficient supermotif bearing-peptides, or a sufficient array of allele-specific motif-bearing peptides, are selected to give broad population coverage. For example, it is preferable to have at least 80% population coverage.
  • a Monte Carlo analysis a statistical evaluation known in the art, can be employed to assess the breadth, or redundancy of, population coverage.
  • Nested epitopes occur where at least two epitopes overlap in a given peptide sequence.
  • a nested peptide sequence can comprise B cell, HLA class I and/or HLA class II epitopes.
  • a general objective is to provide the greatest number of epitopes per sequence.
  • an aspect is to avoid providing a peptide that is any longer than the amino terminus of the amino terminal epitope and the carboxyl terminus of the carboxyl terminal epitope in the peptide.
  • a multi-epitopic sequence such as a sequence comprising nested epitopes, it is generally important to screen the sequence in order to insure that it does not have pathological or other deleterious biological properties.
  • a polyepitopic protein is created, or when creating a minigene, an objective is to generate the smallest peptide that encompasses the epitopes of interest. This principle is similar, if not the same as that employed when selecting a peptide comprising nested epitopes. However, with an artificial polyepitopic peptide, the size minimization objective is balanced against the need to integrate any spacer sequences between epitopes in the polyepitopic protein.
  • Spacer amino acid residues can, for example, be introduced to avoid junctional epitopes (an epitope recognized by the immune system, not present in the target antigen, and only created by the man-made juxtaposition of epitopes), or to facilitate cleavage between epitopes and thereby enhance epitope presentation.
  • Junctional epitopes are generally to be avoided because the recipient may generate an immune response to that non-native epitope. Of particular concern is a junctional epitope that is a “dominant epitope.” A dominant epitope may lead to such a zealous response that immune responses to other epitopes are diminished or suppressed.
  • potential peptide epitopes can also be selected on the basis of their conservancy.
  • a criterion for conservancy may define that the entire sequence of an HLA class I binding peptide or the entire 9-mer core of a class II binding peptide be conserved in a designated percentage of the sequences evaluated for a specific protein antigen.
  • Nucleic acids encoding the peptides of the invention are a particularly useful embodiment of the invention. Epitopes for inclusion in a minigene are preferably selected according to the guidelines set forth in the previous section.
  • a preferred means of administering nucleic acids encoding the peptides of the invention uses minigene constructs encoding a peptide comprising one or multiple epitopes of the invention.
  • a multi-epitope DNA plasmid encoding supermotif- and/or motif-bearing epitopes derived 58P1D12, the PADRE® universal helper T cell epitope or multiple HTL epitopes from 58P1D12 (see e.g., Tables VIII-XXI and XXII to XLIX), and an endoplasmic reticulum-translocating signal sequence can be engineered.
  • a vaccine may also comprise epitopes that are derived from other TAAs.
  • the immunogenicity of a multi-epitopic minigene can be confirmed in transgenic mice to evaluate the magnitude of CTL induction responses against the epitopes tested. Further, the immunogenicity of DNA-encoded epitopes in vivo can be correlated with the in vitro responses of specific CTL lines against target cells transfected with the DNA plasmid. Thus, these experiments can show that the minigene serves to both: 1.) generate a CTL response and 2.) that the induced CTLs recognized cells expressing the encoded epitopes.
  • the amino acid sequences of the epitopes may be reverse translated.
  • a human codon usage table can be used to guide the codon choice for each amino acid.
  • These epitope-encoding DNA sequences may be directly adjoined, so that when translated, a continuous polypeptide sequence is created.
  • additional elements can be incorporated into the minigene design.
  • amino acid sequences that can be reverse translated and included in the minigene sequence include: HLA class I epitopes, HLA class II epitopes, antibody epitopes, a ubiquitination signal sequence, and/or an endoplasmic reticulum targeting signal.
  • HLA presentation of CTL and HTL epitopes may be improved by including synthetic (e.g. poly-alanine) or naturally-occurring flanking sequences adjacent to the CTL or HTL epitopes; these larger peptides comprising the epitope(s) are within the scope of the invention.
  • the minigene sequence may be converted to DNA by assembling oligonucleotides that encode the plus and minus strands of the minigene. Overlapping oligonucleotides (30-100 bases long) may be synthesized, phosphorylated, purified and annealed under appropriate conditions using well known techniques. The ends of the oligonucleotides can be joined, for example, using T4 DNA ligase. This synthetic minigene, encoding the epitope polypeptide, can then be cloned into a desired expression vector.
  • Standard regulatory sequences well known to those of skill in the art are preferably included in the vector to ensure expression in the target cells.
  • a promoter with a down-stream cloning site for minigene insertion a polyadenylation signal for efficient transcription termination; an E. coli origin of replication; and an E. coli selectable marker (e.g. ampicillin or kanamycin resistance).
  • Numerous promoters can be used for this purpose, e.g., the human cytomegalovirus (hCMV) promoter. See, e.g., U.S. Pat. Nos. 5,580,859 and 5,589,466 for other suitable promoter sequences.
  • introns are required for efficient gene expression, and one or more synthetic or naturally-occurring introns could be incorporated into the transcribed region of the minigene.
  • mRNA stabilization sequences and sequences for replication in mammalian cells may also be considered for increasing minigene expression.
  • the minigene is cloned into the polylinker region downstream of the promoter.
  • This plasmid is transformed into an appropriate E. coli strain, and DNA is prepared using standard techniques. The orientation and DNA sequence of the minigene, as well as all other elements included in the vector, are confirmed using restriction mapping and DNA sequence analysis. Bacterial cells harboring the correct plasmid can be stored as a master cell bank and a working cell bank.
  • immunostimulatory sequences appear to play a role in the immunogenicity of DNA vaccines. These sequences may be included in the vector, outside the minigene coding sequence, if desired to enhance immunogenicity.
  • a bi-cistronic expression vector which allows production of both the minigene-encoded epitopes and a second protein (included to enhance or decrease immunogenicity) can be used.
  • proteins or polypeptides that could beneficially enhance the immune response if co-expressed include cytokines (e.g., IL-2, IL-12, GM-CSF), cytokine-inducing molecules (e.g., LeIF), costimulatory molecules, or for HTL responses, pan-DR binding proteins (PADRETM, Epimmune, San Diego, Calif.).
  • Helper (HTL) epitopes can be joined to intracellular targeting signals and expressed separately from expressed CTL epitopes; this allows direction of the HTL epitopes to a cell compartment different than that of the CTL epitopes. If required, this could facilitate more efficient entry of HTL epitopes into the HLA class II pathway, thereby improving HTL induction.
  • immunosuppressive molecules e.g. TGF- ⁇
  • TGF- ⁇ immunosuppressive molecules
  • Therapeutic quantities of plasmid DNA can be produced for example, by fermentation in E. coli , followed by purification. Aliquots from the working cell bank are used to inoculate growth medium, and grown to saturation in shaker flasks or a bioreactor according to well-known techniques. Plasmid DNA can be purified using standard bioseparation technologies such as solid phase anion-exchange resins supplied by QIAGEN, Inc. (Valencia, Calif.). If required, supercoiled DNA can be isolated from the open circular and linear forms using gel electrophoresis or other methods.
  • Purified plasmid DNA can be prepared for injection using a variety of formulations. The simplest of these is reconstitution of lyophilized DNA in sterile phosphate-buffer saline (PBS). This approach, known as “naked DNA,” is currently being used for intramuscular (IM) administration in clinical trials. To maximize the immunotherapeutic effects of minigene DNA vaccines, an alternative method for formulating purified plasmid DNA may be desirable. A variety of methods have been described, and new techniques may become available.
  • Cationic lipids, glycolipids, and fusogenic liposomes can also be used in the formulation (see, e.g., as described by WO 93/24640; Mannino & Gould-Fogerite, BioTechniques 6(7): 682 (1988); U.S. Pat. No. 5,279,833; WO 91/06309; and Feigner, et al., Proc. Nat'l Acad. Sci. USA 84:7413 (1987).
  • peptides and compounds referred to collectively as protective, interactive, non-condensing compounds could also be complexed to purified plasmid DNA to influence variables such as stability, intramuscular dispersion, or trafficking to specific organs or cell types.
  • Target cell sensitization can be used as a functional assay for expression and HLA class I presentation of minigene-encoded CTL epitopes.
  • the plasmid DNA is introduced into a mammalian cell line that is suitable as a target for standard CTL chromium release assays.
  • the transfection method used will be dependent on the final formulation. Electroporation can be used for “naked” DNA, whereas cationic lipids allow direct in vitro transfection.
  • a plasmid expressing green fluorescent protein (GFP) can be co-transfected to allow enrichment of transfected cells using fluorescence activated cell sorting (FACS).
  • FACS fluorescence activated cell sorting
  • HTL epitopes are then chromium-51 (51Cr) labeled and used as target cells for epitope-specific CTL lines; cytolysis, detected by 51Cr release, indicates both production of, and HLA presentation of, minigene-encoded CTL epitopes. Expression of HTL epitopes may be evaluated in an analogous manner using assays to assess HTL activity.
  • 51Cr chromium-51
  • In vivo immunogenicity is a second approach for functional testing of minigene DNA formulations.
  • Transgenic mice expressing appropriate human HLA proteins are immunized with the DNA product.
  • the dose and route of administration are formulation dependent (e.g., IM for DNA in PBS, intraperitoneal (i.p.) for lipid-complexed DNA).
  • Twenty-one days after immunization splenocytes are harvested and restimulated for one week in the presence of peptides encoding each epitope being tested. Thereafter, for CTL effector cells, assays are conducted for cytolysis of peptide-loaded, 51Cr-labeled target cells using standard techniques.
  • Lysis of target cells that were sensitized by HLA loaded with peptide epitopes, corresponding to minigene-encoded epitopes, demonstrates DNA vaccine function for in vivo induction of CTLs. Immunogenicity of HTL epitopes is confirmed in transgenic mice in an analogous manner.
  • nucleic acids can be administered using ballistic delivery as described, for instance, in U.S. Pat. No. 5,204,253.
  • particles comprised solely of DNA are administered.
  • DNA can be adhered to particles, such as gold particles.
  • Minigenes can also be delivered using other bacterial or viral delivery systems well known in the art, e.g., an expression construct encoding epitopes of the invention can be incorporated into a viral vector such as vaccinia.
  • Vaccine compositions comprising CTL peptides of the invention can be modified, e.g., analoged, to provide desired attributes, such as improved serum half life, broadened population coverage or enhanced immunogenicity.
  • the ability of a peptide to induce CTL activity can be enhanced by linking the peptide to a sequence which contains at least one epitope that is capable of inducing a T helper cell response.
  • a CTL peptide can be directly linked to a T helper peptide, often CTL epitope/HTL epitope conjugates are linked by a spacer molecule.
  • the spacer is typically comprised of relatively small, neutral molecules, such as amino acids or amino acid mimetics, which are substantially uncharged under physiological conditions.
  • the spacers are typically selected from, e.g., Ala, Gly, or other neutral spacers of nonpolar amino acids or neutral polar amino acids.
  • the optionally present spacer need not be comprised of the same residues and thus may be a hetero- or homo-oligomer. When present, the spacer will usually be at least one or two residues, more usually three to six residues and sometimes 10 or more residues.
  • the CTL peptide epitope can be linked to the T helper peptide epitope either directly or via a spacer either at the amino or carboxy terminus of the CTL peptide.
  • the amino terminus of either the immunogenic peptide or the T helper peptide may be acylated.
  • the T helper peptide is one that is recognized by T helper cells present in a majority of a genetically diverse population. This can be accomplished by selecting peptides that bind to many, most, or all of the HLA class II molecules.
  • Examples of such amino acid bind many HLA Class II molecules include sequences from antigens such as tetanus toxoid at positions 830-843 QYIKANSKFIGITE; (SEQ ID NO:18), Plasmodium falciparum circumsporozoite (CS) protein at positions 378-398 DIEKKIAKMEKASSVFNVVNS; (SEQ ID NO:19), and Streptococcus 18 kD protein at positions 116-131 GAVDSILGGVATYGAA; (SEQ ID NO:20).
  • Other examples include peptides bearing a DR 1-4-7 supermotif, or either of the DR3 motifs.
  • An alternative of a pan-DR binding epitope comprises all “L” natural amino acids and can be provided in the form of nucleic acids that encode the epitope.
  • HTL peptide epitopes can also be modified to alter their biological properties. For example, they can be modified to include d-amino acids to increase their resistance to proteases and thus extend their serum half life, or they can be conjugated to other molecules such as lipids, proteins, carbohydrates, and the like to increase their biological activity.
  • a T helper peptide can be conjugated to one or more palmitic acid chains at either the amino or carboxyl termini.
  • compositions of the invention at least one component which primes B lymphocytes or T lymphocytes.
  • Lipids have been identified as agents capable of priming CTL in vivo.
  • palmitic acid residues can be attached to the ⁇ - and ⁇ -amino groups of a lysine residue and then linked, e.g., via one or more linking residues such as Gly, Gly-Gly-, Ser, Ser-Ser, or the like, to an immunogenic peptide.
  • the lipidated peptide can then be administered either directly in a micelle or particle, incorporated into a liposome, or emulsified in an adjuvant, e.g., incomplete Freund's adjuvant.
  • a particularly effective immunogenic composition comprises palmitic acid attached to ⁇ - and ⁇ -amino groups of Lys, which is attached via linkage, e.g., Ser-Ser, to the amino terminus of the immunogenic peptide.
  • E. coli lipoproteins such as tripalmitoyl-5-glycerylcysteinlyseryl-serine (P3CSS) can be used to prime virus specific CTL when covalently attached to an appropriate peptide (see, e.g., Deres, et al., Nature 342:561, 1989).
  • Peptides of the invention can be coupled to P3CSS, for example, and the lipopeptide administered to an individual to prime specifically an immune response to the target antigen.
  • P3CSS tripalmitoyl-5-glycerylcysteinlyseryl-serine
  • two such compositions can be combined to more effectively elicit both humoral and cell-mediated responses.
  • Vaccine Compositions Comprising DC Pulsed with CTL and/or HTL Peptides
  • An embodiment of a vaccine composition in accordance with the invention comprises ex vivo administration of a cocktail of epitope-bearing peptides to PBMC, or isolated DC therefrom, from the patient's blood.
  • a pharmaceutical to facilitate harvesting of DC can be used, such as ProgenipoietinTM (Pharmacia-Monsanto, St. Louis, Mo.) or GM-CSF/IL-4. After pulsing the DC with peptides and prior to reinfusion into patients, the DC are washed to remove unbound peptides.
  • a vaccine comprises peptide-pulsed DCs which present the pulsed peptide epitopes complexed with HLA molecules on their surfaces.
  • the DC can be pulsed ex vivo with a cocktail of peptides, some of which stimulate CTL responses to 58P1D12.
  • a helper T cell (HTL) peptide such as a natural or artificial loosely restricted HLA Class II peptide, can be included to facilitate the CTL response.
  • HTL helper T cell
  • a vaccine in accordance with the invention is used to treat a cancer which expresses or overexpresses 58P1D12.
  • Antigenic 58P1D12-related peptides are used to elicit a CTL and/or HTL response ex vivo, as well.
  • the resulting CTL or HTL cells can be used to treat tumors in patients that do not respond to other conventional forms of therapy, or will not respond to a therapeutic vaccine peptide or nucleic acid in accordance with the invention.
  • Ex vivo CTL or HTL responses to a particular antigen are induced by incubating in tissue culture the patient's, or genetically compatible, CTL or HTL precursor cells together with a source of antigen-presenting cells (APC), such as dendritic cells, and the appropriate immunogenic peptide.
  • APC antigen-presenting cells
  • the cells After an appropriate incubation time (typically about 7-28 days), in which the precursor cells are activated and expanded into effector cells, the cells are infused back into the patient, where they will destroy (CTL) or facilitate destruction (HTL) of their specific target cell (e.g., a tumor cell).
  • CTL destroy
  • HTL facilitate destruction
  • Transfected dendritic cells may also be used as antigen presenting cells.
  • compositions of the invention are typically used to treat and/or prevent a cancer that expresses or overexpresses 58P1D12.
  • peptide and/or nucleic acid compositions are administered to a patient in an amount sufficient to elicit an effective B cell, CTL and/or HTL response to the antigen and to cure or at least partially arrest or slow symptoms and/or complications.
  • An amount adequate to accomplish this is defined as “therapeutically effective dose.” Amounts effective for this use will depend on, e.g., the particular composition administered, the manner of administration, the stage and severity of the disease being treated, the weight and general state of health of the patient, and the judgment of the prescribing physician.
  • the immunogenic peptides of the invention are generally administered to an individual already bearing a tumor that expresses 58P1D12.
  • the peptides or DNA encoding them can be administered individually or as fusions of one or more peptide sequences.
  • Patients can be treated with the immunogenic peptides separately or in conjunction with other treatments, such as surgery, as appropriate.
  • administration should generally begin at the first diagnosis of 58P1D12-associated cancer. This is followed by boosting doses until at least symptoms are substantially abated and for a period thereafter.
  • the embodiment of the vaccine composition i.e., including, but not limited to embodiments such as peptide cocktails, polyepitopic polypeptides, minigenes, or TAA-specific CTLs or pulsed dendritic cells
  • delivered to the patient may vary according to the stage of the disease or the patient's health status. For example, in a patient with a tumor that expresses 58P1D12, a vaccine comprising 58P1D12-specific CTL may be more efficacious in killing tumor cells in patient with advanced disease than alternative embodiments.
  • compositions which stimulate helper T cell responses can also be given in accordance with this embodiment of the invention.
  • the dosage for an initial therapeutic immunization generally occurs in a unit dosage range where the lower value is about 1, 5, 50, 500, or 1,000 ⁇ g and the higher value is about 10,000; 20,000; 30,000; or 50,000 ⁇ g.
  • Dosage values for a human typically range from about 500 ⁇ g to about 50,000 ⁇ g per 70 kilogram patient.
  • Boosting dosages of between about 1.0 p.g to about 50,000 ⁇ g of peptide pursuant to a boosting regimen over weeks to months may be administered depending upon the patient's response and condition as determined by measuring the specific activity of CTL and HTL obtained from the patient's blood. Administration should continue until at least clinical symptoms or laboratory tests indicate that the neoplasia, has been eliminated or reduced and for a period thereafter.
  • the dosages, routes of administration, and dose schedules are adjusted in accordance with methodologies known in the art.
  • the peptides and compositions of the present invention are employed in serious disease states, that is, life-threatening or potentially life threatening situations.
  • life-threatening or potentially life threatening situations in certain embodiments, it is possible and may be felt desirable by the treating physician to administer substantial excesses of these peptide compositions relative to these stated dosage amounts.
  • the vaccine compositions of the invention can also be used purely as prophylactic agents.
  • the dosage for an initial prophylactic immunization generally occurs in a unit dosage range where the lower value is about 1, 5, 50, 500, or 1000 ⁇ g and the higher value is about 10,000; 20,000; 30,000; or 50,000 ⁇ g.
  • Dosage values for a human typically range from about 500 ⁇ g to about 50,000 ⁇ g per 70 kilogram patient. This is followed by boosting dosages of between about 1.0 ⁇ g to about 50,000 ⁇ g of peptide administered at defined intervals from about four weeks to six months after the initial administration of vaccine.
  • the immunogenicity of the vaccine can be assessed by measuring the specific activity of CTL and HTL obtained from a sample of the patient's blood.
  • compositions for therapeutic treatment are intended for parenteral, topical, oral, nasal, intrathecal, or local (e.g. as a cream or topical ointment) administration.
  • the pharmaceutical compositions are administered parentally, e.g., intravenously, subcutaneously, intradermally, or intramuscularly.
  • compositions for parenteral administration which comprise a solution of the immunogenic peptides dissolved or suspended in an acceptable carrier, preferably an aqueous carrier.
  • aqueous carriers may be used, e.g., water, buffered water, 0.8% saline, 0.3% glycine, hyaluronic acid and the like. These compositions may be sterilized by conventional, well-known sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration.
  • compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH-adjusting and buffering agents, tonicity adjusting agents, wetting agents, preservatives, and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.
  • auxiliary substances such as pH-adjusting and buffering agents, tonicity adjusting agents, wetting agents, preservatives, and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.
  • concentration of peptides of the invention in the pharmaceutical formulations can vary widely, i.e., from less than about 0.1%, usually at or at least about 2% to as much as 20% to 50% or more by weight, and will be selected primarily by fluid volumes, viscosities, etc., in accordance with the particular mode of administration selected.
  • a human unit dose form of a composition is typically included in a pharmaceutical composition that comprises a human unit dose of an acceptable carrier, in one embodiment an aqueous carrier, and is administered in a volume/quantity that is known by those of skill in the art to be used for administration of such compositions to humans (see, e.g., Remington's Pharmaceutical Sciences, 17th Edition, A. Gennaro, Editor, Mack Publishing Co., Easton, Pa., 1985).
  • a peptide dose for initial immunization can be from about 1 to about 50,000 ⁇ g, generally 100-5,000 ⁇ g, for a 70 kg patient.
  • an initial immunization may be performed using an expression vector in the form of naked nucleic acid administered IM (or SC or ID) in the amounts of 0.5-5 mg at multiple sites.
  • the nucleic acid (0.1 to 1000 ⁇ g) can also be administered using a gene gun.
  • a booster dose is then administered.
  • the booster can be recombinant fowlpox virus administered at a dose of 5-107 to 5 ⁇ 109 pfu.
  • a treatment generally involves repeated administration of the anti-58P1D12 antibody preparation, via an acceptable route of administration such as intravenous injection (IV), typically at a dose in the range of about 0.1 to about 10 mg/kg body weight.
  • IV intravenous injection
  • doses in the range of 10-500 mg mAb per week are effective and well tolerated.
  • an initial loading dose of approximately 4 mg/kg patient body weight IV, followed by weekly doses of about 2 mg/kg IV of the anti-58P1D12 mAb preparation represents an acceptable dosing regimen.
  • various factors can influence the ideal dose in a particular case.
  • Such factors include, for example, half life of a composition, the binding affinity of an Ab, the immunogenicity of a substance, the degree of 58P1D12 expression in the patient, the extent of circulating shed 58P1D12 antigen, the desired steady-state concentration level, frequency of treatment, and the influence of chemotherapeutic or other agents used in combination with the treatment method of the invention, as well as the health status of a particular patient.
  • Non-limiting preferred human unit doses are, for example, 500 ⁇ g-1 mg, 1 mg-50 mg, 50 mg-100 mg, 100 mg-200 mg, 200 mg-300 mg, 400 mg-500 mg, 500 mg-600 mg, 600 mg-700 mg, 700 mg-800 mg, 800 mg-900 mg, 900 mg-1 g, or 1 mg-700 mg.
  • the dose is in a range of 2-5 mg/kg body weight, e.g., with follow on weekly doses of 1-3 mg/kg; 0.5 mg, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 mg/kg body weight followed, e.g., in two, three or four weeks by weekly doses; 0.5-10 mg/kg body weight, e.g., followed in two, three or four weeks by weekly doses; 225, 250, 275, 300, 325, 350, 375, 400 mg m2 of body area weekly; 1-600 mg m2 of body area weekly; 225-400 mg m2 of body area weekly; these does can be followed by weekly doses for 2, 3, 4, 5, 6, 7, 8, 9, 19, 11, 12 or more weeks.
  • human unit dose forms of polynucleotides comprise a suitable dosage range or effective amount that provides any therapeutic effect.
  • a therapeutic effect depends on a number of factors, including the sequence of the polynucleotide, molecular weight of the polynucleotide and route of administration. Dosages are generally selected by the physician or other health care professional in accordance with a variety of parameters known in the art, such as severity of symptoms, history of the patient and the like.
  • a dosage range may be selected from, for example, an independently selected lower limit such as about 0.1, 0.25, 0.5, 1, 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400 or 500 mg/kg up to an independently selected upper limit, greater than the lower limit, of about 60, 80, 100, 200, 300, 400, 500, 750, 1000, 1500, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000 or 10,000 mg/kg.
  • an independently selected lower limit such as about 0.1, 0.25, 0.5, 1, 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400 or 500 mg/kg up to an independently selected upper limit, greater than the lower limit, of about 60, 80, 100, 200, 300, 400, 500, 750, 1000, 1500, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000 or 10,000 mg/kg.
  • a dose may be about any of the following: 0.1 to 100 mg/kg, 0.1 to 50 mg/kg, 0.1 to 25 mg/kg, 0.1 to 10 mg/kg, 1 to 500 mg/kg, 100 to 400 mg/kg, 200 to 300 mg/kg, 1 to 100 mg/kg, 100 to 200 mg/kg, 300 to 400 mg/kg, 400 to 500 mg/kg, 500 to 1000 mg/kg, 500 to 5000 mg/kg, or 500 to 10,000 mg/kg.
  • parenteral routes of administration may require higher doses of polynucleotide compared to more direct application to the nucleotide to diseased tissue, as do polynucleotides of increasing length.
  • human unit dose forms of T-cells comprise a suitable dosage range or effective amount that provides any therapeutic effect.
  • a therapeutic effect depends on a number of factors. Dosages are generally selected by the physician or other health care professional in accordance with a variety of parameters known in the art, such as severity of symptoms, history of the patient and the like.
  • a dose may be about 104 cells to about 106 cells, about 106 cells to about 108 cells, about 108 to about 1011 cells, or about 108 to about 5 ⁇ 1010 cells.
  • a dose may also about 106 cells/m2 to about 1010 cells/m2, or about 106 cells/m2 to about 108 cells/m2.
  • Proteins(s) of the invention, and/or nucleic acids encoding the protein(s), can also be administered via liposomes, which may also serve to: 1) target the proteins(s) to a particular tissue, such as lymphoid tissue; 2) to target selectively to diseases cells; or, 3) to increase the half-life of the peptide composition.
  • liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like.
  • the peptide to be delivered is incorporated as part of a liposome, alone or in conjunction with a molecule which binds to a receptor prevalent among lymphoid cells, such as monoclonal antibodies which bind to the CD45 antigen, or with other therapeutic or immunogenic compositions.
  • liposomes either filled or decorated with a desired peptide of the invention can be directed to the site of lymphoid cells, where the liposomes then deliver the peptide compositions.
  • Liposomes for use in accordance with the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol.
  • lipids are generally guided by consideration of, e.g., liposome size, acid lability and stability of the liposomes in the blood stream.
  • a variety of methods are available for preparing liposomes, as described in, e.g., Szoka, et al., Ann. Rev. Biophys. Bioeng. 9:467 (1980), and U.S. Pat. Nos. 4,235,871, 4,501,728, 4,837,028, and 5,019,369.
  • a ligand to be incorporated into the liposome can include, e.g., antibodies or fragments thereof specific for cell surface determinants of the desired immune system cells.
  • a liposome suspension containing a peptide may be administered intravenously, locally, topically, etc. in a dose which varies according to, inter alia, the manner of administration, the peptide being delivered, and the stage of the disease being treated.
  • nontoxic solid carriers include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like.
  • a pharmaceutically acceptable nontoxic composition is formed by incorporating any of the normally employed excipients, such as those carriers previously listed, and generally 10-95% of active ingredient, that is, one or more peptides of the invention, and more preferably at a concentration of 25%-75%.
  • immunogenic peptides are preferably supplied in finely divided form along with a surfactant and propellant. Typical percentages of peptides are about 0.01%-20% by weight, preferably about 1%-10%.
  • the surfactant must, of course, be nontoxic, and preferably soluble in the propellant.
  • Representative of such agents are the esters or partial esters of fatty acids containing from about 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride.
  • the surfactant may constitute about 0.1%-20% by weight of the composition, preferably about 0.25-5%.
  • the balance of the composition is ordinarily propellant.
  • a carrier can also be included, as desired, as with, e.g., lecithin for intranasal delivery.
  • 58P1D12 polynucleotides, polypeptides, reactive cytotoxic T cells (CTL), reactive helper T cells (HTL) and anti-polypeptide antibodies are used in well known diagnostic, prognostic and therapeutic assays that examine conditions associated with dysregulated cell growth such as cancer, in particular the cancers listed in Table I (see, e.g., both its specific pattern of tissue expression as well as its overexpression in certain cancers as described for example in the Example entitled “Expression analysis of 58P1D12 in normal tissues, and patient specimens”).
  • 58P1D12 can be analogized to a prostate associated antigen PSA, the archetypal marker that has been used by medical practitioners for years to identify and monitor the presence of prostate cancer (see, e.g., Merrill et al., J. Urol. 163(2): 503-5120 (2000); Polascik et al., J. Urol. August; 162(2):293-306 (1999) and Fortier et al., J. Nat. Cancer Inst. 91(19): 1635-1640 (1999)).
  • PSA prostate associated antigen PSA
  • Typical embodiments of diagnostic methods which utilize the 58P1D12 polynucleotides, polypeptides, reactive T cells and antibodies are analogous to those methods from well-established diagnostic assays, which employ, e.g., PSA polynucleotides, polypeptides, reactive T cells and antibodies.
  • PSA polynucleotides are used as probes (for example in Northern analysis, see, e.g., Sharief et al., Biochem. Mol. Biol. Int. 33(3):567-74 (1994)) and primers (for example in PCR analysis, see, e.g., Okegawa et al., J. Urol.
  • the 58P1D12 polynucleotides described herein can be utilized in the same way to detect 58P1D12 overexpression or the metastasis of prostate and other cancers expressing this gene.
  • PSA polypeptides are used to generate antibodies specific for PSA which can then be used to observe the presence and/or the level of PSA proteins in methods to monitor PSA protein overexpression (see, e.g., Stephan et al., Urology 55(4):560-3 (2000)) or the metastasis of prostate cells (see, e.g., Alanen et al., Pathol. Res. Pract. 192(3):233-7 (1996)), the 58P1D12 polypeptides described herein can be utilized to generate antibodies for use in detecting 58P1D12 overexpression or the metastasis of prostate cells and cells of other cancers expressing this gene.
  • metastases involves the movement of cancer cells from an organ of origin (such as the lung or prostate gland etc.) to a different area of the body (such as a lymph node)
  • assays which examine a biological sample for the presence of cells expressing 58P1D12 polynucleotides and/or polypeptides can be used to provide evidence of metastasis.
  • tissue that does not normally contain 58P1D12-expressing cells lymph node
  • 58P1D12-expressing cells such as the 58P1D12 expression seen in LAPC4 and LAPC9
  • 58P1D12 polynucleotides and/or polypeptides can be used to provide evidence of cancer, for example, when cells in a biological sample that do not normally express 58P1D12 or express 58P1D12 at a different level are found to express 58P1D12 or have an increased expression of 58P1D12 (see, e.g., the 58P1D12 expression in the cancers listed in Table I and in patient samples etc. shown in the accompanying Figures).
  • artisans may further wish to generate supplementary evidence of metastasis by testing the biological sample for the presence of a second tissue restricted marker (in addition to 58P1D12) such as PSA, PSCA etc. (see, e.g., Alanen et al., Pathol. Res. Pract. 192(3): 233-237 (1996)).
  • immunohistochemistry to identify the presence of a 58P1D12 polypeptide within a tissue section can indicate an altered state of certain cells within that tissue. It is well understood in the art that the ability of an antibody to localize to a polypeptide that is expressed in cancer cells is a way of diagnosing presence of disease, disease stage, progression and/or tumor aggressiveness. Such an antibody can also detect an altered distribution of the polypeptide within the cancer cells, as compared to corresponding non-malignant tissue.
  • the 58P1D12 polypeptide and immunogenic compositions are also useful in view of the phenomena of altered subcellular protein localization in disease states. Alteration of cells from normal to diseased state causes changes in cellular morphology and is often associated with changes in subcellular protein localization/distribution. For example, cell membrane proteins that are expressed in a polarized manner in normal cells can be altered in disease, resulting in distribution of the protein in a non-polar manner over the whole cell surface.
  • normal breast epithelium is either negative for Her2 protein or exhibits only a basolateral distribution whereas malignant cells can express the protein over the whole cell surface (De Potter, et al, International Journal of Cancer, 44; 969-974 (1989): McCormick, et al, 117; 935-943 (2002)).
  • distribution of the protein may be altered from a surface only localization to include diffuse cytoplasmic expression in the diseased state. Such an example can be seen with MUC1 (Diaz, et al, The Breast Journal, 7: 40-45 (2001)).
  • the 58P1D12 protein and immune responses related thereto are very useful. Accordingly, the ability to determine whether alteration of subcellular protein localization occurred for 24P4C12 make the 58P1D12 protein and immune responses related thereto very useful.
  • Use of the 58P1D12 compositions allows those skilled in the art to make important diagnostic and therapeutic decisions.
  • Immunohistochemical reagents specific to 58P1D12 are also useful to detect metastases of tumors expressing 58P1D12 when the polypeptide appears in tissues where 58P1D12 is not normally produced.
  • 58P1D12 polypeptides and antibodies resulting from immune responses thereto are useful in a variety of important contexts such as diagnostic, prognostic, preventative and/or therapeutic purposes known to those skilled in the art.
  • PSA polynucleotide fragments and polynucleotide variants are employed by skilled artisans for use in methods of monitoring PSA
  • 58P1D12 polynucleotide fragments and polynucleotide variants are used in an analogous manner.
  • typical PSA polynucleotides used in methods of monitoring PSA are probes or primers which consist of fragments of the PSA cDNA sequence.
  • primers used to PCR amplify a PSA polynucleotide must include less than the whole PSA sequence to function in the polymerase chain reaction.
  • PCR reactions In the context of such PCR reactions, skilled artisans generally create a variety of different polynucleotide fragments that can be used as primers in order to amplify different portions of a polynucleotide of interest or to optimize amplification reactions (see, e.g., Caetano-Anolles, G. Biotechniques 25(3): 472-476, 478-480 (1998); Robertson et al., Methods Mol. Biol. 98:121-154 (1998)).
  • variant polynucleotide sequences are typically used as primers and probes for the corresponding mRNAs in PCR and Northern analyses (see, e.g., Sawai et al., Fetal Diagn. Ther. 1996 November-December 11(6):407-13 and Current Protocols In Molecular Biology, Volume 2, Unit 2, Frederick M. Ausubel et al. eds., 1995)).
  • Polynucleotide fragments and variants are useful in this context where they are capable of binding to a target polynucleotide sequence (e.g., a 58P1D12 polynucleotide shown in FIG. 2 or variant thereof) under conditions of high stringency.
  • a target polynucleotide sequence e.g., a 58P1D12 polynucleotide shown in FIG. 2 or variant thereof
  • PSA polypeptides which contain an epitope that can be recognized by an antibody or T cell that specifically binds to that epitope are used in methods of monitoring PSA.
  • 58P1D12 polypeptide fragments and polypeptide analogs or variants can also be used in an analogous manner.
  • This practice of using polypeptide fragments or polypeptide variants to generate antibodies is typical in the art with a wide variety of systems such as fusion proteins being used by practitioners (see, e.g., Current Protocols In Molecular Biology, Volume 2, Unit 16, Frederick M. Ausubel et al. eds., 1995).
  • each epitope(s) functions to provide the architecture with which an antibody or T cell is reactive.
  • polypeptide fragments that can be used in order to generate immune responses specific for different portions of a polypeptide of interest (see, e.g., U.S. Pat. No. 5,840,501 and U.S. Pat. No. 5,939,533).
  • a polypeptide comprising one of the 58P1D12 biological motifs discussed herein or a motif-bearing subsequence which is readily identified by one of skill in the art based on motifs available in the art.
  • Polypeptide fragments, variants or analogs are typically useful in this context as long as they comprise an epitope capable of generating an antibody or T cell specific for a target polypeptide sequence (e.g. a 58P1D12 polypeptide shown in FIG. 3 ).
  • the 58P1D12 polynucleotides and polypeptides exhibit specific properties that make them useful in diagnosing cancers such as those listed in Table I.
  • Diagnostic assays that measure the presence of 58P1D12 gene products in order to evaluate the presence or onset of a disease condition described herein, such as prostate cancer, are used to identify patients for preventive measures or further monitoring, as has been done so successfully with PSA.
  • these materials satisfy a need in the art for molecules having similar or complementary characteristics to PSA in situations where, for example, a definite diagnosis of metastasis of prostatic origin cannot be made on the basis of a test for PSA alone (see, e.g., Alanen et al., Pathol. Res. Pract. 192(3): 233-237 (1996)), and consequently, materials such as 58P1D12 polynucleotides and polypeptides (as well as the 58P1D12 polynucleotide probes and anti-58P1D12 antibodies used to identify the presence of these molecules) need to be employed to confirm a metastases of prostatic origin.
  • the 58P1D12 polynucleotides disclosed herein have a number of other utilities such as their use in the identification of oncogenetic associated chromosomal abnormalities in the chromosomal region to which the 58P1D12 gene maps (see the Example entitled “Chromosomal Mapping of 58P1D12” below).
  • the 58P1D12-related proteins and polynucleotides disclosed herein have other utilities such as their use in the forensic analysis of tissues of unknown origin (see, e.g., Takahama K Forensic Sci Int 1996 Jun. 28; 80(1-2): 63-9).
  • 58P1D12-related proteins or polynucleotides of the invention can be used to treat a pathologic condition characterized by the over-expression of 58P1D12.
  • the amino acid or nucleic acid sequence of FIG. 2 or FIG. 3 , or fragments of either can be used to generate an immune response to a 58P1D12 antigen.
  • Antibodies or other molecules that react with 58P1D12 can be used to modulate the function of this molecule, and thereby provide a therapeutic benefit.
  • the invention includes various methods and compositions for inhibiting the binding of 58P1D12 to its binding partner or its association with other protein(s) as well as methods for inhibiting 58P1D12 function.
  • a recombinant vector that encodes single chain antibodies that specifically bind to 58P1D12 are introduced into 58P1D12 expressing cells via gene transfer technologies. Accordingly, the encoded single chain anti-58P1D12 antibody is expressed intracellularly, binds to 58P1D12 protein, and thereby inhibits its function.
  • Methods for engineering such intracellular single chain antibodies are well known.
  • intracellular antibodies also known as “intrabodies”, are specifically targeted to a particular compartment within the cell, providing control over where the inhibitory activity of the treatment is focused. This technology has been successfully applied in the art (for review, see Richardson and Marasco, 1995, TIBTECH vol. 13).
  • Intrabodies have been shown to virtually eliminate the expression of otherwise abundant cell surface receptors (see, e.g., Richardson et al., 1995, Proc. Natl. Acad. Sci. USA 92: 3137-3141; Beerli et al., 1994, J. Biol. Chem. 289: 23931-23936; Deshane et al., 1994, Gene Ther. 1: 332-337).
  • Single chain antibodies comprise the variable domains of the heavy and light chain joined by a flexible linker polypeptide, and are expressed as a single polypeptide.
  • single chain antibodies are expressed as a single chain variable region fragment joined to the light chain constant region.
  • Well-known intracellular trafficking signals are engineered into recombinant polynucleotide vectors encoding such single chain antibodies in order to target precisely the intrabody to the desired intracellular compartment.
  • intrabodies targeted to the endoplasmic reticulum (ER) are engineered to incorporate a leader peptide and, optionally, a C-terminal ER retention signal, such as the KDEL amino acid motif.
  • Intrabodies intended to exert activity in the nucleus are engineered to include a nuclear localization signal.
  • Lipid moieties are joined to intrabodies in order to tether the intrabody to the cytosolic side of the plasma membrane.
  • Intrabodies can also be targeted to exert function in the cytosol.
  • cytosolic intrabodies are used to sequester factors within the cytosol, thereby preventing them from being transported to their natural cellular destination.
  • intrabodies are used to capture 58P1D12 in the nucleus, thereby preventing its activity within the nucleus.
  • Nuclear targeting signals are engineered into such 58P1D12 intrabodies in order to achieve the desired targeting.
  • Such 58P1D12 intrabodies are designed to bind specifically to a particular 58P1D12 domain.
  • cytosolic intrabodies that specifically bind to a 58P1D12 protein are used to prevent 58P1D12 from gaining access to the nucleus, thereby preventing it from exerting any biological activity within the nucleus (e.g., preventing 58P1D12 from forming transcription complexes with other factors).
  • the transcription of the intrabody is placed under the regulatory control of an appropriate tumor-specific promoter and/or enhancer.
  • an appropriate tumor-specific promoter and/or enhancer In order to target intrabody expression specifically to prostate, for example, the PSA promoter and/or promoter/enhancer can be utilized (See, for example, U.S. Pat. No. 5,919,652 issued 6 Jul. 1999).
  • recombinant molecules bind to 58P1D12 and thereby inhibit 58P1D12 function.
  • these recombinant molecules prevent or inhibit 58P1D12 from accessing/binding to its binding partner(s) or associating with other protein(s).
  • Such recombinant molecules can, for example, contain the reactive part(s) of a 58P1D12 specific antibody molecule.
  • the 58P1D12 binding domain of a 58P1D12 binding partner is engineered into a dimeric fusion protein, whereby the fusion protein comprises two 58P1D12 ligand binding domains linked to the Fc portion of a human IgG, such as human IgG1.
  • Such IgG portion can contain, for example, the CH2 and CH3 domains and the hinge region, but not the CH1 domain.
  • Such dimeric fusion proteins are administered in soluble form to patients suffering from a cancer associated with the expression of 58P1D12, whereby the dimeric fusion protein specifically binds to 58P1D12 and blocks 58P1D12 interaction with a binding partner.
  • Such dimeric fusion proteins are further combined into multimeric proteins using known antibody linking technologies.
  • the present invention also comprises various methods and compositions for inhibiting the transcription of the 58P1D12 gene. Similarly, the invention also provides methods and compositions for inhibiting the translation of 58P1D12 mRNA into protein.
  • a method of inhibiting the transcription of the 58P1D12 gene comprises contacting the 58P1D12 gene with a 58P1D12 antisense polynucleotide.
  • a method of inhibiting 58P1D12 mRNA translation comprises contacting a 58P1D12 mRNA with an antisense polynucleotide.
  • a 58P1D12 specific ribozyme is used to cleave a 58P1D12 message, thereby inhibiting translation.
  • antisense and ribozyme based methods can also be directed to the regulatory regions of the 58P1D12 gene, such as 58P1D12 promoter and/or enhancer elements.
  • proteins capable of inhibiting a 58P1D12 gene transcription factor are used to inhibit 58P1D12 mRNA transcription.
  • the various polynucleotides and compositions useful in the aforementioned methods have been described above.
  • the use of antisense and ribozyme molecules to inhibit transcription and translation is well known in the art.
  • Gene transfer and gene therapy technologies can be used to deliver therapeutic polynucleotide molecules to tumor cells synthesizing 58P1D12 (i.e., antisense, ribozyme, polynucleotides encoding intrabodies and other 58P1D12 inhibitory molecules).
  • 58P1D12 i.e., antisense, ribozyme, polynucleotides encoding intrabodies and other 58P1D12 inhibitory molecules.
  • a number of gene therapy approaches are known in the art.
  • Recombinant vectors encoding 58P1D12 antisense polynucleotides, ribozymes, factors capable of interfering with 58P1D12 transcription, and so forth, can be delivered to target tumor cells using such gene therapy approaches.
  • the above therapeutic approaches can be combined with any one of a wide variety of surgical, chemotherapy or radiation therapy regimens.
  • the therapeutic approaches of the invention can enable the use of reduced dosages of chemotherapy (or other therapies) and/or less frequent administration, an advantage for all patients and particularly for those that do not tolerate the toxicity of the chemotherapeutic agent well.
  • the anti-tumor activity of a particular composition can be evaluated using various in vitro and in vivo assay systems.
  • In vitro assays that evaluate therapeutic activity include cell growth assays, soft agar assays and other assays indicative of tumor promoting activity, binding assays capable of determining the extent to which a therapeutic composition will inhibit the binding of 58P1D12 to a binding partner, etc.
  • a 58P1D12 therapeutic composition can be evaluated in a suitable animal model.
  • xenogenic prostate cancer models can be used, wherein human prostate cancer explants or passaged xenograft tissues are introduced into immune compromised animals, such as nude or SCID mice (Klein et al., 1997, Nature Medicine 3: 402-408).
  • PCT Patent Application WO98/16628 and U.S. Pat. No. 6,107,540 describe various xenograft models of human prostate cancer capable of recapitulating the development of primary tumors, micrometastasis, and the formation of osteoblastic metastases characteristic of late stage disease. Efficacy can be predicted using assays that measure inhibition of tumor formation, tumor regression or metastasis, and the like.
  • xenografts from tumor bearing mice treated with the therapeutic composition can be examined for the presence of apoptotic foci and compared to untreated control xenograft-bearing mice. The extent to which apoptotic foci are found in the tumors of the treated mice provides an indication of the therapeutic efficacy of the composition.
  • Suitable carriers include any material that when combined with the therapeutic composition retains the anti-tumor function of the therapeutic composition and is generally non-reactive with the patient's immune system. Examples include, but are not limited to, any of a number of standard pharmaceutical carriers such as sterile phosphate buffered saline solutions, bacteriostatic water, and the like (see, generally, Remington's Pharmaceutical Sciences 16th Edition, A. Osal., Ed., 1980).
  • Therapeutic formulations can be solubilized and administered via any route capable of delivering the therapeutic composition to the tumor site.
  • Potentially effective routes of administration include, but are not limited to, intravenous, parenteral, intraperitoneal, intramuscular, intratumor, intradermal, intraorgan, orthotopic, and the like.
  • a preferred formulation for intravenous injection comprises the therapeutic composition in a solution of preserved bacteriostatic water, sterile unpreserved water, and/or diluted in polyvinylchloride or polyethylene bags containing 0.9% sterile Sodium Chloride for Injection, USP.
  • Therapeutic protein preparations can be lyophilized and stored as sterile powders, preferably under vacuum, and then reconstituted in bacteriostatic water (containing for example, benzyl alcohol preservative) or in sterile water prior to injection.
  • Dosages and administration protocols for the treatment of cancers using the foregoing methods will vary with the method and the target cancer, and will generally depend on a number of other factors appreciated in the art.
  • screening is performed to identify modulators that induce or suppress a particular expression profile, suppress or induce specific pathways, preferably generating the associated phenotype thereby.
  • having identified differentially expressed genes important in a particular state screens are performed to identify modulators that alter expression of individual genes, either increase or decrease.
  • screening is performed to identify modulators that alter a biological function of the expression product of a differentially expressed gene. Again, having identified the importance of a gene in a particular state, screens are performed to identify agents that bind and/or modulate the biological activity of the gene product.
  • screens are done for genes that are induced in response to a candidate agent.
  • identifying a modulator one that suppresses a cancer expression pattern leading to a normal expression pattern, or a modulator of a cancer gene that leads to expression of the gene as in normal tissue
  • a screen is performed to identify genes that are specifically modulated in response to the agent. Comparing expression profiles between normal tissue and agent-treated cancer tissue reveals genes that are not expressed in normal tissue or cancer tissue, but are expressed in agent treated tissue, and vice versa.
  • agent-specific sequences are identified and used by methods described herein for cancer genes or proteins. In particular these sequences and the proteins they encode are used in marking or identifying agent-treated cells.
  • antibodies are raised against the agent-induced proteins and used to target novel therapeutics to the treated cancer tissue sample.
  • Proteins, nucleic acids, and antibodies of the invention are used in screening assays.
  • the cancer-associated proteins, antibodies, nucleic acids, modified proteins and cells containing these sequences are used in screening assays, such as evaluating the effect of drug candidates on a “gene expression profile,” expression profile of polypeptides or alteration of biological function.
  • the expression profiles are used, preferably in conjunction with high throughput screening techniques to allow monitoring for expression profile genes after treatment with a candidate agent (e.g., Davis, G F, et al, J Biol Screen 7:69 (2002); Zlokamik, et al., Science 279:84-8 (1998); Heid, Genome Res 6:986-94, 1996).
  • the cancer proteins, antibodies, nucleic acids, modified proteins and cells containing the native or modified cancer proteins or genes are used in screening assays. That is, the present invention comprises methods for screening for compositions which modulate the cancer phenotype or a physiological function of a cancer protein of the invention. This is done on a gene itself or by evaluating the effect of drug candidates on a “gene expression profile” or biological function. In one embodiment, expression profiles are used, preferably in conjunction with high throughput screening techniques to allow monitoring after treatment with a candidate agent, see Zlokarnik, supra.
  • assays are executed directed to the genes and proteins of the invention. Assays are run on an individual nucleic acid or protein level. That is, having identified a particular gene as up regulated in cancer, test compounds are screened for the ability to modulate gene expression or for binding to the cancer protein of the invention. “Modulation” in this context includes an increase or a decrease in gene expression. The preferred amount of modulation will depend on the original change of the gene expression in normal versus tissue undergoing cancer, with changes of at least 10%, preferably 50%, more preferably 100-300%, and in some embodiments 300-1000% or greater.
  • a gene exhibits a 4-fold increase in cancer tissue compared to normal tissue, a decrease of about four-fold is often desired; similarly, a 10-fold decrease in cancer tissue compared to normal tissue a target value of a 10-fold increase in expression by the test compound is often desired.
  • Modulators that exacerbate the type of gene expression seen in cancer are also useful, e.g., as an upregulated target in further analyses.
  • the amount of gene expression is monitored using nucleic acid probes and the quantification of gene expression levels, or, alternatively, a gene product itself is monitored, e.g., through the use of antibodies to the cancer protein and standard immunoassays. Proteomics and separation techniques also allow for quantification of expression.
  • gene expression monitoring i.e., an expression profile
  • Such profiles will typically involve one or more of the genes of FIG. 2 .
  • cancer nucleic acid probes are attached to biochips to detect and quantify cancer sequences in a particular cell.
  • PCR can be used.
  • a series e.g., wells of a microtiter plate, can be used with dispensed primers in desired wells. A PCR reaction can then be performed and analyzed for each well.
  • Expression monitoring is performed to identify compounds that modify the expression of one or more cancer-associated sequences, e.g., a polynucleotide sequence set out in FIG. 2 .
  • a test modulator is added to the cells prior to analysis.
  • screens are also provided to identify agents that modulate cancer, modulate cancer proteins of the invention, bind to a cancer protein of the invention, or interfere with the binding of a cancer protein of the invention and an antibody or other binding partner.
  • high throughput screening methods involve providing a library containing a large number of potential therapeutic compounds (candidate compounds). Such “combinatorial chemical libraries” are then screened in one or more assays to identify those library members (particular chemical species or subclasses) that display a desired characteristic activity. The compounds thus identified can serve as conventional “lead compounds,” as compounds for screening, or as therapeutics.
  • combinatorial libraries of potential modulators are screened for an ability to bind to a cancer polypeptide or to modulate activity.
  • new chemical entities with useful properties are generated by identifying a chemical compound (called a “lead compound”) with some desirable property or activity, e.g., inhibiting activity, creating variants of the lead compound, and evaluating the property and activity of those variant compounds.
  • HTS high throughput screening
  • gene expression monitoring is conveniently used to test candidate modulators (e.g., protein, nucleic acid or small molecule).
  • candidate modulators e.g., protein, nucleic acid or small molecule.
  • the target sequence is prepared using known techniques. For example, a sample is treated to lyse the cells, using known lysis buffers, electroporation, etc., with purification and/or amplification such as PCR performed as appropriate. For example, an in vitro transcription with labels covalently attached to the nucleotides is performed. Generally, the nucleic acids are labeled with biotin-FITC or PE, or with cy3 or cy5.
  • the target sequence can be labeled with, e.g., a fluorescent, a chemiluminescent, a chemical, or a radioactive signal, to provide a means of detecting the target sequence's specific binding to a probe.
  • the label also can be an enzyme, such as alkaline phosphatase or horseradish peroxidase, which when provided with an appropriate substrate produces a product that is detected.
  • the label is a labeled compound or small molecule, such as an enzyme inhibitor, that binds but is not catalyzed or altered by the enzyme.
  • the label also can be a moiety or compound, such as, an epitope tag or biotin which specifically binds to streptavidin.
  • the streptavidin is labeled as described above, thereby, providing a detectable signal for the bound target sequence. Unbound labeled streptavidin is typically removed prior to analysis.
  • these assays can be direct hybridization assays or can comprise “sandwich assays”, which include the use of multiple probes, as is generally outlined in U.S. Pat. Nos. 5,681,702; 5,597,909; 5,545,730; 5,594,117; 5,591,584; 5,571,670; 5,580,731; 5,571,670; 5,591,584; 5,624,802; 5,635,352; 5,594,118; 5,359,100; 5,124, 246; and 5,681,697.
  • the target nucleic acid is prepared as outlined above, and then added to the biochip comprising a plurality of nucleic acid probes, under conditions that allow the formation of a hybridization complex.
  • hybridization conditions are used in the present invention, including high, moderate and low stringency conditions as outlined above.
  • the assays are generally run under stringency conditions which allow formation of the label probe hybridization complex only in the presence of target.
  • Stringency can be controlled by altering a step parameter that is a thermodynamic variable, including, but not limited to, temperature, formamide concentration, salt concentration, chaotropic salt concentration pH, organic solvent concentration, etc. These parameters may also be used to control non-specific binding, as is generally outlined in U.S. Pat. No. 5,681,697. Thus, it can be desirable to perform certain steps at higher stringency conditions to reduce non-specific binding.
  • the reactions outlined herein can be accomplished in a variety of ways. Components of the reaction can be added simultaneously, or sequentially, in different orders, with preferred embodiments outlined below.
  • the reaction may include a variety of other reagents. These include salts, buffers, neutral proteins, e.g. albumin, detergents, etc. which can be used to facilitate optimal hybridization and detection, and/or reduce nonspecific or background interactions. Reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., may also be used as appropriate, depending on the sample preparation methods and purity of the target.
  • the assay data are analyzed to determine the expression levels of individual genes, and changes in expression levels as between states, forming a gene expression profile.
  • the invention provides methods identify or screen for a compound that modulates the activity of a cancer-related gene or protein of the invention.
  • the methods comprise adding a test compound, as defined above, to a cell comprising a cancer protein of the invention.
  • the cells contain a recombinant nucleic acid that encodes a cancer protein of the invention.
  • a library of candidate agents is tested on a plurality of cells.
  • the assays are evaluated in the presence or absence or previous or subsequent exposure of physiological signals, e.g. hormones, antibodies, peptides, antigens, cytokines, growth factors, action potentials, pharmacological agents including chemotherapeutics, radiation, carcinogenics, or other cells (i.e., cell-cell contacts).
  • physiological signals e.g. hormones, antibodies, peptides, antigens, cytokines, growth factors, action potentials, pharmacological agents including chemotherapeutics, radiation, carcinogenics, or other cells (i.e., cell-cell contacts).
  • the determinations are made at different stages of the cell cycle process. In this way, compounds that modulate genes or proteins of the invention are identified. Compounds with pharmacological activity are able to enhance or interfere with the activity of the cancer protein of the invention. Once identified, similar structures are evaluated to identify critical structural features of the compound.
  • a method of modulating (e.g., inhibiting) cancer cell division comprises administration of a cancer modulator.
  • a method of modulating (e.g., inhibiting) cancer is provided; the method comprises administration of a cancer modulator.
  • methods of treating cells or individuals with cancer are provided; the method comprises administration of a cancer modulator.
  • a method for modulating the status of a cell that expresses a gene of the invention comprises such art-accepted parameters such as growth, proliferation, survival, function, apoptosis, senescence, location, enzymatic activity, signal transduction, etc. of a cell.
  • a cancer inhibitor is an antibody as discussed above.
  • the cancer inhibitor is an antisense molecule.
  • a variety of cell growth, proliferation, and metastasis assays are known to those of skill in the art, as described herein.
  • the assays to identify suitable modulators are amenable to high throughput screening. Preferred assays thus detect enhancement or inhibition of cancer gene transcription, inhibition or enhancement of polypeptide expression, and inhibition or enhancement of polypeptide activity.
  • modulators evaluated in high throughput screening methods are proteins, often naturally occurring proteins or fragments of naturally occurring proteins.
  • proteins e.g., cellular extracts containing proteins, or random or directed digests of proteinaceous cellular extracts, are used.
  • libraries of proteins are made for screening in the methods of the invention.
  • Particularly preferred in this embodiment are libraries of bacterial, fungal, viral, and mammalian proteins, with the latter being preferred, and human proteins being especially preferred.
  • Particularly useful test compound will be directed to the class of proteins to which the target belongs, e.g., substrates for enzymes, or ligands and receptors.
  • Normal cells require a solid substrate to attach and grow. When cells are transformed, they lose this phenotype and grow detached from the substrate. For example, transformed cells can grow in stirred suspension culture or suspended in semi-solid media, such as semi-solid or soft agar. The transformed cells, when transfected with tumor suppressor genes, can regenerate normal phenotype and once again require a solid substrate to attach to and grow. Soft agar growth or colony formation in assays are used to identify modulators of cancer sequences, which when expressed in host cells, inhibit abnormal cellular proliferation and transformation. A modulator reduces or eliminates the host cells' ability to grow suspended in solid or semisolid media, such as agar.
  • Normal cells typically grow in a flat and organized pattern in cell culture until they touch other cells. When the cells touch one another, they are contact inhibited and stop growing. Transformed cells, however, are not contact inhibited and continue to grow to high densities in disorganized foci. Thus, transformed cells grow to a higher saturation density than corresponding normal cells. This is detected morphologically by the formation of a disoriented monolayer of cells or cells in foci.
  • labeling index with (3H)-thymidine at saturation density is used to measure density limitation of growth, similarly an MTT or Alamar blue assay will reveal proliferation capacity of cells and the ability of modulators to affect same. See Freshney (1994), supra.
  • Transformed cells when transfected with tumor suppressor genes, can regenerate a normal phenotype and become contact inhibited and would grow to a lower density.
  • labeling index with 3H)-thymidine at saturation density is a preferred method of measuring density limitation of growth.
  • Transformed host cells are transfected with a cancer-associated sequence and are grown for 24 hours at saturation density in non-limiting medium conditions.
  • the percentage of cells labeling with (3H)-thymidine is determined by incorporated cpm.
  • a modulator reduces or eliminates contact independent growth, and returns the cells to a normal phenotype.
  • Transformed cells have lower serum dependence than their normal counterparts (see, e.g., Temin, J. Natl. Cancer Inst. 37:167-175 (1966); Eagle et al., J. Exp. Med. 131:836-879 (1970)); Freshney, supra. This is in part due to release of various growth factors by the transformed cells.
  • the degree of growth factor or serum dependence of transformed host cells can be compared with that of control. For example, growth factor or serum dependence of a cell is monitored in methods to identify and characterize compounds that modulate cancer-associated sequences of the invention.
  • Tumor cells release an increased amount of certain factors (hereinafter “tumor specific markers”) than their normal counterparts.
  • plasminogen activator PA
  • PA plasminogen activator
  • Tumor Angiogenesis Factor TAF
  • TAF Tumor Angiogenesis Factor
  • the degree of invasiveness into Matrigel or an extracellular matrix constituent can be used as an assay to identify and characterize compounds that modulate cancer associated sequences.
  • Tumor cells exhibit a positive correlation between malignancy and invasiveness of cells into Matrigel or some other extracellular matrix constituent.
  • tumorigenic cells are typically used as host cells. Expression of a tumor suppressor gene in these host cells would decrease invasiveness of the host cells. Techniques described in Cancer Res. 1999; 59:6010; Freshney (1994), supra, can be used. Briefly, the level of invasion of host cells is measured by using filters coated with Matrigel or some other extracellular matrix constituent.
  • Penetration into the gel, or through to the distal side of the filter, is rated as invasiveness, and rated histologically by number of cells and distance moved, or by prelabeling the cells with 1251 and counting the radioactivity on the distal side of the filter or bottom of the dish. See, e.g., Freshney (1984), supra.
  • Transgenic organisms are prepared in a variety of art-accepted ways. For example, knock-out transgenic organisms, e.g., mammals such as mice, are made, in which a cancer gene is disrupted or in which a cancer gene is inserted. Knock-out transgenic mice are made by insertion of a marker gene or other heterologous gene into the endogenous cancer gene site in the mouse genome via homologous recombination. Such mice can also be made by substituting the endogenous cancer gene with a mutated version of the cancer gene, or by mutating the endogenous cancer gene, e.g., by exposure to carcinogens.
  • transgenic chimeric animals e.g., mice
  • a DNA construct is introduced into the nuclei of embryonic stem cells.
  • Cells containing the newly engineered genetic lesion are injected into a host mouse embryo, which is re-implanted into a recipient female. Some of these embryos develop into chimeric mice that possess germ cells some of which are derived from the mutant cell line. Therefore, by breeding the chimeric mice it is possible to obtain a new line of mice containing the introduced genetic lesion (see, e.g., Capecchi et al., Science 244:1288 (1989)).
  • Chimeric mice can be derived according to U.S. Pat. No. 6,365,797, issued 2 Apr. 2002; U.S. Pat. No.
  • various immune-suppressed or immune-deficient host animals can be used.
  • a genetically athymic “nude” mouse see, e.g., Giovanella et al., J. Natl. Cancer Inst. 52:921 (1974)
  • SCID mouse see, e.g., Giovanella et al., J. Natl. Cancer Inst. 52:921 (1974)
  • SCID mouse e.g., a thymectornized mouse
  • an irradiated mouse see, e.g., Bradley et al., Br. J. Cancer 38:263 (1978); Selby et al., Br. J. Cancer 41:52 (1980)
  • Transplantable tumor cells typically about 106 cells injected into isogenic hosts produce invasive tumors in a high proportion of cases, while normal cells of similar origin will not.
  • cells expressing cancer-associated sequences are injected subcutaneously or orthotopically. Mice are then separated into groups, including control groups and treated experimental groups) e.g. treated with a modulator). After a suitable length of time, preferably 4-8 weeks, tumor growth is measured (e.g., by volume or by its two largest dimensions, or weight) and compared to the control. Tumors that have statistically significant reduction (using, e.g., Student's T test) are said to have inhibited growth.
  • Assays to identify compounds with modulating activity can be performed in vitro.
  • a cancer polypeptide is first contacted with a potential modulator and incubated for a suitable amount of time, e.g., from 0.5 to 48 hours.
  • the cancer polypeptide levels are determined in vitro by measuring the level of protein or mRNA.
  • the level of protein is measured using immunoassays such as Western blotting, ELISA and the like with an antibody that selectively binds to the cancer polypeptide or a fragment thereof.
  • amplification e.g., using PCR, LCR, or hybridization assays, e.g., Northern hybridization, RNAse protection, dot blotting, are preferred.
  • the level of protein or mRNA is detected using directly or indirectly labeled detection agents, e.g., fluorescently or radioactively labeled nucleic acids, radioactively or enzymatically labeled antibodies, and the like, as described herein.
  • a reporter gene system can be devised using a cancer protein promoter operably linked to a reporter gene such as luciferase, green fluorescent protein, CAT, or P-gal.
  • the reporter construct is typically transfected into a cell. After treatment with a potential modulator, the amount of reporter gene transcription, translation, or activity is measured according to standard techniques known to those of skill in the art (Davis GF, supra; Gonzalez, J. & Negulescu, P. Curr. Opin. Biotechnol. 1998: 9:624).
  • in vitro screens are done on individual genes and gene products. That is, having identified a particular differentially expressed gene as important in a particular state, screening of modulators of the expression of the gene or the gene product itself is performed.
  • screening for modulators of expression of specific gene(s) is performed. Typically, the expression of only one or a few genes is evaluated. In another embodiment, screens are designed to first find compounds that bind to differentially expressed proteins. These compounds are then evaluated for the ability to modulate differentially expressed activity. Moreover, once initial candidate compounds are identified, variants can be further screened to better evaluate structure activity relationships.
  • a purified or isolated gene product of the invention is generally used.
  • antibodies are generated to a protein of the invention, and immunoassays are run to determine the amount and/or location of protein.
  • cells comprising the cancer proteins are used in the assays.
  • the methods comprise combining a cancer protein of the invention and a candidate compound such as a ligand, and determining the binding of the compound to the cancer protein of the invention.
  • a cancer protein of the invention utilizes the human cancer protein; animal models of human disease of can also be developed and used.
  • other analogous mammalian proteins also can be used as appreciated by those of skill in the art.
  • variant or derivative cancer proteins are used.
  • the cancer protein of the invention, or the ligand is non-diffusibly bound to an insoluble support.
  • the support can, e.g., be one having isolated sample receiving areas (a microtiter plate, an array, etc.).
  • the insoluble supports can be made of any composition to which the compositions can be bound, is readily separated from soluble material, and is otherwise compatible with the overall method of screening.
  • the surface of such supports can be solid or porous and of any convenient shape.
  • suitable insoluble supports include microtiter plates, arrays, membranes and beads. These are typically made of glass, plastic (e.g., polystyrene), polysaccharide, nylon, nitrocellulose, or TeflonTM, etc. Microtiter plates and arrays are especially convenient because a large number of assays can be carried out simultaneously, using small amounts of reagents and samples. The particular manner of binding of the composition to the support is not crucial so long as it is compatible with the reagents and overall methods of the invention, maintains the activity of the composition and is nondiffusable.
  • Preferred methods of binding include the use of antibodies which do not sterically block either the ligand binding site or activation sequence when attaching the protein to the support, direct binding to “sticky” or ionic supports, chemical crosslinking, the synthesis of the protein or agent on the surface, etc. Following binding of the protein or ligand/binding agent to the support, excess unbound material is removed by washing. The sample receiving areas may then be blocked through incubation with bovine serum albumin (BSA), casein or other innocuous protein or other moiety.
  • BSA bovine serum albumin
  • Binding agents include specific antibodies, non-natural binding agents identified in screens of chemical libraries, peptide analogs, etc.
  • assays to identify agents that have a low toxicity for human cells.
  • a wide variety of assays can be used for this purpose, including proliferation assays, cAMP assays, labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays for protein binding, functional assays (phosphorylation assays, etc.) and the like.
  • a determination of binding of the test compound (ligand, binding agent, modulator, etc.) to a cancer protein of the invention can be done in a number of ways.
  • the test compound can be labeled, and binding determined directly, e.g., by attaching all or a portion of the cancer protein of the invention to a solid support, adding a labeled candidate compound (e.g., a fluorescent label), washing off excess reagent, and determining whether the label is present on the solid support.
  • a labeled candidate compound e.g., a fluorescent label
  • only one of the components is labeled, e.g., a protein of the invention or ligands labeled.
  • more than one component is labeled with different labels, e.g., I125, for the proteins and a fluorophor for the compound.
  • Proximity reagents e.g., quenching or energy transfer reagents are also useful.
  • the binding of the “test compound” is determined by competitive binding assay with a “competitor.”
  • the competitor is a binding moiety that binds to the target molecule (e.g., a cancer protein of the invention). Competitors include compounds such as antibodies, peptides, binding partners, ligands, etc. Under certain circumstances, the competitive binding between the test compound and the competitor displaces the test compound.
  • the test compound is labeled. Either the test compound, the competitor, or both, is added to the protein for a time sufficient to allow binding. Incubations are performed at a temperature that facilitates optimal activity, typically between four and 40° C.
  • Incubation periods are typically optimized, e.g., to facilitate rapid high throughput screening; typically between zero and one hour will be sufficient. Excess reagent is generally removed or washed away. The second component is then added, and the presence or absence of the labeled component is followed, to indicate binding.
  • the competitor is added first, followed by the test compound.
  • Displacement of the competitor is an indication that the test compound is binding to the cancer protein and thus is capable of binding to, and potentially modulating, the activity of the cancer protein.
  • either component can be labeled.
  • the presence of label in the post-test compound wash solution indicates displacement by the test compound.
  • the presence of the label on the support indicates displacement.
  • the test compound is added first, with incubation and washing, followed by the competitor.
  • the absence of binding by the competitor indicates that the test compound binds to the cancer protein with higher affinity than the competitor.
  • the presence of the label on the support, coupled with a lack of competitor binding indicates that the test compound binds to and thus potentially modulates the cancer protein of the invention.
  • the competitive binding methods comprise differential screening to identity agents that are capable of modulating the activity of the cancer proteins of the invention.
  • the methods comprise combining a cancer protein and a competitor in a first sample.
  • a second sample comprises a test compound, the cancer protein, and a competitor.
  • the binding of the competitor is determined for both samples, and a change, or difference in binding between the two samples indicates the presence of an agent capable of binding to the cancer protein and potentially modulating its activity. That is, if the binding of the competitor is different in the second sample relative to the first sample, the agent is capable of binding to the cancer protein.
  • differential screening is used to identify drug candidates that bind to the native cancer protein, but cannot bind to modified cancer proteins.
  • the structure of the cancer protein is modeled and used in rational drug design to synthesize agents that interact with that site, agents which generally do not bind to site-modified proteins.
  • drug candidates that affect the activity of a native cancer protein are also identified by screening drugs for the ability to either enhance or reduce the activity of such proteins.
  • Positive controls and negative controls can be used in the assays.
  • control and test samples are performed in at least triplicate to obtain statistically significant results. Incubation of all samples occurs for a time sufficient to allow for the binding of the agent to the protein. Following incubation, samples are washed free of non-specifically bound material and the amount of bound, generally labeled agent determined For example, where a radiolabel is employed, the samples can be counted in a scintillation counter to determine the amount of bound compound.
  • reagents can be included in the screening assays. These include reagents like salts, neutral proteins, e.g. albumin, detergents, etc. which are used to facilitate optimal protein-protein binding and/or reduce non-specific or background interactions. Also reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., can be used. The mixture of components is added in an order that provides for the requisite binding.
  • Polynucleotide modulators of cancer can be introduced into a cell containing the target nucleotide sequence by formation of a conjugate with a ligand-binding molecule, as described in WO 91/04753.
  • Suitable ligand-binding molecules include, but are not limited to, cell surface receptors, growth factors, other cytokines, or other ligands that bind to cell surface receptors.
  • conjugation of the ligand binding molecule does not substantially interfere with the ability of the ligand binding molecule to bind to its corresponding molecule or receptor, or block entry of the sense or antisense oligonucleotide or its conjugated version into the cell.
  • a polynucleotide modulator of cancer can be introduced into a cell containing the target nucleic acid sequence, e.g., by formation of a polynucleotide-lipid complex, as described in WO 90/10448. It is understood that the use of antisense molecules or knock out and knock in models may also be used in screening assays as discussed above, in addition to methods of treatment.
  • the activity of a cancer-associated protein is down-regulated, or entirely inhibited, by the use of antisense polynucleotide or inhibitory small nuclear RNA (snRNA), i.e., a nucleic acid complementary to, and which can preferably hybridize specifically to, a coding mRNA nucleic acid sequence, e.g., a cancer protein of the invention, mRNA, or a subsequence thereof. Binding of the antisense polynucleotide to the mRNA reduces the translation and/or stability of the mRNA.
  • snRNA inhibitory small nuclear RNA
  • antisense polynucleotides can comprise naturally occurring nucleotides, or synthetic species formed from naturally occurring subunits or their close homologs. Antisense polynucleotides may also have altered sugar moieties or inter-sugar linkages. Exemplary among these are the phosphorothioate and other sulfur containing species which are known for use in the art. Analogs are comprised by this invention so long as they function effectively to hybridize with nucleotides of the invention. See, e.g., Isis Pharmaceuticals, Carlsbad, Calif.; Sequitor, Inc., Natick, Mass.
  • antisense polynucleotides can readily be synthesized using recombinant means, or can be synthesized in vitro. Equipment for such synthesis is sold by several vendors, including Applied Biosystems. The preparation of other oligonucleotides such as phosphorothioates and alkylated derivatives is also well known to those of skill in the art.
  • Antisense molecules as used herein include antisense or sense oligonucleotides.
  • Sense oligonucleotides can, e.g., be employed to block transcription by binding to the anti-sense strand.
  • the antisense and sense oligonucleotide comprise a single stranded nucleic acid sequence (either RNA or DNA) capable of binding to target mRNA (sense) or DNA (antisense) sequences for cancer molecules.
  • Antisense or sense oligonucleotides, according to the present invention comprise a fragment generally at least about 12 nucleotides, preferably from about 12 to 30 nucleotides.
  • ribozymes can be used to target and inhibit transcription of cancer-associated nucleotide sequences.
  • a ribozyme is an RNA molecule that catalytically cleaves other RNA molecules.
  • Different kinds of ribozymes have been described, including group I ribozymes, hammerhead ribozymes, hairpin ribozymes, RNase P, and axhead ribozymes (see, e.g., Castanotto et al., Adv. in Pharmacology 25: 289-317 (1994) for a general review of the properties of different ribozymes).
  • hairpin ribozymes are described, e.g., in Hampel et al., Nucl. Acids Res. 18:299-304 (1990); European Patent Publication No. 0360257; U.S. Pat. No. 5,254,678.
  • Methods of preparing are well known to those of skill in the art (see, e.g., WO 94/26877; Ojwang et al., Proc. Natl. Acad. Sci. USA 90:6340-6344 (1993); Yamada et al., Human Gene Therapy 1:39-45 (1994); Leavitt et al., Proc. Natl. Acad. Sci. USA 92:699-703 (1995); Leavitt et al., Human Gene Therapy 5: 1151-120 (1994); and Yamada et al., Virology 205: 121-126 (1994)).
  • a test compound is administered to a population of cancer cells, which have an associated cancer expression profile.
  • administration or “contacting” herein is meant that the modulator is added to the cells in such a manner as to allow the modulator to act upon the cell, whether by uptake and intracellular action, or by action at the cell surface.
  • a nucleic acid encoding a proteinaceous agent i.e., a peptide
  • a viral construct such as an adenoviral or retroviral construct
  • expression of the peptide agent is accomplished, e.g., PCT US97/01019.
  • Regulatable gene therapy systems can also be used.
  • the cells are washed if desired and are allowed to incubate under preferably physiological conditions for some period.
  • the cells are then harvested and a new gene expression profile is generated.
  • cancer tissue is screened for agents that modulate, e.g., induce or suppress, the cancer phenotype.
  • a change in at least one gene, preferably many, of the expression profile indicates that the agent has an effect on cancer activity.
  • altering a biological function or a signaling pathway is indicative of modulator activity.
  • screens are done to assess genes or gene products. That is, having identified a particular differentially expressed gene as important in a particular state, screening of modulators of either the expression of the gene or the gene product itself is performed.
  • Measurements of cancer polypeptide activity, or of the cancer phenotype are performed using a variety of assays. For example, the effects of modulators upon the function of a cancer polypeptide(s) are measured by examining parameters described above. A physiological change that affects activity is used to assess the influence of a test compound on the polypeptides of this invention.
  • a variety of effects can be assesses such as, in the case of a cancer associated with solid tumors, tumor growth, tumor metastasis, neovascularization, hormone release, transcriptional changes to both known and uncharacterized genetic markers (e.g., by Northern blots), changes in cell metabolism such as cell growth or pH changes, and changes in intracellular second messengers such as cGNIP.
  • the invention provides methods for identifying cells containing variant cancer genes, e.g., determining the presence of, all or part, the sequence of at least one endogenous cancer gene in a cell. This is accomplished using any number of sequencing techniques.
  • the invention comprises methods of identifying the cancer genotype of an individual, e.g., determining all or part of the sequence of at least one gene of the invention in the individual. This is generally done in at least one tissue of the individual, e.g., a tissue set forth in Table I, and may include the evaluation of a number of tissues or different samples of the same tissue.
  • the method may include comparing the sequence of the sequenced gene to a known cancer gene, i.e., a wild-type gene to determine the presence of family members, homologies, mutations or variants.
  • the sequence of all or part of the gene can then be compared to the sequence of a known cancer gene to determine if any differences exist. This is done using any number of known homology programs, such as BLAST, Bestfit, etc.
  • the presence of a difference in the sequence between the cancer gene of the patient and the known cancer gene correlates with a disease state or a propensity for a disease state, as outlined herein.
  • the cancer genes are used as probes to determine the number of copies of the cancer gene in the genome.
  • the cancer genes are used as probes to determine the chromosomal localization of the cancer genes.
  • Information such as chromosomal localization finds use in providing a diagnosis or prognosis in particular when chromosomal abnormalities such as translocations, and the like are identified in the cancer gene locus.
  • kits are within the scope of the invention.
  • kits can comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in the method, along with a label or insert comprising instructions for use, such as a use described herein.
  • the container(s) can comprise a probe that is or can be detectably labeled.
  • probe can be an antibody or polynucleotide specific for a protein or a gene or message of the invention, respectively.
  • kits can also have containers containing nucleotide(s) for amplification of the target nucleic acid sequence.
  • Kits can comprise a container comprising a reporter, such as a biotin-binding protein, such as avidin or streptavidin, bound to a reporter molecule, such as an enzymatic, fluorescent, or radioisotope label; such a reporter can be used with, e.g., a nucleic acid or antibody.
  • the kit can include all or part of the amino acid sequences in FIG. 2 or FIG. 3 or analogs thereof, or a nucleic acid molecule that encodes such amino acid sequences.
  • the kit of the invention will typically comprise the container described above and one or more other containers associated therewith that comprise materials desirable from a commercial and user standpoint, including buffers, diluents, filters, needles, syringes; carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use.
  • a label can be present on or with the container to indicate that the composition is used for a specific therapy or non-therapeutic application, such as a prognostic, prophylactic, diagnostic or laboratory application, and can also indicate directions for either in vivo or in vitro use, such as those described herein. Directions and or other information can also be included on an insert(s) or label(s) which is included with or on the kit.
  • the label can be on or associated with the container.
  • a label a can be on a container when letters, numbers or other characters forming the label are molded or etched into the container itself; a label can be associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert.
  • the label can indicate that the composition is used for diagnosing, treating, prophylaxing or prognosing a condition, such as a neoplasia of a tissue set forth in Table I.
  • an article(s) of manufacture containing compositions such as amino acid sequence(s), small molecule(s), nucleic acid sequence(s), and/or antibody(s), e.g., materials useful for the diagnosis, prognosis, prophylaxis and/or treatment of neoplasias of tissues such as those set forth in Table I.
  • the article of manufacture typically comprises at least one container and at least one label. Suitable containers include, for example, bottles, vials, syringes, and test tubes.
  • the containers can be formed from a variety of materials such as glass, metal or plastic.
  • the container can hold amino acid sequence(s), small molecule(s), nucleic acid sequence(s), cell population(s) and/or antibody(s).
  • the container holds a polynucleotide for use in examining the mRNA expression profile of a cell, together with reagents used for this purpose.
  • a container comprises an antibody, binding fragment thereof or specific binding protein for use in evaluating protein expression of 58P1D12 in cells and tissues, or for relevant laboratory, prognostic, diagnostic, prophylactic and therapeutic purposes; indications and/or directions for such uses can be included on or with such container, as can reagents and other compositions or tools used for these purposes.
  • a container comprises materials for eliciting a cellular or humoral immune response, together with associated indications and/or directions.
  • a container comprises materials for adoptive immunotherapy, such as cytotoxic T cells (CTL) or helper T cells (HTL), together with associated indications and/or directions; reagents and other compositions or tools used for such purpose can also be included.
  • CTL cytotoxic T cells
  • HTL helper T cells
  • the container can alternatively hold a composition that is effective for treating, diagnosis, prognosing or prophylaxing a condition and can have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle).
  • the active agents in the composition can be an antibody capable of specifically binding 58P1D12 and modulating the function of 58P1D12.
  • the article of manufacture can further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution and/or dextrose solution. It can further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, stirrers, needles, syringes, and/or package inserts with indications and/or instructions for use.
  • a pharmaceutically-acceptable buffer such as phosphate-buffered saline, Ringer's solution and/or dextrose solution.
  • It can further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, stirrers, needles, syringes, and/or package inserts with indications and/or instructions for use.
  • SSH Suppression Subtractive Hybridization
  • the patient cancer and normal tissues were purchased from different sources such as the NDR1 (Philadelphia, Pa.).
  • mRNA for some normal tissues were purchased from different companies such as Clontech, Palo Alto, Calif.
  • Tissues were homogenized in Trizol reagent (Life Technologies, Gibco BRL) using 10 ml/g tissue to isolate total RNA.
  • Poly A RNA was purified from total RNA using Qiagen's Oligotex mRNA Mini and Midi kits. Total and mRNA were quantified by spectrophotometric analysis (O.D. 260/280 nm) and analyzed by gel electrophoresis.
  • DPNCDN (cDNA synthesis primer): (SEQ ID NO: 22) 5′TTTTGATCAAGCTT 30 3′ Adaptor 1: (SEQ ID NO: 23) 5′CTAATACGACTCACTATAGGGCTCGAGCGGCCGCCCGGGCAG3′ (SEQ ID NO: 24) 3′GGCCCGTCCTAG5′ Adaptor 2: (SEQ ID NO: 25) 5′GTAATACGACTCACTATAGGGCAGCGTGGTCGCGCGGCCGAG3′ (SEQ ID NO: 26) 3′CGGCTCCTAG5′ PCR primer 1: (SEQ ID NO: 27) 5′CTAATACGACTCACTATAGGGC3′ Nested primer (NP)1: (SEQ ID NO: 28) 5′TCGAGCGGCCGCCCGGGCAGGA3′ Nested primer (NP)2: (SEQ ID NO: 29) 5′AGCGTGGTCGCGCGCGAGGA3′
  • SSH Suppression Subtractive Hybridization
  • the gene 58P1D12 sequence was derived from the androgen dependent xenograft LAPC-9AD minus the androgen independent xenograft LAPC-9AI cDNA subtraction.
  • the SSH DNA sequence 58P1D12 ( FIG. 1 ) was identified.
  • the cDNA derived from LAPC-9AI was used as the source of the “driver” cDNA, while the cDNA from LAPC-9AD was used as the source of the “tester” cDNA.
  • Double stranded cDNAs corresponding to tester and driver cDNAs were synthesized from 2 ⁇ g of poly(A) + RNA isolated from the relevant xenograft tissue, as described above, using CLONTECH's PCR-Select cDNA Subtraction Kit and 1 ng of oligonucleotide DPNCDN as primer. First- and second-strand synthesis were carried out as described in the Kit's user manual protocol (CLONTECH Protocol No. PT1117-1, Catalog No. K1804-1). The resulting cDNA was digested with Dpn II for 3 hrs at 37° C. Digested cDNA was extracted with phenol/chloroform (1:1) and ethanol precipitated.
  • Tester cDNA was generated by diluting 1 ⁇ l of Dpn II digested cDNA from the relevant tissue source (see above) (400 ng) in 5 ⁇ l of water. The diluted cDNA (2 ⁇ l, 160 ng) was then ligated to 2 ⁇ l of Adaptor 1 and Adaptor 2 (10 ⁇ M), in separate ligation reactions, in a total volume of 10 ⁇ l at 16° C. overnight, using 400 ⁇ l of T4 DNA ligase (CLONTECH). Ligation was terminated with 1 ⁇ l of 0.2 M EDTA and heating at 72° C. for 5 min
  • the first hybridization was performed by adding 1.5 ⁇ l (600 ng) of driver cDNA to each of two tubes containing 1.5 ⁇ l (20 ng) Adaptor 1- and Adaptor 2-ligated tester cDNA. In a final volume of 4 ⁇ l, the samples were overlaid with mineral oil, denatured in an MJ Research thermal cycler at 98° C. for 1.5 minutes, and then were allowed to hybridize for 8 hrs at 68° C. The two hybridizations were then mixed together with an additional 1 ⁇ l of fresh denatured driver cDNA and were allowed to hybridize overnight at 68° C. The second hybridization was then diluted in 200 ⁇ l of 20 mM Hepes, pH 8.3, 50 mM NaCl, 0.2 mM EDTA, heated at 70° C. for 7 min. and stored at ⁇ 20° C.
  • PCR Amplification, Cloning and Sequencing of Gene Fragments Generated from SSH To amplify gene fragments resulting from SSH reactions, two PCR amplifications were performed. In the primary PCR reaction 1 ⁇ l of the diluted final hybridization mix was added to 1 ⁇ l of PCR primer 1 (10 ⁇ M), 0.5 ⁇ l dNTP mix (10 ⁇ M), 2.5 ⁇ l 10 ⁇ reaction buffer (CLONTECH) and 0.5 ⁇ l 50 ⁇ Advantage cDNA polymerase Mix (CLONTECH) in a final volume of 25 ⁇ l. PCR 1 was conducted using the following conditions: 75° C. for 5 min., 94° C. for 25 sec., then 27 cycles of 94° C. for 10 sec, 66° C.
  • PCR 2 was performed using 10-12 cycles of 94° C. for 10 sec, 68° C. for 30 sec, and 72° C. for 1.5 minutes. The PCR products were analyzed using 2% agarose gel electrophoresis.
  • PCR products were inserted into pCR2.1 using the T/A vector cloning kit (Invitrogen). Transformed E. coli were subjected to blue/white and ampicillin selection. White colonies were picked and arrayed into 96 well plates and were grown in liquid culture overnight. To identify inserts, PCR amplification was performed on 1 ⁇ l of bacterial culture using the conditions of PCR1 and NP1 and NP2 as primers. PCR products were analyzed using 2% agarose gel electrophoresis.
  • Bacterial clones were stored in 20% glycerol in a 96 well format. Plasmid DNA was prepared, sequenced, and subjected to nucleic acid homology searches of the GenBank, dBest, and NCI-CGAP databases.
  • First strand cDNAs can be generated from 1 ⁇ g of mRNA with oligo (dT)12-18 priming using the Gibco-BRL Superscript Preamplification system. The manufacturer's protocol was used which included an incubation for 50 min at 42° C. with reverse transcriptase followed by RNAse H treatment at 37° C. for 20 min. After completing the reaction, the volume can be increased to 200 ⁇ l with water prior to normalization. First strand cDNAs from 16 different normal human tissues can be obtained from Clontech.
  • Normalization of the first strand cDNAs from multiple tissues was performed by using the primers 5′ atatcgccgcgctcgtcgtcgacaa3′ (SEQ ID NO:30) and 5′ agccacacgcagetcattgtagaagg 3′ (SEQ ID NO:31) to amplify ⁇ -actin.
  • First strand cDNA (5 ⁇ l) were amplified in a total volume of 50 ⁇ l containing 0.4 ⁇ M primers, 0.2 ⁇ M each dNTPs, 1 ⁇ PCR buffer (Clontech, 10 mM Tris-HCL, 1.5 mM MgCl 2 , 50 mM KCl, pH8.3) and 1 ⁇ Klentaq DNA polymerase (Clontech).
  • Five ⁇ l of the PCR reaction can be removed at 18, 20, and 22 cycles and used for agarose gel electrophoresis.
  • PCR was performed using an MJ Research thermal cycler under the following conditions: Initial denaturation can be at 94° C. for 15 sec, followed by a 18, 20, and 22 cycles of 94° C. for 15, 65° C.
  • 58P1D12.1 5′-CCTGCTTCAGTAACAACCACATTCT-3′ (SEQ ID NO: 32)
  • 58P1D12.2 5′-CTTTACCAGTGGAATGATGACAGG-3′ (SEQ ID NO: 33)
  • FIG. 14 A typical RT-PCR expression analysis is shown in FIG. 14 .
  • the 58P1D12 SSH sequence of 427 by ( FIG. 1 ) was identified from LAPC xenograft SSH experiment.
  • a full length cDNA clone for 58P1D12 was isolated from an LAPC-9 AD library.
  • the cDNA (clone 2) is 2550 by in length and encodes a 273 amino acid ORF ( FIG. 2 and FIG. 3 ).
  • 58P1D12 v.1 exhibited 100% homology to human chondrolectin.
  • Other variants of 58P1D12 were also identified and these are listed in FIG. 2 and FIG. 3 .
  • Chromosomal localization can implicate genes in disease pathogenesis.
  • chromosome mapping approaches include fluorescent in situ hybridization (FISH), human/hamster radiation hybrid (RH) panels (Walter et al., 1994; Nature Genetics 7:22; Research Genetics, Huntsville Ala.), human-rodent somatic cell hybrid panels such as is available from the Coriell Institute (Camden, N.J.), and genomic viewers utilizing BLAST homologies to sequenced and mapped genomic clones (NCBI, Bethesda, Md.).
  • 58P1D12 expression in normal and patient cancer tissues was tested by PCR ( FIG. 14 ).
  • First strand cDNA was prepared from normal tissues (bladder, brain, heart, kidney, liver, lung, prostate, spleen, skeletal muscle, testis, pancreas, colon and stomach), and from pools of patient cancer specimens (prostate cancer pool, bladder cancer pool, lung cancer pool, ovary cancer pool, prostate cancer xenograft pool, bone and melanoma cancer pool, and cervical cancer pool). Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using primers to 58P1D12, was performed at 30 cycles of amplification. Results expression of 58P1D12 predominantly in testis amongst the normal tissues tested. 58P1D12 is also expressed in prostate cancer, bladder cancer, lung cancer, ovary cancer, prostate cancer xenograft, bone/melanoma cancer pool, and cervical cancer.
  • FIG. 15 shows expression of 58P1D12 in normal tissues by Northern blot analysis.
  • FIG. 17 shows 58P1D12 expression in normal and patient cancer specimens.
  • RNA was extracted from a pool of 3 ovary tumor patient specimens, normal prostate (NP), normal bladder (NB), normal kidney (NK), and normal colon (NC).
  • Northern blots with 10 ⁇ g of total RNA were probed with the 58P1D12 sequence. Size standards in kilobases are on the side. Results show expression of 58P1D12 in ovary tumor patient specimens but not in the normal tissues tested.
  • 58P1D12 expression was tested in a large panel of ovarian cancer patient specimens ( FIG. 19 ).
  • First strand cDNA was prepared from a panel of ovary patient cancer specimens as well as two different normal ovary tissues. Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using primers to 58P1D12, was performed at 26 and 30 cycles of amplification. Samples were run on an agarose gel, and PCR products were quantitated using the AlphaImager software. Expression was recorded as negative, weak, medium or strong. Results show expression of 58P1D12 in the majority of patient cancer specimens tested (75%), but not in normal ovary.
  • Transcript variants are variants of mature mRNA from the same gene which arise by alternative transcription or alternative splicing.
  • Alternative transcripts are transcripts from the same gene but start transcription at different points.
  • Splice variants are mRNA variants spliced differently from the same transcript.
  • a given gene can have zero to many alternative transcripts and each transcript can have zero to many splice variants.
  • Each transcript variant has a unique exon makeup, and can have different coding and/or non-coding (5′ or 3′ end) portions, from the original transcript.
  • Transcript variants can code for similar or different proteins with the same or a similar function or can encode proteins with different functions, and can be expressed in the same tissue at the same time, or in different tissues at the same time, or in the same tissue at different times, or in different tissues at different times. Proteins encoded by transcript variants can have similar or different cellular or extracellular localizations, e.g., secreted versus intracellular.
  • Transcript variants are identified by a variety of art-accepted methods. For example, alternative transcripts and splice variants are identified by full-length cloning experiment, or by use of full-length transcript and EST sequences. First, all human ESTs were grouped into clusters which show direct or indirect identity with each other. Second, ESTs in the same cluster were further grouped into sub-clusters and assembled into a consensus sequence. The original gene sequence is compared to the consensus sequence(s) or other full-length sequences. Each consensus sequence is a potential splice variant for that gene (see, e.g., the World Wide Web address doubletwist.com/products/c11_agentsOverview.jhtml). Even when a variant is identified that is not a full-length clone, that portion of the variant is very useful for antigen generation and for further cloning of the full-length splice variant, using techniques known in the art.
  • Genomic-based transcript variant identification programs include FgenesH (A. Salamov and V. Solovyev, “Ab initio gene finding in Drosophila genomic DNA,” Genome Research. 2000 April; 10(4):516-22); Grail (URL compbio.ornl.gov/Grail-bin/EmptyGrailForm) and GenScan (URL genes.mit.edu/GENSCAN.html).
  • FgenesH A. Salamov and V. Solovyev, “Ab initio gene finding in Drosophila genomic DNA,” Genome Research. 2000 April; 10(4):516-22
  • Grail URL compbio.ornl.gov/Grail-bin/EmptyGrailForm
  • GenScan URL genes.mit.edu/GENSCAN.html.
  • splice variant identification protocols see., e.g., Southan, C., A genomic perspective on human proteases, FEBS Lett. 2001 Jun. 8; 498(2-3):214-8; de Souza, S.
  • PCR-based Validation Wellmann S, et al., Specific reverse transcription-PCR quantification of vascular endothelial growth factor (VEGF) splice variants by LightCycler technology, Clin Chem. 2001 April; 47(4):654-60; Jia, H. P., et al., Discovery of new human beta-defensins using a genomics-based approach, Gene. 2001 Jan. 24; 263(1-2):211-8.
  • VEGF vascular endothelial growth factor
  • genomic regions are modulated in cancers.
  • the alternative transcripts or splice variants of the gene are modulated as well.
  • 58P1D12 has a particular expression profile related to cancer.
  • Alternative transcripts and splice variants of 58P1D12 may also be involved in cancers in the same or different tissues, thus serving as tumor-associated markers/antigens.
  • transcript variants were identified, designated as 58P1D12 v.2, v.3, v.4 and v.5.
  • the boundaries of the exon in the original transcript, 58P1D12 v.1 were shown in Table LI.
  • Exon compositions of the variants were shown in FIG. 10 . Theoretically, each different combination of exons in spatial order, e.g. exon 1 of v.2 and exons 3, 4, 5 and 6 of v.1, is a potential splice variant.
  • Tables LII(a)-(d) through LV(a)-(d) are set forth on a variant-by-variant bases.
  • LII(a)-(d) shows nucleotide sequence of the transcript variant.
  • Table LIII(a)-(d) shows the alignment of the transcript variant with nucleic acid sequence of 58P1D12 v.1.
  • LIV(a)-(d) lays out amino acid translation of the transcript variant for the identified reading frame orientation.
  • Table LV(a)-(d) displays alignments of the amino acid sequence encoded by the splice variant with that of 58P1D12 v.1.
  • a Single Nucleotide Polymorphism is a single base pair variation in a nucleotide sequence at a specific location.
  • SNP Single Nucleotide Polymorphism
  • A/T refers to the specific base pair sequence of one or more locations in the genome of an individual.
  • Haplotype refers to the base pair sequence of more than one location on the same DNA molecule (or the same chromosome in higher organisms), often in the context of one gene or in the context of several tightly linked genes.
  • SNP that occurs on a cDNA is called cSNP. This cSNP may change amino acids of the protein encoded by the gene and thus change the functions of the protein.
  • SNP and/or combinations of alleles have many applications, including diagnosis of inherited diseases, determination of drug reactions and dosage, identification of genes responsible for diseases, and analysis of the genetic relationship between individuals (P. Nowotny, J. M. Kwon and A. M. Goate, “SNP analysis to dissect human traits,” Curr. Opin. Neurobiol. 2001 October; 11(5):637-641; M. Pirmohamed and B. K. Park, “Genetic susceptibility to adverse drug reactions,” Trends Pharmacol. Sci. 2001 June; 22(6):298-305; J. H. Riley, C. J.
  • SNP are identified by a variety of art-accepted methods (P. Bean, “The promising voyage of SNP target discovery,” Am. Clin. Lab. 2001 October-November; 20(9):18-20; K. M. Weiss, “In search of human variation,” Genome Res. 1998 July; 8(7):691-697; M. M. She, “Enabling large-scale pharmacogenetic studies by high-throughput mutation detection and genotyping technologies,” Clin. Chem. 2001 February; 47(2):164-172).
  • SNP can be identified by sequencing DNA fragments that show polymorphism by gel-based methods such as restriction fragment length polymorphism (RFLP) and denaturing gradient gel electrophoresis (DGGE).
  • RFLP restriction fragment length polymorphism
  • DGGE denaturing gradient gel electrophoresis
  • FIG. 12 shows the schematic alignment of the SNP variants.
  • FIG. 11 shows the schematic alignment of protein variants, corresponding to nucleotide variants.
  • Nucleotide variants that code for the same amino acid sequence as v.1 are not shown in FIG. 11 . These alleles of the SNP, though shown separately here, can occur in different combinations (haplotypes) and in any one of the transcript variants (such as 58P1D12 v.2) that contains the site of the SNP.
  • 58P1D12 and 58P1D12 variants are cloned into any one of a variety of expression vectors known in the art.
  • One or more of the following regions of 58P1D12 variants are expressed: the full length sequence presented in FIGS. 2 and 3 , or any 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more contiguous amino acids from 58P1D12, variants, or analogs thereof.
  • pCRII To generate 58P1D12 sense and anti-sense RNA probes for RNA in situ investigations, pCRII constructs (Invitrogen, Carlsbad Calif.) are generated encoding either all or fragments of the 58P1D12 cDNA.
  • the pCRII vector has Sp6 and T7 promoters flanking the insert to drive the transcription of 58P1D12 RNA for use as probes in RNA in situ hybridization experiments. These probes are used to analyze the cell and tissue expression of 58P1D12 at the RNA level.
  • Transcribed 58P1D12 RNA representing the cDNA amino acid coding region of the 58P1D12 gene is used in in vitro translation systems such as the TnTTM Coupled Reticulolysate System (Promega, Corp., Madison, Wis.) to synthesize 58P1D12 protein.
  • TnTTM Coupled Reticulolysate System Promega, Corp., Madison, Wis.
  • pGEX Constructs To generate recombinant 58P1D12 proteins in bacteria that are fused to the Glutathione S-transferase (GST) protein, all or parts of the 58P1D12 cDNA protein coding sequence are cloned into the pGEX family of GST-fusion vectors (Amersham Pharmacia Biotech, Piscataway, N.J.).
  • GST Glutathione S-transferase
  • constructs allow controlled expression of recombinant 58P1D12 protein sequences with GST fused at the amino-terminus and a six histidine epitope (6 ⁇ His) at the carboxyl-terminus
  • the GST and 6 ⁇ His tags permit purification of the recombinant fusion protein from induced bacteria with the appropriate affinity matrix and allow recognition of the fusion protein with anti-GST and anti-His antibodies.
  • the 6 ⁇ His tag is generated by adding 6 histidine codons to the cloning primer at the 3′ end, e.g., of the open reading frame (ORF).
  • a proteolytic cleavage site such as the PreScissionTM recognition site in pGEX-6P-1, may be employed such that it permits cleavage of the GST tag from 58P1D12-related protein.
  • the ampicillin resistance gene and pBR322 origin permits selection and maintenance of the pGEX plasmids in E. coli.
  • pMAL Constructs To generate, in bacteria, recombinant 58P1D12 proteins that are fused to maltose-binding protein (MBP), all or parts of the 58P1D12 cDNA protein coding sequence are fused to the MBP gene by cloning into the pMAL-c2X and pMAL-p2X vectors (New England Biolabs, Beverly, Mass.).
  • MBP maltose-binding protein
  • constructs allow controlled expression of recombinant 58P1D12 protein sequences with MBP fused at the amino-terminus and a 6 ⁇ His epitope tag at the carboxyl-terminus
  • the MBP and 6 ⁇ His tags permit purification of the recombinant protein from induced bacteria with the appropriate affinity matrix and allow recognition of the fusion protein with anti-MBP and anti-His antibodies.
  • the 6 ⁇ His epitope tag is generated by adding 6 histidine codons to the 3′ cloning primer.
  • a Factor Xa recognition site permits cleavage of the pMAL tag from 58P1D12.
  • the pMAL-c2X and pMAL-p2X vectors are optimized to express the recombinant protein in the cytoplasm or periplasm respectively. Periplasm expression enhances folding of proteins with disulfide bonds.
  • pET Constructs To express 58P1D12 in bacterial cells, all or parts of the 58P1D12 cDNA protein coding sequence are cloned into the pET family of vectors (Novagen, Madison, Wis.). These vectors allow tightly controlled expression of recombinant 58P1D12 protein in bacteria with and without fusion to proteins that enhance solubility, such as NusA and thioredoxin (Trx), and epitope tags, such as 6 ⁇ His and S-TagTM that aid purification and detection of the recombinant protein.
  • Nrx NusA and thioredoxin
  • epitope tags such as 6 ⁇ His and S-TagTM that aid purification and detection of the recombinant protein.
  • constructs are made utilizing pET NusA fusion system 43.1 such that regions of the 58P1D12 protein are expressed as amino-terminal fusions to NusA.
  • the cDNA encoding amino acids 22-213 of 58P1D12 was cloned into the pET-21b vector, expressed, and purified from bacteria. The recombinant protein was used to generate rabbit polyclonal antibodies.
  • pESC Constructs To express 58P1D12 in the yeast species Saccharomyces cerevisiae for generation of recombinant protein and functional studies, all or parts of the 58P1D12 cDNA protein coding sequence are cloned into the pESC family of vectors each of which contain 1 of 4 selectable markers, HIS3, TRP1, LEU2, and URA3 (Stratagene, La Jolla, Calif.). These vectors allow controlled expression from the same plasmid of up to 2 different genes or cloned sequences containing either FlagTM or Myc epitope tags in the same yeast cell. This system is useful to confirm protein-protein interactions of 58P1D12. In addition, expression in yeast yields similar post-translational modifications, such as glycosylations and phosphorylations that are found when expressed in eukaryotic cells.
  • pESP Constructs To express 58P1D12 in the yeast species Saccharomyces pombe , all or parts of the 58P1D12 cDNA protein coding sequence are cloned into the pESP family of vectors. These vectors allow controlled high level of expression of a 58P1D12 protein sequence that is fused at either the amino terminus or at the carboxyl terminus to GST which aids purification of the recombinant protein. A FlagTM epitope tag allows detection of the recombinant protein with anti-FlagTM antibody.
  • 58P1D12 To express recombinant 58P1D12 in eukaryotic cells, the full or partial length 58P1D12 cDNA sequences were cloned into any one of a variety of expression vectors known in the art. One or more of the following regions of 58P1D12 were expressed in these constructs, amino acids 1 to 273 or any 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more contiguous amino acids from 58P1D12 v.1; amino acids 1 to 232 of v.2; amino acids 1 to 236 of v.4; or any 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or more contiguous amino acids from 58P1D12 variants, or analogs thereof.
  • constructs can be transfected into any one of a wide variety of mammalian cells such as 293T cells.
  • Transfected 293T cell lysates can be probed with the anti-58P1D12 polyclonal serum, described herein.
  • pcDNA4/H isMax Constructs: To express 58P1D12 in mammalian cells, a 58P1D12 ORF, or portions thereof, of 58P1D12 are cloned into pcDNA4/H isMax Version A (Invitrogen, Carlsbad, Calif.). Protein expression is driven from the cytomegalovirus (CMV) promoter and the SP16 translational enhancer. The recombinant protein has XpressTM and six histidine (6 ⁇ His) epitopes fused to the amino-terminus.
  • CMV cytomegalovirus
  • the pcDNA4/H isMax vector also contains the bovine growth hormone (BGH) polyadenylation signal and transcription termination sequence to enhance mRNA stability along with the SV40 origin for episomal replication and simple vector rescue in cell lines expressing the large T antigen.
  • BGH bovine growth hormone
  • the Zeocin resistance gene allows for selection of mammalian cells expressing the protein and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli.
  • pcDNA3.1/MycHis Constructs To express 58P1D12 in mammalian cells, a 58P1D12 ORF, or portions thereof, of 58P1D12 with a consensus Kozak translation initiation site was cloned into pcDNA3.1/MycHis Version A (Invitrogen, Carlsbad, Calif.). Protein expression is driven from the cytomegalovirus (CMV) promoter.
  • CMV cytomegalovirus
  • the recombinant proteins have the myc epitope and 6 ⁇ His epitope fused to the carboxyl-terminus
  • the pcDNA3.1/MycHis vector also contains the bovine growth hormone (BGH) polyadenylation signal and transcription termination sequence to enhance mRNA stability, along with the SV40 origin for episomal replication and simple vector rescue in cell lines expressing the large T antigen.
  • BGH bovine growth hormone
  • the Neomycin resistance gene can be used, as it allows for selection of mammalian cells expressing the protein and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli.
  • pcDNA3.1/CT-GFP-TOPO Construct To express 58P1D12 in mammalian cells and to allow detection of the recombinant proteins using fluorescence, a 58P1D12 ORF, or portions thereof, with a consensus Kozak translation initiation site are cloned into pcDNA3.1/CT-GFP-TOPO (Invitrogen, CA). Protein expression is driven from the cytomegalovirus (CMV) promoter. The recombinant proteins have the Green Fluorescent Protein (GFP) fused to the carboxyl-terminus facilitating non-invasive, in vivo detection and cell biology studies.
  • CMV cytomegalovirus
  • the pcDNA3.1CT-GFP-TOPO vector also contains the bovine growth hormone (BGH) polyadenylation signal and transcription termination sequence to enhance mRNA stability along with the SV40 origin for episomal replication and simple vector rescue in cell lines expressing the large T antigen.
  • BGH bovine growth hormone
  • the Neomycin resistance gene allows for selection of mammalian cells that express the protein and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli . Additional constructs with an amino-terminal GFP fusion are made in pcDNA3.1/NT-GFP-TOPO spanning the entire length of a 58P1D12 protein.
  • PAPtag A 58P1D12 ORF, or portions thereof, were cloned into pAPtag-5 (GenHunter Corp. Nashville, Tenn.). This construct generates an alkaline phosphatase fusion at the carboxyl-terminus of a 58P1D12 protein while fusing the IgG ⁇ signal sequence to the amino-terminus. Constructs are also generated in which alkaline phosphatase with an amino-terminal IgG ⁇ signal sequence is fused to the amino-terminus of a 58P1D12 protein.
  • the resulting recombinant 58P1D12 proteins are optimized for secretion into the media of transfected mammalian cells and can be used to identify proteins such as ligands or receptors that interact with 58P1D12 proteins.
  • Protein expression is driven from the CMV promoter and the recombinant proteins also contain myc and 6 ⁇ His epitopes fused at the carboxyl-terminus that facilitates detection and purification.
  • the Zeocin resistance gene present in the vector allows for selection of mammalian cells expressing the recombinant protein and the ampicillin resistance gene permits selection of the plasmid in E. coli.
  • pTag5 A 58P1D12 ORF, or portions thereof, were cloned into pTag-5.
  • This vector is similar to pAPtag but without the alkaline phosphatase fusion.
  • This construct generated 58P1D12 protein with an amino-terminal IgG ⁇ signal sequence and myc and 6 ⁇ His epitope tags at the carboxyl-terminus that facilitate detection and affinity purification.
  • the resulting recombinant 58P1D12 protein was optimized for secretion into the media of transfected mammalian cells, and was used as immunogen or ligand to identify proteins such as ligands or receptors that interact with the 58P1D12 proteins. Protein expression is driven from the CMV promoter.
  • the Zeocin resistance gene present in the vector allows for selection of mammalian cells expressing the protein, and the ampicillin resistance gene permits selection of the plasmid in E. coli.
  • PsecFc A 58P1D12 ORF, or portions thereof, were cloned into psecFc.
  • the psecFc vector was assembled by cloning the human immunoglobulin G1 (IgG) Fc (hinge, CH2, CH3 regions) into pSecTag2 (Invitrogen, California). This construct generated an IgG1 Fc fusion at the carboxyl-terminus of the 58P1D12 proteins, while fusing the IgGK signal sequence to N-terminus 58P1D12 fusions utilizing the murine IgG1 Fc region are also used.
  • IgG human immunoglobulin G1
  • the resulting recombinant 58P1D12 proteins are optimized for secretion into the media of transfected mammalian cells, and can be used as immunogens or to identify proteins such as ligands or receptors that interact with 58P1D12 protein. Protein expression is driven from the CMV promoter.
  • the hygromycin resistance gene present in the vector allows for selection of mammalian cells that express the recombinant protein, and the ampicillin resistance gene permits selection of the plasmid in E. coli.
  • pSR ⁇ Constructs To generate mammalian cell lines that express 58P1D12 constitutively, 58P1D12 ORF, or portions thereof, were cloned into pSR ⁇ constructs. Amphotropic and ecotropic retroviruses were generated by transfection of pSR ⁇ constructs into the 293T-10A1 packaging line or co-transfection of pSR ⁇ and a helper plasmid (containing deleted packaging sequences) into the 293 cells, respectively. The retrovirus is used to infect a variety of mammalian cell lines, resulting in the integration of the cloned gene, 58P1D12, into the host cell-lines. Protein expression is driven from a long terminal repeat (LTR).
  • LTR long terminal repeat
  • Neomycin resistance gene present in the vector allows for selection of mammalian cells that express the protein, and the ampicillin resistance gene and ColE1 origin permit selection and maintenance of the plasmid in E. coli .
  • the retroviral vectors can thereafter be used for infection and generation of various cell lines using, for example, PC3, NIH 3T3, TsuPrl, 293 or rat-1 cells.
  • Additional pSR ⁇ constructs are made that fuse an epitope tag such as the FLAG tag to the carboxyl-terminus of 58P1D12 sequences to allow detection using anti-Flag antibodies.
  • the FLAG sequence 5′ gat tac aag gat gac gac gat aag 3′ (SEQ ID NO:34) is added to cloning primer at the 3′ end of the ORF.
  • Additional pSR ⁇ constructs are made to produce both amino-terminal and carboxyl-terminal GFP and myc/6 ⁇ His fusion proteins of the full-length 58P1D12 proteins.
  • Additional Viral Vectors Additional constructs are made for viral-mediated delivery and expression of 58P1D12. High virus titer leading to high level expression of 58P1D12 is achieved in viral delivery systems such as adenoviral vectors and herpes amplicon vectors.
  • a 58P1D12 coding sequence or fragments thereof are amplified by PCR and subcloned into the AdEasy shuttle vector (Stratagene). Recombination and virus packaging are performed according to the manufacturer's instructions to generate adenoviral vectors.
  • 58P1D12 coding sequences or fragments thereof are cloned into the HSV-1 vector (Imgenex) to generate herpes viral vectors.
  • the viral vectors are thereafter used for infection of various cell lines such as PC3, NIH 3T3, 293 or rat-1 cells.
  • coding sequences of 58P1D12, or portions thereof are cloned into regulated mammalian expression systems such as the T-Rex System (Invitrogen), the GeneSwitch System (Invitrogen) and the tightly-regulated Ecdysone System (Sratagene). These systems allow the study of the temporal and concentration dependent effects of recombinant 58P1D12. These vectors are thereafter used to control expression of 58P1D12 in various cell lines such as PC3, NIH 3T3, 293 or rat-1 cells.
  • regulated mammalian expression systems such as the T-Rex System (Invitrogen), the GeneSwitch System (Invitrogen) and the tightly-regulated Ecdysone System (Sratagene). These systems allow the study of the temporal and concentration dependent effects of recombinant 58P1D12. These vectors are thereafter used to control expression of 58P1D12 in various cell lines such as PC3, NIH 3T3, 293 or rat-1 cells.
  • 58P1D12 ORF To generate recombinant 58P1D12 proteins in a baculovirus expression system, 58P1D12 ORF, or portions thereof, are cloned into the baculovirus transfer vector pBlueBac 4.5 (Invitrogen), which provides a His-tag at the N-terminus.
  • pBlueBac-58P1D12 is co-transfected with helper plasmid pBac-N-Blue (Invitrogen) into SF9 ( Spodoptera frugiperda ) insect cells to generate recombinant baculovirus (see Invitrogen instruction manual for details). Baculovirus is then collected from cell supernatant and purified by plaque assay.
  • Recombinant 58P1D12 protein is then generated by infection of HighFive insect cells (Invitrogen) with purified baculovirus.
  • Recombinant 58P1D12 protein can be detected using anti-58P1D12 or anti-His-tag antibody.
  • 58P1D12 protein can be purified and used in various cell-based assays or as immunogen to generate polyclonal and monoclonal antibodies specific for 58P1D12.
  • FIG. 5A-C , FIG. 6A-C , FIG. 7A-C , FIG. 8A-C , and FIG. 9A-C depict graphically five amino acid profiles of 58P1D12 variant 1, variant 2, and variant 3, A through C respectively, each assessment available by accessing the ProtScale website (at the World Wide Web address expasy.ch/cgi-bin/protscale.pl) on the ExPasy molecular biology server.
  • FIG. 5 Hydrophilicity, (Hopp T. P., Woods K. R., 1981. Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828);
  • FIG. 6 Hydropathicity, (Kyte J., Doolittle R.F., 1982. J. Mol. Biol. 157:105-132);
  • FIG. 7 Percentage Accessible Residues (Janin J., 1979 Nature 277:491-492);
  • FIG. 8 Average Flexibility, (Bhaskaran R., and Ponnuswamy P.K., 1988. Int. J. Pept. Protein Res. 32:242-255);
  • FIG. 5 Hydrophilicity, (Hopp T. P., Woods K. R., 1981. Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828);
  • FIG. 6 Hydropathicity, (Kyte J., Doolittle R.F., 1982. J
  • Beta-turn (Deleage, G., Roux B. 1987 Protein Engineering 1:289-294); and optionally others available in the art, such as on the ProtScale website, were used to identify antigenic regions of each of the 58P1D12 variant proteins.
  • Each of the above amino acid profiles of 58P1D12 variants were generated using the following ProtScale parameters for analysis: 1) A window size of 9; 2) 100% weight of the window edges compared to the window center; and, 3) amino acid profile values normalized to lie between 0 and 1.
  • Hydrophilicity ( FIG. 5 ), Hydropathicity ( FIG. 6 ) and Percentage Accessible Residues ( FIG. 7 ) profiles were used to determine stretches of hydrophilic amino acids (i.e., values greater than 0.5 on the Hydrophilicity and Percentage Accessible Residues profile, and values less than 0.5 on the Hydropathicity profile). Such regions are likely to be exposed to the aqueous environment, be present on the surface of the protein, and thus available for immune recognition, such as by antibodies.
  • Average Flexibility ( FIG. 8 ) and Beta-turn ( FIG. 9 ) profiles determine stretches of amino acids (i.e., values greater than 0.5 on the Beta-turn profile and the Average Flexibility profile) that are not constrained in secondary structures such as beta sheets and alpha helices. Such regions are also more likely to be exposed on the protein and thus accessible to immune recognition, such as by antibodies.
  • Antigenic sequences of the 58P1D12 variant proteins indicated, e.g., by the profiles set forth in FIG. 5 , FIG. 6 , FIG. 7 , FIG. 8 , and/or FIG. 9 are used to prepare immunogens, either peptides or nucleic acids that encode them, to generate therapeutic and diagnostic anti-58P1D12 antibodies.
  • the immunogen can be any 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50 or more than 50 contiguous amino acids, or the corresponding nucleic acids that encode them, from the 58P1D12 protein variants listed in FIGS. 2 and 3 .
  • peptide immunogens of the invention can comprise, a peptide region of at least 5 amino acids of FIGS. 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profiles of FIG. 5 ; a peptide region of at least 5 amino acids of FIGS. 2 and 3 in any whole number increment that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile of FIG. 6 ; a peptide region of at least 5 amino acids of FIGS. 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profiles of FIG. 7 ; a peptide region of at least 5 amino acids of FIGS.
  • Peptide immunogens of the invention can also comprise nucleic acids that encode any of the forgoing.
  • All immunogens of the invention, peptide or nucleic acid can be embodied in human unit dose form, or comprised by a composition that includes a pharmaceutical excipient compatible with human physiology.
  • the secondary structures of 58P1D12 protein variants 1, 2, and 3, namely the predicted presence and location of alpha helices, extended strands, and random coils, are predicted from their primary amino acid sequences using the HNN—Hierarchical Neural Network method (NPS@: Network Protein Sequence Analysis TIBS 2000 March Vol. 25, No 3 [291]:147-150 Combet C., Blanchet C., Geourjon C.
  • NPS@ Network Protein Sequence Analysis TIBS 2000 March Vol. 25, No 3 [291]:147-150 Combet C., Blanchet C., Geourjon C.
  • 58P1D12 variant 1 is composed of 24.18% alpha helix, 18.68% extended strand, and 57.14% random coil ( FIG. 13A ).
  • 58P1D12 variant 2 is composed of 19.83% alpha helix, 18.97% extended strand, and 61.21% random coil ( FIG. 13B ).
  • 58P1D12 variant 3 is composed of 32.20% alpha helix, 15.25% extended strand, and 52.54% random coil ( FIG. 13C ).
  • Polyclonal antibodies can be raised in a mammal, for example, by one or more injections of an immunizing agent and, if desired, an adjuvant.
  • the immunizing agent and/or adjuvant will be injected in the mammal by multiple subcutaneous or intraperitoneal injections.
  • computer algorithms are employed in design of immunogens that, based on amino acid sequence analysis contain characteristics of being antigenic and available for recognition by the immune system of the immunized host (see the Example entitled “Antigenicity Profiles and Secondary Structure”). Such regions would be predicted to be hydrophilic, flexible, in beta-turn conformations, and be exposed on the surface of the protein (see, e.g., FIG. 5 , FIG. 6 , FIG. 7 , FIG. 8 , or FIG. 9 for amino acid profiles that indicate such regions of 58P1D12 protein variant 1).
  • recombinant bacterial fusion proteins or peptides containing hydrophilic, flexible, beta-turn regions of 58P1D12 protein variants are used as antigens to generate polyclonal antibodies in New Zealand White rabbits or monoclonal antibodies as described in Example 11.
  • such regions include, but are not limited to, amino acids 19-30, amino acids 49-66, amino acids 70-82, amino acids 88-115, amino acids 131-145, amino acids 165-203, amino acids 243-255, and amino acids 262-273. It is useful to conjugate the immunizing agent to a protein known to be immunogenic in the mammal being immunized.
  • immunogenic proteins examples include, but are not limited to, keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor.
  • KLH keyhole limpet hemocyanin
  • serum albumin serum albumin
  • bovine thyroglobulin bovine thyroglobulin
  • soybean trypsin inhibitor soybean trypsin inhibitor.
  • a peptide encoding amino acids 102-115 of 58P1D12 variant 1 was conjugated to KLH and used to immunize a rabbit.
  • the immunizing agent may include all or portions of the 58P1D12 variant proteins, analogs or fusion proteins thereof.
  • the 58P1D12 variant 1 amino acid sequence can be fused using recombinant DNA techniques to any one of a variety of fusion protein partners that are well known in the art, such as glutathione-S-transferase (GST) and HIS tagged fusion proteins.
  • GST glutathione-S-transferase
  • amino acids 22-213 of 58P1D12 variant 1 is fused to H is using recombinant techniques and the pET21b expression vector. The protein was then expressed, purified, and used to immunize 2 rabbits.
  • Such fusion proteins are purified from induced bacteria using the appropriate affinity matrix.
  • recombinant bacterial fusion proteins that may be employed include maltose binding protein, LacZ, thioredoxin, NusA, or an immunoglobulin constant region (see the section entitled “Production of 58P1D12 in Prokaryotic Systems” and Current Protocols In Molecular Biology, Volume 2, Unit 16, Frederick M. Ausubul et al. eds., 1995; Linsley, P.S., Brady, W., Urnes, M., Grosmaire, L., Damle, N., and Ledbetter, J. (1991) J. Exp. Med. 174, 561-566).
  • mammalian expressed protein antigens are also used. These antigens are expressed from mammalian expression vectors such as the Tag5 and Fc-fusion vectors (see the section entitled “Production of Recombinant 58P1D12 in Eukaryotic Systems”), and retain post-translational modifications such as glycosylations found in native protein.
  • amino acids 22-213 of 58P1D12 variant 1 was cloned into the Tag5 mammalian secretion vector, and expressed in 293T cells.
  • the recombinant protein was purified by metal chelate chromatography from tissue culture supernatants of 293T cells stably expressing the recombinant vector.
  • the purified Tag5 58P1D12 protein was then used as immunogen.
  • adjuvants include, but are not limited to, complete Freund's adjuvant (CFA) and MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).
  • CFA complete Freund's adjuvant
  • MPL-TDM adjuvant monophosphoryl Lipid A, synthetic trehalose dicorynomycolate
  • rabbits are initially immunized subcutaneously with up to 200 ⁇ g, typically 100-200 ⁇ g, of fusion protein or peptide conjugated to KLH mixed in complete Freund's adjuvant (CFA). Rabbits are then injected subcutaneously every two weeks with up to 200 ⁇ g, typically 100-200 ⁇ g, of the immunogen in incomplete Freund's adjuvant (IFA). Test bleeds are taken approximately 7-10 days following each immunization and used to monitor the titer of the antiserum by ELISA.
  • CFA complete Freund's adjuvant
  • 58P1D12 variant 1 cDNA was cloned into pcDNA 3.1 myc-his expression vector (Invitrogen, see the Example entitled “Production of Recombinant 58P1D12 in Eukaryotic Systems”). After transfection of the constructs into 293T cells, cell lysates are probed with the anti-58P1D12 serum and with anti-His antibody (Santa Cruz Biotechnologies, Santa Cruz, Calif.) to determine specific reactivity to denatured 58P1D12 protein using the Western blot technique.
  • the immune serum is tested by fluorescence microscopy, flow cytometry and immunoprecipitation against 293T and other recombinant 58P1D12-expressing cells to determine specific recognition of native protein.
  • Western blot, immunoprecipitation, fluorescent microscopy, and flow cytometric techniques using cells that endogenously express 58P1D12 are also carried out to test reactivity and specificity.
  • Anti-serum from rabbits immunized with 58P1D12 variant fusion proteins are purified by depletion of antibodies reactive to the fusion partner sequence by passage over an affinity column containing the fusion partner either alone or in the context of an irrelevant fusion protein.
  • antiserum derived from a GST-58P1D12 variant 1 fusion protein is first purified by passage over a column of GST protein covalently coupled to AffiGel matrix (BioRad, Hercules, Calif.). The antiserum is then affinity purified by passage over a column composed of a MBP-58P1D12 fusion protein covalently coupled to Affigel matrix.
  • the serum is then further purified by protein G affinity chromatography to isolate the IgG fraction.
  • Sera from other His-tagged antigens and peptide immunized rabbits as well as fusion partner depleted sera are affinity purified by passage over a column matrix composed of the original protein immunogen or free peptide.
  • therapeutic mAbs to 58P1D12 variants comprise those that react with epitopes specific for each variant protein or specific to sequences in common between the variants that would disrupt or modulate the biological function of the 58P1D12 variants, for example those that would disrupt the interaction with ligands and binding partners
  • Immunogens for generation of such mAbs include those designed to encode or contain the entire 58P1D12 protein variant sequence, regions predicted to contain functional motifs, and regions of the 58P1D12 protein variants predicted to be antigenic from computer analysis of the amino acid sequence (see, e.g., FIG. 5 , FIG. 6 , FIG. 7 , FIG. 8 , or FIG.
  • Immunogens include peptides, recombinant bacterial proteins, and mammalian expressed Tag 5 proteins and human and murine IgG FC fusion proteins.
  • cells engineered to express high levels of a respective 58P1D12 variant such as Rat1-58P1D12 variant 1 or 300.19-58P1D12 variant lmurine Pre-B cells, were used to immunize mice.
  • mice are first immunized intraperitoneally (IP) with, typically, 10-50 ⁇ g of protein immunogen or 10 7 58P1D12-expressing cells mixed in complete Freund's adjuvant. Mice are then subsequently immunized IP every 2-4 weeks with, typically, 10-50 ⁇ g of protein immunogen or 10 7 cells mixed in incomplete Freund's adjuvant.
  • IP intraperitoneally
  • MPL-TDM adjuvant is used in immunizations.
  • a DNA-based immunization protocol is employed in which a mammalian expression vector encoding a 58P1D12 variant sequence is used to immunize mice by direct injection of the plasmid DNA.
  • amino acids 22-213 of 58P1D12 of variant 1 is cloned into the Tag5 mammalian secretion vector and the recombinant vector will then be used as immunogen.
  • the same amino acids are cloned into an Fc-fusion secretion vector in which the 58P1D12 variant 1 sequence is fused at the amino-terminus to an IgK leader sequence and at the carboxyl-terminus to the coding sequence of the human or murine IgG Fc region.
  • This recombinant vector is then used as immunogen.
  • the plasmid immunization protocols are used in combination with purified proteins expressed from the same vector and with cells expressing the respective 58P1D12 variant.
  • test bleeds are taken 7-10 days following an injection to monitor titer and specificity of the immune response. Once appropriate reactivity and specificity is obtained as determined by ELISA, Western blotting, immunoprecipitation, fluorescence microscopy, and flow cytometric analyses, fusion and hybridoma generation is then carried out with established procedures well known in the art (see, e.g., Harlow and Lane, 1988).
  • amino acids 22-213 of 58P1D12 variant 1 was cloned into the pTag5 vector, the protein was then expressed in 293T cells and purified by metal chelate chromatography.
  • Balb C mice were immunized with 10 ⁇ g of recombinant variant 1 protein mixed in adjuvant. Mice were subsequently immunized over several weeks with the protein.
  • ELISA using the antigen determined the titer of serum from the immunized mice. Reactivity and specificity of the serum to full length 58P1D12 variant 1 protein was monitored by flow cytometry using Rat-58P1D12 cells compared to Rat1-neo control cells ( FIG. 20 ).
  • 58P1D12 variant 1-expressing cells or cells endogenously expressing 58P1D12 variant 1 are also used. Mice showing the strongest reactivity were rested and given a final injection of antigen and sacrificed for fusion. The lymph nodes of the sacrificed mice were harvested and fused to SP2/0 myeloma cells using standard procedures (Harlow and Lane, 1988). Supernatants from HAT selected growth wells were analyzed by flow cytometry to identify specific-58P1D12 surface binding MAbs ( FIG. 21 ). Supernatants are also be screened by ELISA, Western blot, immunoprecipitation, and fluorescent microscopy to identify 58P1D12 specific antibody-producing clones.
  • 58P1D12 variant 3 specific MAbs are generated by employing immunogens that encode amino acid sequences unique to variant 3.
  • immunogens that encode amino acid sequences unique to variant 3.
  • a GST-fusion protein is constructed to encode amino acids 171-236, expressed, purified, and used to immunize mice. Hybridomas resulting from fusion of the B-cells from the mice are screened on cells expressing 58P1D12 variant 3 protein and cross-screened on cells expressing variant 1 protein to identify variant 3-specific MAbs.
  • the binding affinity of 58P1D12 variant specific monoclonal antibodies are determined using standard technologies. Affinity measurements quantify the strength of antibody to epitope binding and are used to help define which 58P1D12 variant monoclonal antibodies preferred for diagnostic or therapeutic use, as appreciated by one of skill in the art.
  • the BIAcore system (Uppsala, Sweden) is a preferred method for determining binding affinity. The BIAcore system uses surface plasmon resonance (SPR, Welford K. 1991, Opt. Quant. Elect. 23:1; Morton and Myszka, 1998, Methods in Enzymology 295: 268) to monitor biomolecular interactions in real time.
  • BIAcore analysis conveniently generates association rate constants, dissociation rate constants, equilibrium dissociation constants, and affinity constants.
  • equilibrium binding analysis of a dilution series of the MAb is also used to determine affinity defined by the dissociation constant (KD).
  • KD is determined by non-linear regression of the equilibrium binding data of the concentration series.
  • the KD is defined as the concentration at which half-maximal binding of the MAb to the antigen is attained.
  • HLA class I and class II binding assays using purified HLA molecules are performed in accordance with disclosed protocols (e.g., PCT publications WO 94/20127 and WO 94/03205; Sidney et al., Current Protocols in Immunology 18.3.1 (1998); Sidney, et al., J. Immunol. 154:247 (1995); Sette, et al., Mol. Immunol. 31:813 (1994)). Briefly, purified MHC molecules (5 to 500 nM) are incubated with various unlabeled peptide inhibitors and 1-10 nM 125I-radiolabeled probe peptides as described.
  • MHC-peptide complexes are separated from free peptide by gel filtration and the fraction of peptide bound is determined.
  • each MHC preparation is titered in the presence of fixed amounts of radiolabeled peptides to determine the concentration of HLA molecules necessary to bind 10-20% of the total radioactivity. All subsequent inhibition and direct binding assays are performed using these HLA concentrations.
  • Binding assays as outlined above may be used to analyze HLA supermotif and/or HLA motif-bearing peptides (see Table IV).
  • HLA vaccine compositions of the invention can include multiple epitopes.
  • the multiple epitopes can comprise multiple HLA supermotifs or motifs to achieve broad population coverage. This example illustrates the identification and confirmation of supermotif- and motif-bearing epitopes for the inclusion in such a vaccine composition. Calculation of population coverage is performed using the strategy described below.
  • Identified A2-, A3-, and DR-supermotif sequences are scored using polynomial algorithms to predict their capacity to bind to specific HLA-Class I or Class II molecules. These polynomial algorithms account for the impact of different amino acids at different positions, and are essentially based on the premise that the overall affinity (or AG) of peptide-HLA molecule interactions can be approximated as a linear polynomial function of the type:
  • aji is a coefficient which represents the effect of the presence of a given amino acid (j) at a given position (i) along the sequence of a peptide of n amino acids.
  • the crucial assumption of this method is that the effects at each position are essentially independent of each other (i.e., independent binding of individual side-chains).
  • residue j occurs at position i in the peptide, it is assumed to contribute a constant amount ji to the free energy of binding of the peptide irrespective of the sequence of the rest of the peptide.
  • the ARB values corresponding to the sequence of the peptide are multiplied. If this product exceeds a chosen threshold, the peptide is predicted to bind. Appropriate thresholds are chosen as a function of the degree of stringency of prediction desired.
  • Protein sequences from 58P1D12 are scanned utilizing motif identification software, to identify 8-, 9-10- and 11-mer sequences containing the HLA-A2-supermotif main anchor specificity. Typically, these sequences are then scored using the protocol described above and the peptides corresponding to the positive-scoring sequences are synthesized and tested for their capacity to bind purified HLA-A*0201 molecules in vitro (HLA-A*0201 is considered a prototype A2 supertype molecule).
  • A2-supertype molecules A*0202, A*0203, A*0206, and A*6802.
  • Peptides that bind to at least three of the five A2-supertype alleles tested are typically deemed A2-supertype cross-reactive binders.
  • Preferred peptides bind at an affinity equal to or less than 500 nM to three or more HLA-A2 supertype molecules.
  • the 58P1D12 protein sequence(s) scanned above is also examined for the presence of peptides with the HLA-A3-supermotif primary anchors. Peptides corresponding to the HLA A3 supermotif-bearing sequences are then synthesized and tested for binding to HLA-A*0301 and HLA-A*1101 molecules, the molecules encoded by the two most prevalent A3-supertype alleles.
  • the peptides that bind at least one of the two alleles with binding affinities of 500 nM, often 200 nM, are then tested for binding cross-reactivity to the other common A3-supertype alleles (e.g., A*3101, A*3301, and A*6801) to identify those that can bind at least three of the five HLA-A3-supertype molecules tested.
  • A3-supertype alleles e.g., A*3101, A*3301, and A*6801
  • the 58P1D12 protein(s) scanned above is also analyzed for the presence of 8-, 9-10-, or 11-mer peptides with the HLA-B7-supermotif.
  • Corresponding peptides are synthesized and tested for binding to HLA-B*0702, the molecule encoded by the most common B7-supertype allele (i.e., the prototype B7 supertype allele).
  • Peptides binding B*0702 with IC50 of ⁇ 500 nM are identified using standard methods. These peptides are then tested for binding to other common B7-supertype molecules (e.g., B*3501, B*5101, B*5301, and B*5401). Peptides capable of binding to three or more of the five B7-supertype alleles tested are thereby identified.
  • HLA-A1 and -A24 epitopes can also be incorporated into vaccine compositions.
  • An analysis of the 58P1D12 protein can also be performed to identify HLA-A1- and A24-motif-containing sequences.
  • Cross-reactive candidate CTL A2-supermotif-bearing peptides that are identified as described herein are selected to confirm in vitro immunogenicity. Confirmation is performed using the following methodology:
  • the 0.221A2.1 cell line produced by transferring the HLA-A2.1 gene into the HLA-A, -B, -C null mutant human B-lymphoblastoid cell line 721.221, is used as the peptide-loaded target to measure activity of HLA-A2.1-restricted CTL.
  • This cell line is grown in RPMI-1640 medium supplemented with antibiotics, sodium pyruvate, nonessential amino acids and 10% (v/v) heat inactivated FCS.
  • Cells that express an antigen of interest, or transfectants comprising the gene encoding the antigen of interest can be used as target cells to confirm the ability of peptide-specific CTLs to recognize endogenous antigen.
  • DC Dendritic Cells
  • the wells are washed a total of three times with 3 ml RPMI to remove most of the non-adherent and loosely adherent cells.
  • Three ml of complete medium containing 50 ng/ml of GM-CSF and 1,000 U/ml of IL-4 are then added to each well.
  • TNF ⁇ is added to the DCs on day 6 at 75 ng/ml and the cells are used for CTL induction cultures on day 7.
  • CD8+ T-cells are isolated by positive selection with Dynal immunomagnetic beads (Dynabeads® M-450) and the Detacha-bead® reagent. Typically about 200 ⁇ 250 ⁇ 106 PBMC are processed to obtain 24 ⁇ 106 CD8+ T-cells (enough for a 48-well plate culture). Briefly, the PBMCs are thawed in RPMI with 30 ⁇ g/ml DNAse, washed once with PBS containing 1% human AB serum and resuspended in PBS/1% AB serum at a concentration of 20 ⁇ 106 cells/ml.
  • the magnetic beads are washed 3 times with PBS/AB serum, added to the cells (140 ⁇ l beads/20 ⁇ 106 cells) and incubated for 1 hour at 4° C. with continuous mixing.
  • the beads and cells are washed 4 ⁇ with PBS/AB serum to remove the nonadherent cells and resuspended at 100 ⁇ 106 cells/ml (based on the original cell number) in PBS/AB serum containing 100 ⁇ l/ml Detacha-bead® reagent and 30 ⁇ g/ml DNAse.
  • the mixture is incubated for 1 hour at room temperature with continuous mixing.
  • the beads are washed again with PBS/AB/DNAse to collect the CD8+ T-cells.
  • the DC are collected and centrifuged at 1300 rpm for 5-7 minutes, washed once with PBS with 1% BSA, counted and pulsed with 40 ⁇ g/ml of peptide at a cell concentration of 1-2 ⁇ 106/ml in the presence of 3 ⁇ g/ml ⁇ 2-microglobulin for 4 hours at 20° C.
  • the DC are then irradiated (4,200 rads), washed 1 time with medium and counted again.
  • cytokine-generated DC at 1 ⁇ 105 cells/ml
  • CD8+ T-cells at 2 ⁇ 106 cell/ml
  • Recombinant human IL-10 is added the next day at a final concentration of 10 ng/ml and rhuman IL-2 is added 48 hours later at 10 IU/ml.
  • PBMCs are thawed and washed twice with RPMI and DNAse. The cells are resuspended at 5 ⁇ 106 cells/ml and irradiated at ⁇ 4200 rads. The PBMCs are plated at 2 ⁇ 106 in 0.5 ml complete medium per well and incubated for 2 hours at 37° C.
  • the plates are washed twice with RPMI by tapping the plate gently to remove the nonadherent cells and the adherent cells pulsed with 10 ⁇ g/ml of peptide in the presence of 3 ⁇ g/ml 132 microglobulin in 0.25 ml RPMI/5% AB per well for 2 hours at 37° C.
  • Peptide solution from each well is aspirated and the wells are washed once with RPMI. Most of the media is aspirated from the induction cultures (CD8+ cells) and brought to 0.5 ml with fresh media. The cells are then transferred to the wells containing the peptide-pulsed adherent cells.
  • recombinant human IL-10 is added at a final concentration of 10 ng/ml and recombinant human IL2 is added the next day and again 2-3 days later at 50 IU/ml (Tsai et al., Critical Reviews in Immunology 18(1-2):65-75, 1998). Seven days later, the cultures are assayed for CTL activity in a 51Cr release assay. In some experiments the cultures are assayed for peptide-specific recognition in the in situ IFN ⁇ ELISA at the time of the second restimulation followed by assay of endogenous recognition 7 days later. After expansion, activity is measured in both assays for a side-by-side comparison.
  • cytotoxicity is determined in a standard (5 hr) 51Cr release assay by assaying individual wells at a single E:T.
  • Peptide-pulsed targets are prepared by incubating the cells with 10 ⁇ g/ml peptide overnight at 37° C.
  • Adherent target cells are removed from culture flasks with trypsin-EDTA.
  • Target cells are labeled with 200 ⁇ Ci of 51Cr sodium chromate (Dupont, Wilmington, Del.) for 1 hour at 37° C.
  • Labeled target cells are resuspended at 106 per ml and diluted 1:10 with K562 cells at a concentration of 3.3 ⁇ 106/ml (an NK-sensitive erythroblastoma cell line used to reduce non-specific lysis).
  • Target cells (100 ⁇ l) and effectors (100 ⁇ l) are plated in 96 well round-bottom plates and incubated for 5 hours at 37° C. At that time, 100 ⁇ l of supernatant are collected from each well and percent lysis is determined according to the formula:
  • a positive culture is defined as one in which the specific lysis (sample-background) is 10% or higher in the case of individual wells and is 15% or more at the two highest E:T ratios when expanded cultures are assayed.
  • Immulon 2 plates are coated with mouse anti-human IFN ⁇ monoclonal antibody (4 ⁇ g/ml 0.1M NaHCO 3 , pH8.2) overnight at 4° C.
  • the plates are washed with Ca2+, Mg2+-free PBS/0.05% Tween 20 and blocked with PBS/10% FCS for two hours, after which the CTLs (100 ⁇ l/well) and targets (100 ⁇ l/well) are added to each well, leaving empty wells for the standards and blanks (which received media only).
  • the target cells either peptide-pulsed or endogenous targets, are used at a concentration of 1 ⁇ 106 cells/ml.
  • the plates are incubated for 48 hours at 37° C. with 5% CO2.
  • Recombinant human IFN-gamma is added to the standard wells starting at 400 pg or 1200 pg/100 microliter/well and the plate incubated for two hours at 37° C.
  • the plates are washed and 100 ⁇ l of biotinylated mouse anti-human IFN-gamma monoclonal antibody (2 microgram/ml in PBS/3% FCS/0.05% Tween 20) are added and incubated for 2 hours at room temperature. After washing again, 100 microliter HRP-streptavidin (1:4000) are added and the plates incubated for one hour at room temperature.
  • the plates are then washed 6 ⁇ with wash buffer, 100 microliter/well developing solution (TMB 1:1) are added, and the plates allowed to develop for 5-15 minutes.
  • TMB 1:1 microliter/well developing solution
  • the reaction is stopped with 50 microliter/well 1M H3PO4 and read at OD450.
  • a culture is considered positive if it measured at least 50 pg of IFN-gam
  • PBMC autologous or allogeneic
  • EBV-transformed cells 5 ⁇ 104 CD8+ cells are added to a T25 flask containing the following: 1 ⁇ 106 irradiated (4,200 rad) PBMC (autologous or allogeneic) per ml, 2 ⁇ 105 irradiated (8,000 rad) EBV-transformed cells per ml, and OKT3 (anti-CD3) at 30 ng per ml in RPMI-1640 containing 10% (v/v) human AB serum, non-essential amino acids, sodium pyruvate, 25 ⁇ M 2-mercaptoethanol, L-glutamine and penicillin/streptomycin.
  • Recombinant human IL2 is added 24 hours later at a final concentration of 200 IU/ml and every three days thereafter with fresh media at 50 IU/ml.
  • the cells are split if the cell concentration exceeds 1 ⁇ 106/ml and the cultures are assayed between days 13 and 15 at E:T ratios of 30, 10, 3 and 1:1 in the 51Cr release assay or at 1 ⁇ 106/ml in the in situ IFN ⁇ assay using the same targets as before the expansion.
  • Cultures are expanded in the absence of anti-CD3+ as follows. Those cultures that demonstrate specific lytic activity against peptide and endogenous targets are selected and 5 ⁇ 104 CD8+ cells are added to a T25 flask containing the following: 1 ⁇ 106 autologous PBMC per ml which have been peptide-pulsed with 10 ⁇ g/ml peptide for two hours at 37° C. and irradiated (4,200 rad); 2 ⁇ 105 irradiated (8,000 rad) EBV-transformed cells per ml RPMI-1640 containing 10% (v/v) human AB serum, non-essential AA, sodium pyruvate, 25 mM 2-ME, L-glutamine and gentamicin.
  • A2-supermotif cross-reactive binding peptides are tested in the cellular assay for the ability to induce peptide-specific CTL in normal individuals.
  • a peptide is typically considered to be an epitope if it induces peptide-specific CTLs in at least individuals, and preferably, also recognizes the endogenously expressed peptide.
  • PBMCs isolated from patients bearing a tumor that expresses 58P1D12. Briefly, PBMCs are isolated from patients, re-stimulated with peptide-pulsed monocytes and assayed for the ability to recognize peptide-pulsed target cells as well as transfected cells endogenously expressing the antigen.
  • HLA-A3 supermotif-bearing cross-reactive binding peptides are also evaluated for immunogenicity using methodology analogous for that used to evaluate the immunogenicity of the HLA-A2 supermotif peptides.
  • HLA motifs and supermotifs are useful in the identification and preparation of highly cross-reactive native peptides, as demonstrated herein.
  • the definition of HLA motifs and supermotifs also allows one to engineer highly cross-reactive epitopes by identifying residues within a native peptide sequence which can be analoged to confer upon the peptide certain characteristics, e.g. greater cross-reactivity within the group of HLA molecules that comprise a supertype, and/or greater binding affinity for some or all of those HLA molecules. Examples of analoging peptides to exhibit modulated binding affinity are set forth in this example.
  • Peptide engineering strategies are implemented to further increase the cross-reactivity of the epitopes.
  • the main anchors of A2-supermotif-bearing peptides are altered, for example, to introduce a preferred L, I, V, or M at position 2, and I or V at the C-terminus.
  • each engineered analog is initially tested for binding to the prototype A2 supertype allele A*0201, then, if A*0201 binding capacity is maintained, for A2-supertype cross-reactivity.
  • a peptide is confirmed as binding one or all supertype members and then analoged to modulate binding affinity to any one (or more) of the supertype members to add population coverage.
  • the selection of analogs for immunogenicity in a cellular screening analysis is typically further restricted by the capacity of the parent wild type (WT) peptide to bind at least weakly, i.e., bind at an IC50 of 5000 nM or less, to three of more A2 supertype alleles.
  • WT wild type
  • the rationale for this requirement is that the WT peptides must be present endogenously in sufficient quantity to be biologically relevant.
  • Analoged peptides have been shown to have increased immunogenicity and cross-reactivity by T cells specific for the parent epitope (see, e.g., Parkhurst et al., J. Immunol. 157:2539, 1996; and Pogue et al., Proc. Natl. Acad. Sci. USA 92:8166, 1995).
  • analog-specific CTLs are also able to recognize the wild-type peptide and, when possible, target cells that endogenously express the epitope.
  • Analogs of HLA-A3 supermotif-bearing epitopes are generated using strategies similar to those employed in analoging HLA-A2 supermotif-bearing peptides. For example, peptides binding to 3/5 of the A3-supertype molecules are engineered at primary anchor residues to possess a preferred residue (V, S, M, or A) at position 2.
  • analog peptides are then tested for the ability to bind A*03 and A*11 (prototype A3 supertype alleles). Those peptides that demonstrate 500 nM binding capacity are then confirmed as having A3-supertype cross-reactivity.
  • B7 supermotif-bearing peptides are, for example, engineered to possess a preferred residue (V, I, L, or F) at the C-terminal primary anchor position, as demonstrated by Sidney et al. (J. Immunol. 157:3480-3490, 1996).
  • analog-specific CTLs are also able to recognize the wild-type peptide and, when possible, targets that endogenously express the epitope.
  • HLA supermotifs are of value in engineering highly cross-reactive peptides and/or peptides that bind HLA molecules with increased affinity by identifying particular residues at secondary anchor positions that are associated with such properties. For example, the binding capacity of a B7 supermotif-bearing peptide with an F residue at position 1 is analyzed. The peptide is then analoged to, for example, substitute L for F at position 1. The analoged peptide is evaluated for increased binding affinity, binding half life and/or increased cross-reactivity. Such a procedure identifies analoged peptides with enhanced properties.
  • Engineered analogs with sufficiently improved binding capacity or cross-reactivity can also be tested for immunogenicity in HLA-B7-transgenic mice, following for example, IFA immunization or lipopeptide immunization.
  • Analoged peptides are additionally tested for the ability to stimulate a recall response using PBMC from patients with 58P1D12-expressing tumors.
  • cysteine Another form of peptide analoging, unrelated to anchor positions, involves the substitution of a cysteine with ⁇ -amino butyric acid. Due to its chemical nature, cysteine has the propensity to form disulfide bridges and sufficiently alter the peptide structurally so as to reduce binding capacity. Substitution of ⁇ -amino butyric acid for cysteine not only alleviates this problem, but has been shown to improve binding and crossbinding capabilities in some instances (see, e.g., the review by Sette et al., In: Persistent Viral Infections, Eds. R. Ahmed and I. Chen, John Wiley & Sons, England, 1999).
  • the binding properties and/or cross-reactivity of peptide ligands for HLA supertype molecules can be modulated.
  • Peptide epitopes bearing an HLA class II supermotif or motif are identified and confirmed as outlined below using methodology similar to that described for HLA Class I peptides.
  • a 58P1D12 antigen is analyzed for the presence of sequences bearing an HLA-DR-motif or supermotif. Specifically, 15-mer sequences are selected comprising a DR-supermotif, comprising a 9-mer core, and three-residue N- and C-terminal flanking regions (15 amino acids total).
  • Protocols for predicting peptide binding to DR molecules have been developed (Southwood et al., J. Immunol. 160:3363-3373, 1998). These protocols, specific for individual DR molecules, allow the scoring, and ranking, of 9-mer core regions. Each protocol not only scores peptide sequences for the presence of DR-supermotif primary anchors (i.e., at position 1 and position 6) within a 9-mer core, but additionally evaluates sequences for the presence of secondary anchors. Using allele-specific selection tables (see, e.g., Southwood et al., ibid.), it has been found that these protocols efficiently select peptide sequences with a high probability of binding a particular DR molecule. Additionally, it has been found that performing these protocols in tandem, specifically those for DR1, DR4w4, and DR7, can efficiently select DR cross-reactive peptides.
  • the 58P1D12-derived peptides identified above are tested for their binding capacity for various common HLA-DR molecules. All peptides are initially tested for binding to the DR molecules in the primary panel: DR1, DR4w4, and DR7. Peptides binding at least two of these three DR molecules are then tested for binding to DR2w2 ⁇ 1, DR2w2 ⁇ 2, DR6w19, and DR9 molecules in secondary assays. Finally, peptides binding at least two of the four secondary panel DR molecules, and thus cumulatively at least four of seven different DR molecules, are screened for binding to DR4w15, DR5w11, and DR8w2 molecules in tertiary assays.
  • Peptides binding at least seven of the ten DR molecules comprising the primary, secondary, and tertiary screening assays are considered cross-reactive DR binders.
  • 58P1D12-derived peptides found to bind common HLA-DR alleles are of particular interest.
  • DR3 binding capacity is a relevant criterion in the selection of HTL epitopes.
  • peptides shown to be candidates may also be assayed for their DR3 binding capacity.
  • peptides binding only to DR3 can also be considered as candidates for inclusion in a vaccine formulation.
  • target 58P1D12 antigens are analyzed for sequences carrying one of the two DR3-specific binding motifs reported by Geluk et al. (J. Immunol. 152:5742-5748, 1994).
  • the corresponding peptides are then synthesized and confirmed as having the ability to bind DR3 with an affinity of 1 ⁇ M or better, i.e., less than 1 ⁇ M.
  • Peptides are found that meet this binding criterion and qualify as HLA class II high affinity binders.
  • DR3 binding epitopes identified in this manner are included in vaccine compositions with DR supermotif-bearing peptide epitopes.
  • the class II motif-bearing peptides are analoged to improve affinity or cross-reactivity.
  • aspartic acid at position 4 of the 9-mer core sequence is an optimal residue for DR3 binding, and substitution for that residue often improves DR 3 binding.
  • This example determines immunogenic DR supermotif- and DR3 motif-bearing epitopes among those identified using the methodology set forth herein.
  • Immunogenicity of HTL epitopes are confirmed in a manner analogous to the determination of immunogenicity of CTL epitopes, by assessing the ability to stimulate HTL responses and/or by using appropriate transgenic mouse models Immunogenicity is determined by screening for: 1.) in vitro primary induction using normal PBMC or 2.) recall responses from patients who have 58P1D12-expressing tumors.
  • This example illustrates the assessment of the breadth of population coverage of a vaccine composition comprised of multiple epitopes comprising multiple supermotifs and/or motifs.
  • the A3-like supertype may also include A34, A66, and A*7401, these alleles were not included in overall frequency calculations.
  • confirmed members of the A2-like supertype family are A*0201, A*0202, A*0203, A*0204, A*0205, A*0206, A*0207, A*6802, and A*6901.
  • the B7-like supertype-confirmed alleles are: B7, B*3501-03, B51, B*5301, B*5401, B*5501-2, B*5601, B*6701, and B*7801 (potentially also B*1401, B*3504-06, B*4201, and B*5602).
  • Population coverage achieved by combining the A2-, A3- and B7-supertypes is approximately 86% in five major ethnic groups. Coverage may be extended by including peptides bearing the A1 and A24 motifs. On average, A1 is present in 12% and A24 in 29% of the population across five different major ethnic groups (Caucasian, North American Black, Chinese, Japanese, and Hispanic). Together, these alleles are represented with an average frequency of 39% in these same ethnic populations. The total coverage across the major ethnicities when A1 and A24 are combined with the coverage of the A2-, A3- and B7-supertype alleles is >95%, see, e.g., Table IV (G). An analogous approach can be used to estimate population coverage achieved with combinations of class II motif-bearing epitopes.
  • an average population coverage is predicted to be greater than 95% in each of five major ethnic populations.
  • the game theory Monte Carlo simulation analysis which is known in the art (see e.g., Osborne, M.J. and Rubinstein, A. “A course in game theory” MIT Press, 1994), can be used to estimate what percentage of the individuals in a population comprised of the Caucasian, North American Black, Japanese, Chinese, and Hispanic ethnic groups would recognize the vaccine epitopes described herein. A preferred percentage is 90%. A more preferred percentage is 95%.
  • Effector cells isolated from transgenic mice that are immunized with peptide epitopes are re-stimulated in vitro using peptide-coated stimulator cells. Six days later, effector cells are assayed for cytotoxicity and the cell lines that contain peptide-specific cytotoxic activity are further re-stimulated. An additional six days later, these cell lines are tested for cytotoxic activity on 51 Cr labeled Jurkat-A2.1/K b target cells in the absence or presence of peptide, and also tested on 51 Cr labeled target cells bearing the endogenously synthesized antigen, i.e. cells that are stably transfected with 58P1D12 expression vectors.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Microbiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Analytical Chemistry (AREA)
  • Mycology (AREA)
  • Hematology (AREA)
  • Physics & Mathematics (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Oncology (AREA)
  • General Physics & Mathematics (AREA)
  • Wood Science & Technology (AREA)
  • Food Science & Technology (AREA)
  • Pathology (AREA)
  • Hospice & Palliative Care (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)

Abstract

A novel gene 58P1D12 and its encoded protein, and variants thereof, are described wherein 58P1D12 exhibits tissue specific expression in normal adult tissue, and is aberrantly expressed in the cancers listed in Table I. Consequently, 58P1D12 provides a diagnostic, prognostic, prophylactic and/or therapeutic target for cancer. The 58P1D12 gene or fragment thereof, or its encoded protein, or variants thereof, or a fragment thereof, can be used to elicit a humoral or cellular immune response; antibodies or T cells reactive with 58P1D12 can be used in active or passive immunization.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. patent application Ser. No. 10/919,654, filed 16 Aug. 2004, now allowed, which is a continuation-in-part of U.S. patent application Ser. No. 10/460,512, filed 11 Jun. 2003, now U.S. Pat. No. 7,153,940, issued 26 Dec. 2006, which is a divisional of U.S. patent application Ser. No. 09/638,203, filed 11 Aug. 2000, now U.S. Pat. No. 6,602,501, issued 5 Aug. 2003, which claims the benefit of priority of U.S. Provisional Patent Application No. 60/148,935, filed 12 Aug. 1999. The contents of each of these applications are hereby incorporated by reference in their entirety.
  • STATEMENT OF RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH
  • Not applicable.
  • REFERENCE TO SEQUENCE LISTING SUBMITTED VIA EFS-WEB
  • The entire content of the following electronic submission of the sequence listing via the USPTO EFS-WEB server, as authorized and set forth in MPEP §1730 II.B.2(a)(A), is incorporated herein by reference in its entirety for all purposes. The sequence listing is identified on the electronically filed text file as follows:
  • File Name Date of Creation Size (bytes)
    511582002003Seqlist.txt Jul. 9, 2010 115,201 bytes
  • REFERENCE TO LIST OF TABLES (APPENDIX) SUBMITTED VIA EFS-WEB
  • The entire content of the following electronic submission of the List of Tables Appendix via the USPTO EFS-WEB server, as authorized and set forth in MPEP §1730 II.B.2(a)(A), is incorporated herein by reference in its entirety for all purposes. The List of Tables Appendix is identified on the electronically filed .pdf file as follows:
  • File Name Date of Creation Size (bytes) Pages (total)
    511582002003TablesI-LVd.pdf Jul. 14, 2010 824,998 bytes 148 pages
  • FIELD OF THE INVENTION
  • The invention described herein relates to genes and their encoded proteins, termed 58P1D12 and variants thereof, expressed in certain cancers, and to diagnostic and therapeutic methods and compositions useful in the management of cancers that express 58P1D12.
  • BACKGROUND OF THE INVENTION
  • Cancer is the second leading cause of human death next to coronary disease. Worldwide, millions of people die from cancer every year. In the United States alone, as reported by the American Cancer Society, cancer causes the death of well over a half-million people annually, with over 1.2 million new cases diagnosed per year. While deaths from heart disease have been declining significantly, those resulting from cancer generally are on the rise. In the early part of the next century, cancer is predicted to become the leading cause of death.
  • Worldwide, several cancers stand out as the leading killers. In particular, carcinomas of the lung, prostate, breast, colon, pancreas, and ovary represent the primary causes of cancer death. These and virtually all other carcinomas share a common lethal feature. With very few exceptions, metastatic disease from a carcinoma is fatal. Moreover, even for those cancer patients who initially survive their primary cancers, common experience has shown that their lives are dramatically altered. Many cancer patients experience strong anxieties driven by the awareness of the potential for recurrence or treatment failure. Many cancer patients experience physical debilitations following treatment. Furthermore, many cancer patients experience a recurrence.
  • Worldwide, prostate cancer is the fourth most prevalent cancer in men. In North America and Northern Europe, it is by far the most common cancer in males and is the second leading cause of cancer death in men. In the United States alone, well over 30,000 men die annually of this disease—second only to lung cancer. Despite the magnitude of these figures, there is still no effective treatment for metastatic prostate cancer. Surgical prostatectomy, radiation therapy, hormone ablation therapy, surgical castration and chemotherapy continue to be the main treatment modalities. Unfortunately, these treatments are ineffective for many and are often associated with undesirable consequences.
  • On the diagnostic front, the lack of a prostate tumor marker that can accurately detect early-stage, localized tumors remains a significant limitation in the diagnosis and management of this disease. Although the serum prostate specific antigen (PSA) assay has been a very useful tool, however its specificity and general utility is widely regarded as lacking in several important respects.
  • Progress in identifying additional specific markers for prostate cancer has been improved by the generation of prostate cancer xenografts that can recapitulate different stages of the disease in mice. The LAPC (Los Angeles Prostate Cancer) xenografts are prostate cancer xenografts that have survived passage in severe combined immune deficient (SCID) mice and have exhibited the capacity to mimic the transition from androgen dependence to androgen independence (Klein et al., 1997, Nat. Med. 3:402). More recently identified prostate cancer markers include PCTA-1 (Su et al., 1996, Proc. Natl. Acad. Sci. USA 93: 7252), prostate-specific membrane (PSM) antigen (Pinto et al., Clin Cancer Res 1996 September 2 (9): 1445-51), STEAP (Hubert, et al., Proc Natl Acad Sci U S A. 1999 Dec. 7; 96(25): 14523-8) and prostate stem cell antigen (PSCA) (Reiter et al., 1998, Proc. Natl. Acad. Sci. USA 95: 1735).
  • While previously identified markers such as PSA, PSM, PCTA and PSCA have facilitated efforts to diagnose and treat prostate cancer, there is need for the identification of additional markers and therapeutic targets for prostate and related cancers in order to further improve diagnosis and therapy.
  • Renal cell carcinoma (RCC) accounts for approximately 3 percent of adult malignancies. Once adenomas reach a diameter of 2 to 3 cm, malignant potential exists. In the adult, the two principal malignant renal tumors are renal cell adenocarcinoma and transitional cell carcinoma of the renal pelvis or urethras. The incidence of renal cell adenocarcinoma is estimated at more than 29,000 cases in the United States, and more than 11,600 patients died of this disease in 1998. Transitional cell carcinoma is less frequent, with an incidence of approximately 500 cases per year in the United States.
  • Surgery has been the primary therapy for renal cell adenocarcinoma for many decades. Until recently, metastatic disease has been refractory to any systemic therapy. With recent developments in systemic therapies, particularly immunotherapies, metastatic renal cell carcinoma may be approached aggressively in appropriate patients with a possibility of durable responses. Nevertheless, there is a remaining need for effective therapies for these patients.
  • Of all new cases of cancer in the United States, bladder cancer represents approximately 5 percent in men (fifth most common neoplasm) and 3 percent in women (eighth most common neoplasm). The incidence is increasing slowly, concurrent with an increasing older population. In 1998, there was an estimated 54,500 cases, including 39,500 in men and 15,000 in women. The age-adjusted incidence in the United States is 32 per 100,000 for men and eight per 100,000 in women. The historic male/female ratio of 3:1 may be decreasing related to smoking patterns in women. There were an estimated 11,000 deaths from bladder cancer in 1998 (7,800 in men and 3,900 in women). Bladder cancer incidence and mortality strongly increase with age and will be an increasing problem as the population becomes more elderly.
  • Most bladder cancers recur in the bladder. Bladder cancer is managed with a combination of transurethral resection of the bladder (TUR) and intravesical chemotherapy or immunotherapy. The multifocal and recurrent nature of bladder cancer points out the limitations of TUR. Most muscle-invasive cancers are not cured by TUR alone. Radical cystectomy and urinary diversion is the most effective means to eliminate the cancer but carry an undeniable impact on urinary and sexual function. There continues to be a significant need for treatment modalities that are beneficial for bladder cancer patients.
  • An estimated 130,200 cases of colorectal cancer occurred in 2000 in the United States, including 93,800 cases of colon cancer and 36,400 of rectal cancer. Colorectal cancers are the third most common cancers in men and women. Incidence rates declined significantly during 1992-1996 (−2.1% per year). Research suggests that these declines have been due to increased screening and polyp removal, preventing progression of polyps to invasive cancers. There were an estimated 56,300 deaths (47,700 from colon cancer, 8,600 from rectal cancer) in 2000, accounting for about 11% of all U.S. cancer deaths.
  • At present, surgery is the most common form of therapy for colorectal cancer, and for cancers that have not spread, it is frequently curative. Chemotherapy, or chemotherapy plus radiation, is given before or after surgery to most patients whose cancer has deeply perforated the bowel wall or has spread to the lymph nodes. A permanent colostomy (creation of an abdominal opening for elimination of body wastes) is occasionally needed for colon cancer and is infrequently required for rectal cancer. There continues to be a need for effective diagnostic and treatment modalities for colorectal cancer.
  • There were an estimated 164,100 new cases of lung and bronchial cancer in 2000, accounting for 14% of all U.S. cancer diagnoses. The incidence rate of lung and bronchial cancer is declining significantly in men, from a high of 86.5 per 100,000 in 1984 to 70.0 in 1996. In the 1990s, the rate of increase among women began to slow. In 1996, the incidence rate in women was 42.3 per 100,000.
  • Lung and bronchial cancer caused an estimated 156,900 deaths in 2000, accounting for 28% of all cancer deaths. During 1992-1996, mortality from lung cancer declined significantly among men (−1.7% per year) while rates for women were still significantly increasing (0.9% per year). Since 1987, more women have died each year of lung cancer than breast cancer, which, for over 40 years, was the major cause of cancer death in women. Decreasing lung cancer incidence and mortality rates most likely resulted from decreased smoking rates over the previous 30 years; however, decreasing smoking patterns among women lag behind those of men. Of concern, although the declines in adult tobacco use have slowed, tobacco use in youth is increasing again.
  • Treatment options for lung and bronchial cancer are determined by the type and stage of the cancer and include surgery, radiation therapy, and chemotherapy. For many localized cancers, surgery is usually the treatment of choice. Because the disease has usually spread by the time it is discovered, radiation therapy and chemotherapy are often needed in combination with surgery. Chemotherapy alone or combined with radiation is the treatment of choice for small cell lung cancer; on this regimen, a large percentage of patients experience remission, which in some cases is long lasting. There is however, an ongoing need for effective treatment and diagnostic approaches for lung and bronchial cancers.
  • An estimated 182,800 new invasive cases of breast cancer were expected to occur among women in the United States during 2000. Additionally, about 1,400 new cases of breast cancer were expected to be diagnosed in men in 2000. After increasing about 4% per year in the 1980s, breast cancer incidence rates in women have leveled off in the 1990s to about 110.6 cases per 100,000.
  • In the U.S. alone, there were an estimated 41,200 deaths (40,800 women, 400 men) in 2000 due to breast cancer. Breast cancer ranks second among cancer deaths in women. According to the most recent data, mortality rates declined significantly during 1992-1996 with the largest decreases in younger women, both white and black. These decreases were probably the result of earlier detection and improved treatment.
  • Taking into account the medical circumstances and the patient's preferences, treatment of breast cancer may involve lumpectomy (local removal of the tumor) and removal of the lymph nodes under the arm; mastectomy (surgical removal of the breast) and removal of the lymph nodes under the arm; radiation therapy; chemotherapy; or hormone therapy. Often, two or more methods are used in combination. Numerous studies have shown that, for early stage disease, long-term survival rates after lumpectomy plus radiotherapy are similar to survival rates after modified radical mastectomy. Significant advances in reconstruction techniques provide several options for breast reconstruction after mastectomy. Recently, such reconstruction has been done at the same time as the mastectomy.
  • Local excision of ductal carcinoma in situ (DCIS) with adequate amounts of surrounding normal breast tissue may prevent the local recurrence of the DCIS. Radiation to the breast and/or tamoxifen may reduce the chance of DCIS occurring in the remaining breast tissue. This is important because DCIS, if left untreated, may develop into invasive breast cancer. Nevertheless, there are serious side effects or sequelae to these treatments. There is, therefore, a need for efficacious breast cancer treatments.
  • There were an estimated 23,100 new cases of ovarian cancer in the United States in 2000. It accounts for 4% of all cancers among women and ranks second among gynecologic cancers. During 1992-1996, ovarian cancer incidence rates were significantly declining. Consequent to ovarian cancer, there were an estimated 14,000 deaths in 2000. Ovarian cancer causes more deaths than any other cancer of the female reproductive system.
  • Surgery, radiation therapy, and chemotherapy are treatment options for ovarian cancer. Surgery usually includes the removal of one or both ovaries, the fallopian tubes (salpingo-oophorectomy), and the uterus (hysterectomy). In some very early tumors, only the involved ovary will be removed, especially in young women who wish to have children. In advanced disease, an attempt is made to remove all intra-abdominal disease to enhance the effect of chemotherapy. There continues to be an important need for effective treatment options for ovarian cancer.
  • There were an estimated 28,300 new cases of pancreatic cancer in the United States in 2000. Over the past 20 years, rates of pancreatic cancer have declined in men. Rates among women have remained approximately constant but may be beginning to decline. Pancreatic cancer caused an estimated 28,200 deaths in 2000 in the United States. Over the past 20 years, there has been a slight but significant decrease in mortality rates among men (about −0.9% per year) while rates have increased slightly among women.
  • Surgery, radiation therapy, and chemotherapy are treatment options for pancreatic cancer. These treatment options can extend survival and/or relieve symptoms in many patients but are not likely to produce a cure for most. There is a significant need for additional therapeutic and diagnostic options for pancreatic cancer.
  • SUMMARY OF THE INVENTION
  • The present invention relates to a gene, designated 58P1D12, that has now been found to be over-expressed in the cancer(s) listed in Table I. Northern blot expression analysis of 58P1D12 gene expression in normal tissues shows a restricted expression pattern in adult tissues. The nucleotide (FIG. 2) and amino acid (FIG. 2, and FIG. 3) sequences of 58P1D12 are provided. The tissue-related profile of 58P1D12 in normal adult tissues, combined with the over-expression observed in the tissues listed in Table I, shows that 58P1D12 is aberrantly over-expressed in at least some cancers, and thus serves as a useful diagnostic, prophylactic, prognostic, and/or therapeutic target for cancers of the tissue(s) such as those listed in Table I.
  • The invention provides polynucleotides corresponding or complementary to all or part of the 58P1D12 genes, mRNAs, and/or coding sequences, preferably in isolated form, including polynucleotides encoding 58P1D12-related proteins and fragments of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or more than 25 contiguous amino acids; at least 30, 35, 40, 45, 50, 55, 60, 65, 70, 80, 85, 90, 95, 100 or more than 100 contiguous amino acids of a 58P1D12-related protein, as well as the peptides/proteins themselves; DNA, RNA, DNA/RNA hybrids, and related molecules, polynucleotides or oligonucleotides complementary or having at least a 90% homology to the 58P1D12 genes or mRNA sequences or parts thereof, and polynucleotides or oligonucleotides that hybridize to the 58P1D12 genes, mRNAs, or to 58P1D12-encoding polynucleotides. Also provided are means for isolating cDNAs and the genes encoding 58P1D12. Recombinant DNA molecules containing 58P1D12 polynucleotides, cells transformed or transduced with such molecules, and host-vector systems for the expression of 58P1D12 gene products are also provided. The invention further provides antibodies that bind to 58P1D12 proteins and polypeptide fragments thereof, including polyclonal and monoclonal antibodies, murine and other mammalian antibodies, chimeric antibodies, humanized and fully human antibodies, and antibodies labeled with a detectable marker or therapeutic agent. In certain embodiments, there is a proviso that the entire nucleic acid sequence of FIG. 2 is not encoded and/or the entire amino acid sequence of FIG. 2 is not prepared. In certain embodiments, the entire nucleic acid sequence of FIG. 2 is encoded and/or the entire amino acid sequence of FIG. 2 is prepared, either of which are in respective human unit dose forms.
  • The invention further provides methods for detecting the presence and status of 58P1D12 polynucleotides and proteins in various biological samples, as well as methods for identifying cells that express 58P1D12. A typical embodiment of this invention provides methods for monitoring 58P1D12 gene products in a tissue or hematology sample having or suspected of having some form of growth dysregulation such as cancer.
  • The invention further provides various immunogenic or therapeutic compositions and strategies for treating cancers that express 58P1D12 such as cancers of tissues listed in Table I, including therapies aimed at inhibiting the transcription, translation, processing or function of 58P1D12 as well as cancer vaccines. In one aspect, the invention provides compositions, and methods comprising them, for treating a cancer that expresses 58P1D12 in a human subject wherein the composition comprises a carrier suitable for human use and a human unit dose of one or more than one agent that inhibits the production or function of 58P1D12. Preferably, the carrier is a uniquely human carrier. In another aspect of the invention, the agent is a moiety that is immunoreactive with 58P1D12 protein. Non-limiting examples of such moieties include, but are not limited to, antibodies (such as single chain, monoclonal, polyclonal, humanized, chimeric, or human antibodies), functional equivalents thereof (whether naturally occurring or synthetic), and combinations thereof. The antibodies can be conjugated to a diagnostic or therapeutic moiety. In another aspect, the agent is a small molecule as defined herein.
  • In another aspect, the agent comprises one or more than one peptide which comprises a cytotoxic T lymphocyte (CTL) epitope that binds an HLA class I molecule in a human to elicit a CTL response to 58P1D12 and/or one or more than one peptide which comprises a helper T lymphocyte (HTL) epitope which binds an HLA class II molecule in a human to elicit an HTL response. The peptides of the invention may be on the same or on one or more separate polypeptide molecules. In a further aspect of the invention, the agent comprises one or more than one nucleic acid molecule that expresses one or more than one of the CTL or HTL response stimulating peptides as described above. In yet another aspect of the invention, the one or more than one nucleic acid molecule may express a moiety that is immunologically reactive with 58P1D12 as described above. The one or more than one nucleic acid molecule may also be, or encodes, a molecule that inhibits production of 58P1D12. Non-limiting examples of such molecules include, but are not limited to, those complementary to a nucleotide sequence essential for production of 58P1D12 (e.g. antisense sequences or molecules that form a triple helix with a nucleotide double helix essential for 58P1D12 production) or a ribozyme effective to lyse 58P1D12 mRNA.
  • Note that to determine the starting position of any peptide set forth in Tables VIII-XXI and XXII to XLIX (collectively HLA Peptide Tables) respective to its parental protein, e.g., variant 1, variant 2, etc., reference is made to three factors: the particular variant, the length of the peptide in an HLA Peptide Table, and the Search Peptides in Table VII. Generally, a unique Search Peptide is used to obtain HLA peptides of a particular for a particular variant. The position of each Search Peptide relative to its respective parent molecule is listed in Table VII. Accordingly, if a Search Peptide begins at position “X”, one must add the value “X−1” to each position in Tables VIII-XXI and XXII to XLIX to obtain the actual position of the HLA peptides in their parental molecule. For example, if a particular Search Peptide begins at position 150 of its parental molecule, one must add 150−1, i.e., 149 to each HLA peptide amino acid position to calculate the position of that amino acid in the parent molecule.
  • One embodiment of the invention comprises an HLA peptide, that occurs at least twice in Tables VIII-XXI and XXII to XLIX collectively, or an oligonucleotide that encodes the HLA peptide. Another embodiment of the invention comprises an HLA peptide that occurs at least once in Tables VIII-XXI and at least once in tables XXII to XLIX, or an oligonucleotide that encodes the HLA peptide.
  • Another embodiment of the invention is antibody epitopes, which comprise a peptide regions, or an oligonucleotide encoding the peptide region, that has one two, three, four, or five of the following characteristics:
  • i) a peptide region of at least 5 amino acids of a particular peptide of FIG. 3, in any whole number increment up to the full length of that protein in FIG. 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Hydrophilicity profile of FIG. 5;
  • ii) a peptide region of at least 5 amino acids of a particular peptide of FIG. 3, in any whole number increment up to the full length of that protein in FIG. 3, that includes an amino acid position having a value equal to or less than 0.5, 0.4, 0.3, 0.2, 0.1, or having a value equal to 0.0, in the Hydropathicity profile of FIG. 6;
  • iii) a peptide region of at least 5 amino acids of a particular peptide of FIG. 3, in any whole number increment up to the full length of that protein in FIG. 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Percent Accessible Residues profile of FIG. 7;
  • iv) a peptide region of at least 5 amino acids of a particular peptide of FIG. 3, in any whole number increment up to the full length of that protein in FIG. 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Average Flexibility profile of FIG. 8; or
  • v) a peptide region of at least 5 amino acids of a particular peptide of FIG. 3, in any whole number increment up to the full length of that protein in FIG. 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Beta-turn profile of FIG. 9. [!!! The Figure descriptions need to be revised when using this as a template]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1. The 58P1D12 SSH sequence of 427 nucleotides.
  • FIG. 2A The cDNA and amino acid sequence of 58P1D12 variant 1 (also called “58P1D12 v.1” or “58P1D12 variant 1”) is shown in FIG. 2A. The start methionine is underlined. The open reading frame extends from nucleic acid 380-1201 including the stop codon.
  • FIG. 2B. The cDNA and amino acid sequence of 58P1D12 variant 2 (also called “58P1D12 v.2”) is shown in FIG. 2B. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 388-1086 including the stop codon.
  • FIG. 2C. The cDNA and amino acid sequence of 58P1D12 variant 3 (also called “58P1D12 v.3”) is shown in FIG. 2C. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 206-904 including the stop codon.
  • FIG. 2D. The cDNA and amino acid sequence of 58P1D12 variant 4 (also called “58P1D12 v.4”) is shown in FIG. 2D. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 206-916 including the stop codon.
  • FIG. 2E. The cDNA and amino acid sequence of 58P1D12 variant 5 (also called “58P1D12 v.5”) is shown in FIG. 2E. The codon for the start methionine is underlined. The open reading frame extends from nucleic acid 106-816 including the stop codon.
  • FIG. 2F. Variants 58P1D12 v.6 through v.15 are variants with single nucleotide difference from 58P1D12 v.1. Though these SNP variants are shown separately, they can also occur in any combinations and in any of the transcript variants listed above in FIGS. 2A-2F.
  • FIG. 3A. The amino acid sequence of 58P1D12 v.1 is shown in FIG. 3A; it has 273 amino acids.
  • FIG. 3B. The amino acid sequence of 58P1D12 v.2 is shown in FIG. 3B; it has 232 amino acids.
  • FIG. 3C. The amino acid sequence of 58P1D12 v.3 is shown in FIG. 3C; it has 236 amino acids.
  • As used herein, a reference to 58P1D12 includes all variants thereof, including those shown in FIGS. 2, 3, 10, 11, and 12 unless the context clearly indicates otherwise.
  • FIG. 4. Intratibial tumor growth of 3T3-58P1D12 cells. 3T3 fibroblasts were infected with empty vector control amphotropic virus (Neo) or virus containing the 58P1D12 gene. Stable transfectants were selected in G-418 (geneticin) and then grown in 10% FBS. For each cell type, 3T3-Neo and 3T3-58P1D12, 1×106 cells were mixed with Matrigel® and injected into the flanks of 7 male SCID mice. After 31 days, the mice were sacrified, and the tumors were extracted with the hind quarters. The data are representative of the 7 mice treated with the two cell types. The data indicate that the expression of 58P1D12 in the non-tumor forming 3T3 line has increased tumor growth potential in vivo.
  • FIG. 5(A)-(C). Hydrophilicity amino acid profile of 58P1D12 v.1-v.3 determined by computer algorithm sequence analysis using the method of Hopp and Woods (Hopp T. P., Woods K. R., 1981. Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828) accessed on the Protscale website located on the World Wide Web at (expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.
  • FIG. 6(A)-(C). Hydropathicity amino acid profile of 58P1D12 v.1-v.9 determined by computer algorithm sequence analysis using the method of Kyte and Doolittle (Kyte J., Doolittle R. F., 1982. J. Mol. Biol. 157:105-132) accessed on the ProtScale website located on the World Wide Web at (.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.
  • FIG. 7(A)-(C). Percent accessible residues amino acid profile of 58P1D12 v.1-v.9 determined by computer algorithm sequence analysis using the method of Janin (Janin J., 1979 Nature 277:491-492) accessed on the ProtScale website located on the World Wide Web at (.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.
  • FIG. 8(A)-(C). Average flexibility amino acid profile of 58P1D12 v.1-v.9 determined by computer algorithm sequence analysis using the method of Bhaskaran and Ponnuswamy (Bhaskaran R., and Ponnuswamy P.K., 1988. Int. J. Pept. Protein Res. 32:242-255) accessed on the ProtScale website located on the World Wide Web at (.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.
  • FIG. 9(A)-(C). Beta-turn amino acid profile of 58P1D12 v.1-v.9 determined by computer algorithm sequence analysis using the method of Deleage and Roux (Deleage, G., Roux B. 1987 Protein Engineering 1:289-294) accessed on the ProtScale website located on the World Wide Web at (.expasy.ch/cgi-bin/protscale.pl) through the ExPasy molecular biology server.
  • FIG. 10. Structures of transcript variants of 58P1D12. Variant 58P1D12 v.2 through v.5 were transcript variants of 58P1D12 v.1. Variants 58P1D12 v.2 and v.3, each had two different exons in the 5′ end, shared five exons in the 3′ end with v.1. Variants v.4 and v.5 were similar to v.3, with deletions of one or two exons from v.3. All the exons in different transcript variants were aligned in relative order as on the genome. Poly A tails and SNP are not shown here. Numbers in “( )” underneath the boxes correspond to those of 58P1D12 v.1. Lengths of introns and exons are not proportional.
  • FIG. 11. Schematic alignment of protein variants of 58P1D12. Protein variants corresponded to nucleotide variants. Nucleotide variants 58P1D12 v.3 coded for the same protein as variant v.2, and variant v.5 coded for the same protein as v.4. SNP variants v.6 through v.15 coded for the same protein as v.1. Nucleotide variants 58P1D12 v.6 through v.15 each had one alternative allele of a SNP as compared with variant v.1, as shown in FIG. 12. Proteins of v.2 and v.3 were portion of v.1 protein, while variants v.4 and v.5 had a portion that was different from any port of v.1. Numbers underneath the box correspond to 58P1D12 v.1. Black boxes represent the same sequence as 58P1D12 v.1.
  • FIG. 12. Schematic alignment of SNP variants of 58P1D12. Variants 58P1D12 v.6 through v.12 were variants with single nucleotide differences as compared to variant v.1. Though these SNP variants were shown separately, they could also occur in any combinations (called haplotype), and in any transcript variants that contained the base pairs, such as v.2 shown in FIG. 10. Numbers correspond to those of 58P1D12 v.1. Black box shows the same sequence as 58P1D12 v.1. SNPs are indicated above the box.
  • FIG. 13. Secondary structure and transmembrane domains prediction for 58P1D12 protein variants.
  • FIG. 13A-13C: The secondary structures of 58P1D12 protein variants 1 (SEQ ID NO:15), variants 2 (SEQ ID NO:16), variant 3 (SEQ ID NO:17), respectively, were predicted using the HNN—Hierarchical Neural Network method (NPS@: Network Protein Sequence Analysis TIBS 2000 March Vol. 25, No 3 [291]:147-150 Combet C., Blanchet C., Geourjon C. and Deléage G., at the World Wide Web address pbillbcp.fr/cgi-bin/npsa_automat.pl?page=npsa_nn.html), accessed from the ExPasy molecular biology server located on the World Wide Web at (.expasy.ch/tools/). This method predicts the presence and location of alpha helices, extended strands, and random coils from the primary protein sequence. The percent of the protein variant in a given secondary structure is also listed.
  • FIG. 13D, FIG. 13F, FIG. 13H: Schematic representation of the probability of existence of transmembrane regions of 58P1D12 protein variants 1, 2, 3, respectively, based on the TMpred algorithm of Hofmann and Stoffel which utilizes TMBASE (K. Hofmann, W. Stoffel. TMBASE—A database of membrane spanning protein segments Biol. Chem. Hoppe-Seyler 374:166, 1993).
  • FIG. 13E, FIG. 13G, FIG. 13I: Schematic representation of the probability of the existence of transmembrane regions of 58P1D12 variants 1, 2, 3, respectively, based on the TMHMM algorithm of Sonnhammer, von Heijne, and Krogh (Erik L.L. Sonnhammer, Gunnar von Heijne, and Anders Krogh: A hidden Markov model for predicting transmembrane helices in protein sequences. In Proc. of Sixth Int. Conf. on Intelligent Systems for Molecular Biology, p 175-182 Ed J. Glasgow, T. Littlejohn, F. Major, R. Lathrop, D. Sankoff, and C. Sensen Menlo Park, Calif.: AAAI Press, 1998). The TMpred and TMHMM algorithms are accessed from the ExPasy molecular biology server located on the World Wide Web at (.expasy.ch/tools/).
  • FIG. 14. 58P1D12 Expression in Normal and Patient Cancer Tissues. First strand cDNA was prepared from normal tissues (bladder, brain, heart, kidney, liver, lung, prostate, spleen, skeletal muscle, testis, pancreas, colon and stomach), and from pools of patient cancer specimens (prostate cancer pool, bladder cancer pool, lung cancer pool, ovary cancer pool, prostate cancer xenograft pool, bone and melanoma cancer pool, and cervical cancer pool). Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using primers to 58P1D12, was performed at 30 cycles of amplification. Results expression of 58P1D12 predominantly in testis amongst the normal tissues tested. 58P1D12 is also expressed in prostate cancer, bladder cancer, lung cancer, ovary cancer, prostate cancer xenograft, bone/melanoma cancer pool, and cervical cancer.
  • FIG. 15. Expression of 58P1D12 in normal tissues. Two multiple tissue northern blots (Clontech) both with 2 ug of mRNA/lane were probed with the 58P1D12 sequence. Size standards in kilobases (kb) are indicated on the side. Results show predominant expression of 58P1D12 transcript in normal testis. A significantly weaker signal is detected in spleen, lung, kidney and pancreas.
  • FIG. 16. 58P1D12 Expression in Prostate Cancer Xenograft tissues. RNA was extracted from LAPC prostate cancer xenografts, LAPC-4AD, LAPC-4AI, LAPC-9AD and LAPC-9AI, as well as from normal prostate. Northern blots with 10 ug of total RNA were probed with the 58P1D12 sequence. Size standards in kilobases are on the side. Results show expression of 58P1D12 in LAPC-9AD but not in the other tissues tested.
  • FIG. 17. 58P1D12 Expression in Normal and Patient Cancer Specimens. RNA was extracted from a pool of 3 ovary tumor patient specimens, normal prostate (NP), normal bladder (NB), normal kidney (NK), and normal colon (NC). Northern blots with 10 ug of total RNA were probed with the 58P1D12 sequence. Size standards in kilobases are on the side. Results show expression of 58P1D12 in ovary tumor patient specimens but not in the normal tissues tested.
  • FIG. 18. 58P1D12 Expression in Ovarian Cancer Patient Specimens. RNA was extracted from normal ovary, LAPC-9AD prostate cancer xenograft, and a panel of ovary tumor patient specimens (Ovary T). Northern blots with 10 ug of total RNA were probed with the 58P1D12 sequence. Size standards in kilobases are on the side. Results show expression of 58P1D12 in most of the patient specimens tested as well as in the prostate xenograft.
  • FIG. 19. 58P1D12 Expression in a Large Panel of Ovarian Cancer Patient Specimens. First strand cDNA was prepared from a panel of ovary patient cancer specimens as well as two different normal ovary tissues. Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using primers to 58P1D12, was performed at 26 and 30 cycles of amplification. Samples were run on an agarose gel, and PCR products were quantitated using the AlphaImager software. Expression was recorded as negative, weak, medium or strong. Results show expression of 58P1D12 in the majority of patient cancer specimens tested (75%), but not in normal ovary.
  • FIG. 20. Specific cell surface staining of Rat1-58P1D12 cells with serum from mice immunized with recombinant 58P1D12 protein. Rat1-58P1D12 and control Rat1-neo cells (˜100,000 cells) were incubated with various dilutions of serum obtained from 2 different mice immunized with recombinant Tag5-58P1D12 protein. Cells were washed and then incubated with goat anti-mouse IgG-PE secondary Ab (1:100 dilution in PBS+2% FBS) on ice for 1 hour. Cells were washed again, fixed in 2% paraformaldehyde in PBS, and subjected to FACS analysis. The Rat1-58P1D12 cells, but not the Rat1-neo cells, show a shift in fluorescent intensity over a large dilution range demonstrating specific binding of anti-58P1D12 Abs present in the serum from the 2 mice.
  • FIG. 21. Specific cell surface staining of Rat1-58P1D12 cells with monoclonal antibody hybridoma supernatants. Rat1-58P1D12 and control Rat1-neo cells (100,000 cells) were incubated with supernatants derived from hybridomas M8-143(5).10.1 and M8-143(5).45.1. on ice for 1 hour. Cells were washed and then incubated with goat anti-mouse IgG-PE secondary Ab (1:100 dilution in PBS+2% FBS) on ice for 1 hour. Cells were washed again, fixed in 2% paraformaldehyde in PBS, and subjected to FACS analysis. The Rat1-58P1D12 cells, but not the Rat1-neo cells, show a shift in fluorescent intensity demonstrating specific binding of the MAb to 58P1D12 protein on the cell surface.
  • FIG. 22. Affinity determination of MAb M8-143(5).10.1 by FACS-based equilibrium binding analysis. 3T3-58P1D12 cells were incubated at 4 C for 2 hours with various concentrations of MAb M8-143(5).10.1 in PBS+2% FBS+0.04% NaN3. Cells were washed, and then incubated at 4 C for 1 hour with a 1:100 dilution of goat anti-mouse IgG-PE secondary Ab. After washing, cells were fixed in 2% paraformaldehyde in PBS and subjected to FACS analysis. Shown on the left are the histograms of cells stained with various concentrations of MAb. On the right is a graphical representation of the data plotting MAb concentration versus the mean fluorescent intensity of the stained populations. Non-linear regression of the binding data was carried out with the GraphPad Prism software (Version 3.00 for Windows, GraphPad Software, San Diego Calif. USA) to determine the dissociation constant (KD). The analysis shows that MAb M8-143(5).10.1 is a high affinity MAb with an apparent KD of 0.086 nM on 3T3-58P1D12 cells.
  • FIG. 23. 58P1D12 induces angiogenesis (tube formation) in umbilical vein endothelial cells (HUVEC). Primary umbilical vein endothelial cells were cultured in 10% FBS, then removed with EDTA and mixed with media containing either 10% FBS, 0.1% FBS, 0.1% FBS+50 ng/ml VEGF or 0.1% FBS+5 □g/ml 58P1D12 ECD protein. The mixtures were plated onto a 200 □l layer of Matrigel® and then incubated at 37° C. for 16 hours. The plates were then analyzed for the formation of tubes by microscopy. Images of representative fields are shown. The data show that HUVEC cells form tubes in 10% FBS but not in 0.1% FBS, thereby allowing the assessment of 58P1D12-induced angiogenesis in low serum conditions. At 5 □g/ml, 58P1D12 ECD exhibited the capacity to induce the formation of endothelial tubes similarly as the positive control VEGF.
  • FIG. 24. 58P1D12 induces angiogenesis (tube formation) in lung microvascular endothelial cells (HMVEC). Primary lung endothelial cells were cultured in 10% FBS, then removed with EDTA and mixed with media containing either 0.1% FBS, 0.1% FBS+50 ng/ml VEGF or 0.1% FBS+5 □g/ml 58P1D12 ECD protein. The mixtures were plated onto a 200 □l layer of Matrigel® and then incubated at 37° C. for 16 hours. The plates were then analyzed for the formation of tubes by microscopy. Images of representative fields are shown. The data show that HMVEC cells do not form tubes in 0.1% FBS, thereby allowing the assessment of 58P1D12-induced angiogenesis in low serum conditions. At 5 □g/ml, 58P1D12 ECD exhibited the capacity to induce the formation of endothelial tubes similarly as the positive control VEGF.
  • FIG. 25. Monoclonal antibody to 58P1D12 inhibits angiogenesis (tube formation) in umbilical vein endothelial cells (HUVEC). Primary umbilical vein endothelial cells were cultured in 10% FBS, then removed with EDTA and mixed with media containing either 0.1% FBS, 5% FBS, 0.1% FBS+3 □g/ml 58P1D12 ECD protein, 0.1% FBS+3 □g/ml 58P1D12 ECD protein+30 □g/ml MAb M8-143(5)10 or 0.1% FBS+3 □g/ml 58P1D12 ECD protein+30 □g/ml MAb M3-47.24. The mixtures were plated onto a 200 □l layer of Matrigel® and then incubated at 37° C. for 16 hours. The plates were then analyzed for the formation of tubes by microscopy. The total tube counts are shown. The data show that HUVEC cells form tubes in 5% FBS but not in 0.1% FBS, thereby allowing the assessment of 58P1D12-induced angiogenesis in low serum conditions. At 3 □g/ml, 58P1D12 ECD exhibited the capacity to induce the formation of endothelial tubes, and that incubation with MAb M8-143(5)10, but not MAb M3-47.24, inhibited the tube formation by 45%.
  • FIG. 26. 58P1D12 ECD enhances survival of HUVEC. Primary umbilical vein endothelial cells were cultured in 10% FBS. Cells were trypsinized and 3000 cells per well were plated onto 96-well dishes for 72 hours. Cell viability was assayed by Alamar blue staining for 3 hours, followed by fluorescence measurement. The assay was performed in triplicate with standard deviations represented. In low serum conditions (0.1% FBS), cell viability was low, but was enhanced by increasing concentrations of 58P1D12 ECD. For positive control, the cells were treated with 50 ng/ml VEGF. Cell viability at 10 □g/ml 58P1D12 ECD was similar to that observed using 50 ng/ml VEGF (a known survival signal for endothelium).
  • FIG. 27. 58P1D12 ECD enhances proliferation of HMVEC. Primary lung microvascular endothelial cells (HMVEC) were cultured in 10% FBS. Cells were trypsinized and 3000 cells per well were plated onto 96-well dishes in 2% FBS. The cells were either left untreated, or were incubated with 50 ng/ml VEGF, 5 □g/ml 58P1D12 ECD or 10 □g/ml 58P1D12 ECD. After 48 hours, the cells were incubated with 3H-thymidine and allowed to grow overnight. Incorporation of 3H-thymidine into cell DNA was assayed by standard protocols. The assay was performed in triplicate with standard deviations represented. The data indicate that basal proliferation could be measured in 2% FBS, where increased proliferation was observed with 58P1D12 ECD. Similar levels of enhanced proliferation were seen with 10 □g/ml 58P1D12 ECD compared with the VEGF positive control.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Outline of Sections
  • I.) Definitions
  • II.) 58P1D12 Polynucleotides
      • II.A.) Uses of 58P1D12 Polynucleotides
        • II.A.1.) Monitoring of Genetic Abnormalities
        • II.A.2.) Antisense Embodiments
        • II.A.3.) Primers and Primer Pairs
        • II.A.4.) Isolation of 58P1D12-Encoding Nucleic Acid Molecules
        • II.A.5.) Recombinant Nucleic Acid Molecules and Host-Vector Systems
  • III.) 58P1D12-related Proteins
      • III.A.) Motif-bearing Protein Embodiments
      • III.B.) Expression of 58P1D12-related Proteins
      • III.C.) Modifications of 58P1D12-related Proteins
      • III.D.) Uses of 58P1D12-related Proteins
  • IV.) 58P1D12 Antibodies
  • V.) 58P1D12 Cellular Immune Responses
  • VI.) 58P1D12 Transgenic Animals
  • VII.) Methods for the Detection of 58P1D12
  • VIII.) Methods for Monitoring the Status of 58P1D12-related Genes and Their Products
  • IX.) Identification of Molecules That Interact With 58P1D12
  • X.) Therapeutic Methods and Compositions
      • X.A.) Anti-Cancer Vaccines
      • X.B.) 58P1D12 as a Target for Antibody-Based Therapy
      • X.C.) 58P1D12 as a Target for Cellular Immune Responses
        • X.C.1. Minigene Vaccines
        • X.C.2. Combinations of CTL Peptides with Helper Peptides
        • X.C.3. Combinations of CTL Peptides with T Cell Priming Agents
        • X.C.4. Vaccine Compositions Comprising DC Pulsed with CTL and/or HTL Peptides
        • X.D.) Adoptive Immunotherapy
        • X.E.) Administration of Vaccines for Therapeutic or Prophylactic Purposes
  • XI.) Diagnostic and Prognostic Embodiments of 58P1D12.
  • XII) Inhibition of 58P1D12 Protein Function
      • XII.A.) Inhibition of 58P1D12 With Intracellular Antibodies
      • XII.B.) Inhibition of 58P1D12 with Recombinant Proteins
      • XII.C.) Inhibition of 58P1D12 Transcription or Translation
      • XII.D.) General Considerations for Therapeutic Strategies
  • XIII.) Identification, Characterization and Use of Modulators of 109P1D1
  • XIV.) KITS/Articles of Manufacture
  • I.) DEFINITIONS
  • Unless otherwise defined, all terms of art, notations and other scientific terms or terminology used herein are intended to have the meanings commonly understood by those of skill in the art to which this invention pertains. In some cases, terms with commonly understood meanings are defined herein for clarity and/or for ready reference, and the inclusion of such definitions herein should not necessarily be construed to represent a substantial difference over what is generally understood in the art. Many of the techniques and procedures described or referenced herein are well understood and commonly employed using conventional methodology by those skilled in the art, such as, for example, the widely utilized molecular cloning methodologies described in Sambrook et al., Molecular Cloning: A Laboratory Manual 2nd. edition (1989) Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. As appropriate, procedures involving the use of commercially available kits and reagents are generally carried out in accordance with manufacturer defined protocols and/or parameters unless otherwise noted.
  • The terms “advanced prostate cancer”, “locally advanced prostate cancer”, “advanced disease” and “locally advanced disease” mean prostate cancers that have extended through the prostate capsule, and are meant to include stage C disease under the American Urological Association (AUA) system, stage C1-C2 disease under the Whitmore-Jewett system, and stage T3-T4 and N+ disease under the TNM (tumor, node, metastasis) system. In general, surgery is not recommended for patients with locally advanced disease, and these patients have substantially less favorable outcomes compared to patients having clinically localized (organ-confined) prostate cancer. Locally advanced disease is clinically identified by palpable evidence of induration beyond the lateral border of the prostate, or asymmetry or induration above the prostate base. Locally advanced prostate cancer is presently diagnosed pathologically following radical prostatectomy if the tumor invades or penetrates the prostatic capsule, extends into the surgical margin, or invades the seminal vesicles.
  • “Altering the native glycosylation pattern” is intended for purposes herein to mean deleting one or more carbohydrate moieties found in native sequence 58P1D12 (either by removing the underlying glycosylation site or by deleting the glycosylation by chemical and/or enzymatic means), and/or adding one or more glycosylation sites that are not present in the native sequence 58P1D12. In addition, the phrase includes qualitative changes in the glycosylation of the native proteins, involving a change in the nature and proportions of the various carbohydrate moieties present.
  • The term “analog” refers to a molecule which is structurally similar or shares similar or corresponding attributes with another molecule (e.g. a 58P1D12-related protein). For example, an analog of a 58P1D12 protein can be specifically bound by an antibody or T cell that specifically binds to 58P1D12.
  • The term “antibody” is used in the broadest sense. Therefore, an “antibody” can be naturally occurring or man-made such as monoclonal antibodies produced by conventional hybridoma technology. Anti-58P1D12 antibodies comprise monoclonal and polyclonal antibodies as well as fragments containing the antigen-binding domain and/or one or more complementarity determining regions of these antibodies.
  • An “antibody fragment” is defined as at least a portion of the variable region of the immunoglobulin molecule that binds to its target, i.e., the antigen-binding region. In one embodiment it specifically covers single anti-58P1D12 antibodies and clones thereof (including agonist, antagonist and neutralizing antibodies) and anti-58P1D12 antibody compositions with polyepitopic specificity.
  • The term “codon optimized sequences” refers to nucleotide sequences that have been optimized for a particular host species by replacing any codons having a usage frequency of less than about 20%. Nucleotide sequences that have been optimized for expression in a given host species by elimination of spurious polyadenylation sequences, elimination of exon/intron splicing signals, elimination of transposon-like repeats and/or optimization of GC content in addition to codon optimization are referred to herein as an “expression enhanced sequences.”
  • A “combinatorial library” is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis by combining a number of chemical “building blocks” such as reagents. For example, a linear combinatorial chemical library, such as a polypeptide (e.g., mutein) library, is formed by combining a set of chemical building blocks called amino acids in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound). Numerous chemical compounds are synthesized through such combinatorial mixing of chemical building blocks (Gallop et al., J. Med. Chem. 37(9): 1233-1251 (1994)).
  • Preparation and screening of combinatorial libraries is well known to those of skill in the art. Such combinatorial chemical libraries include, but are not limited to, peptide libraries (see, e.g., U.S. Pat. No. 5,010,175, Furka, Pept. Prot. Res. 37:487-493 (1991), Houghton et al., Nature, 354:84-88 (1991)), peptoids (PCT Publication No WO 91/19735), encoded peptides (PCT Publication WO 93/20242), random bio-oligomers (PCT Publication WO 92/00091), benzodiazepines (U.S. Pat. No. 5,288,514), diversomers such as hydantoins, benzodiazepines and dipeptides (Hobbs et al., Proc. Nat. Acad. Sci. USA 90:6909-6913 (1993)), vinylogous polypeptides (Hagihara et al., J. Amer. Chem. Soc. 114:6568 (1992)), nonpeptidal peptidomimetics with a Beta-D-Glucose scaffolding (Hirschmann et al., J. Amer. Chem. Soc. 114:9217-9218 (1992)), analogous organic syntheses of small compound libraries (Chen et al., J. Amer. Chem. Soc. 116:2661 (1994)), oligocarbarnates (Cho, et al., Science 261:1303 (1993)), and/or peptidyl phosphonates (Campbell et al., J. Org. Chem. 59:658 (1994)). See, generally, Gordon et al., J. Med. Chem. 37:1385 (1994), nucleic acid libraries (see, e.g., Stratagene, Corp.), peptide nucleic acid libraries (see, e.g., U.S. Pat. No. 5,539,083), antibody libraries (see, e.g., Vaughn et al., Nature Biotechnology 14(3): 309-314 (1996), and PCT/US96/10287), carbohydrate libraries (see, e.g., Liang et al., Science 274:1520-1522 (1996), and U.S. Pat. No. 5,593,853), and small organic molecule libraries (see, e.g., benzodiazepines, Baum, C&EN, Jan 18, page 33 (1993); isoprenoids, U.S. Pat. No. 5,569,588; thiazolidinones and metathiazanones, U.S. Pat. No. 5,549,974; pyrrolidines, U.S. Pat. Nos. 5,525,735 and 5,519,134; morpholino compounds, U.S. Pat. No. 5,506,337; benzodiazepines, U.S. Pat. No. 5,288,514; and the like).
  • Devices for the preparation of combinatorial libraries are commercially available (see, e.g., 357 NIPS, 390 NIPS, Advanced Chem Tech, Louisville Ky.; Symphony, Rainin, Woburn, Mass.; 433A, Applied Biosystems, Foster City, Calif.; 9050, Plus, Millipore, Bedford, NIA). A number of well-known robotic systems have also been developed for solution phase chemistries. These systems include automated workstations such as the automated synthesis apparatus developed by Takeda Chemical Industries, LTD. (Osaka, Japan) and many robotic systems utilizing robotic arms (Zymate H, Zymark Corporation, Hopkinton, Mass.; Orca, Hewlett-Packard, Palo Alto, Calif.), which mimic the manual synthetic operations performed by a chemist. Any of the above devices are suitable for use with the present invention. The nature and implementation of modifications to these devices (if any) so that they can operate as discussed herein will be apparent to persons skilled in the relevant art. In addition, numerous combinatorial libraries are themselves commercially available (see, e.g., ComGenex, Princeton, N.J.; Asinex, Moscow, RU; Tripos, Inc., St. Louis, Mo.; ChemStar, Ltd, Moscow, RU; 3D Pharmaceuticals, Exton, Pa.; Martek Biosciences, Columbia, Md.; etc.).
  • The term “cytotoxic agent” refers to a substance that inhibits or prevents the expression activity of cells, function of cells and/or causes destruction of cells. The term is intended to include radioactive isotopes chemotherapeutic agents, and toxins such as small molecule toxins or enzymatically active toxins of bacterial, fungal, plant or animal origin, including fragments and/or variants thereof. Examples of cytotoxic agents include, but are not limited to auristatins, auromycins, maytansinoids, yttrium, bismuth, ricin, ricin A-chain, combrestatin, duocarmycins, dolostatins, doxorubicin, daunorubicin, taxol, cisplatin, cc1065, ethidium bromide, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicine, dihydroxy anthracin dione, actinomycin, diphtheria toxin, Pseudomonas exotoxin (PE) A, PE40, abrin, abrin A chain, modeccin A chain, alpha-sarcin, gelonin, mitogellin, retstrictocin, phenomycin, enomycin, curicin, crotin, calicheamicin, Sapaonaria officinalis inhibitor, and glucocorticoid and other chemotherapeutic agents, as well as radioisotopes such as At211, I131, I125, Y90, Re186, Re188, Sm153, Bi212 or 213, P32 and radioactive isotopes of Lu including Lu177. Antibodies may also be conjugated to an anti-cancer pro-drug activating enzyme capable of converting the pro-drug to its active form.
  • The “gene product” is sometimes referred to herein as a protein or mRNA. For example, a “gene product of the invention” is sometimes referred to herein as a “cancer amino acid sequence”, “cancer protein”, “protein of a cancer listed in Table I”, a “cancer mRNA”, “mRNA of a cancer listed in Table I”, etc. In one embodiment, the cancer protein is encoded by a nucleic acid of FIG. 2. The cancer protein can be a fragment, or alternatively, be the full-length protein to the fragment encoded by the nucleic acids of FIG. 2. In one embodiment, a cancer amino acid sequence is used to determine sequence identity or similarity. In another embodiment, the sequences are naturally occurring allelic variants of a protein encoded by a nucleic acid of FIG. 2. In another embodiment, the sequences are sequence variants as further described herein.
  • “High throughput screening” assays for the presence, absence, quantification, or other properties of particular nucleic acids or protein products are well known to those of skill in the art. Similarly, binding assays and reporter gene assays are similarly well known. Thus, e.g., U.S. Pat. No. 5,559,410 discloses high throughput screening methods for proteins; U.S. Pat. No. 5,585,639 discloses high throughput screening methods for nucleic acid binding (i.e., in arrays); while U.S. Pat. Nos. 5,576,220 and 5,541,061 disclose high throughput methods of screening for ligand/antibody binding.
  • In addition, high throughput screening systems are commercially available (see, e.g., Amersham Biosciences, Piscataway, N.J.; Zymark Corp., Hopkinton, Mass.; Air Technical Industries, Mentor, Ohio; Beckman Instruments, Inc. Fullerton, Calif.; Precision Systems, Inc., Natick, Mass.; etc.). These systems typically automate entire procedures, including all sample and reagent pipetting, liquid dispensing, timed incubations, and final readings of the microplate in detector(s) appropriate for the assay. These configurable systems provide high throughput and rapid start up as well as a high degree of flexibility and customization. The manufacturers of such systems provide detailed protocols for various high throughput systems. Thus, e.g., Zymark Corp. provides technical bulletins describing screening systems for detecting the modulation of gene transcription, ligand binding, and the like.
  • The term “homolog” refers to a molecule which exhibits homology to another molecule, by for example, having sequences of chemical residues that are the same or similar at corresponding positions.
  • “Human Leukocyte Antigen” or “HLA” is a human class I or class II Major Histocompatibility Complex (MHC) protein (see, e.g., Stites, et al., Immunology, 8th Ed, Lange Publishing, Los Altos, Calif. (1994).
  • The terms “hybridize”, “hybridizing”, “hybridizes” and the like, used in the context of polynucleotides, are meant to refer to conventional hybridization conditions, preferably such as hybridization in 50% formamide/6×SSC/0.1% SDS/100 μg/ml ssDNA, in which temperatures for hybridization are above 37 degrees C. and temperatures for washing in 0.1×SSC/0.1% SDS are above 55 degrees C.
  • The phrases “isolated” or “biologically pure” refer to material which is substantially or essentially free from components which normally accompany the material as it is found in its native state. Thus, isolated peptides in accordance with the invention preferably do not contain materials normally associated with the peptides in their in situ environment. For example, a polynucleotide is said to be “isolated” when it is substantially separated from contaminant polynucleotides that correspond or are complementary to genes other than the 58P1D12 genes or that encode polypeptides other than 58P1D12 gene product or fragments thereof. A skilled artisan can readily employ nucleic acid isolation procedures to obtain an isolated 58P1D12 polynucleotide. A protein is said to be “isolated,” for example, when physical, mechanical or chemical methods are employed to remove the 58P1D12 proteins from cellular constituents that are normally associated with the protein. A skilled artisan can readily employ standard purification methods to obtain an isolated 58P1D12 protein. Alternatively, an isolated protein can be prepared by chemical means.
  • The term “mammal” refers to any organism classified as a mammal, including mice, rats, rabbits, dogs, cats, cows, horses and humans. In one embodiment of the invention, the mammal is a mouse. In another embodiment of the invention, the mammal is a human.
  • The terms “metastatic prostate cancer” and “metastatic disease” mean prostate cancers that have spread to regional lymph nodes or to distant sites, and are meant to include stage D disease under the AUA system and stage T×N×M+ under the TNM system. As is the case with locally advanced prostate cancer, surgery is generally not indicated for patients with metastatic disease, and hormonal (androgen ablation) therapy is a preferred treatment modality. Patients with metastatic prostate cancer eventually develop an androgen-refractory state within 12 to 18 months of treatment initiation. Approximately half of these androgen-refractory patients die within 6 months after developing that status. The most common site for prostate cancer metastasis is bone. Prostate cancer bone metastases are often osteoblastic rather than osteolytic (i.e., resulting in net bone formation). Bone metastases are found most frequently in the spine, followed by the femur, pelvis, rib cage, skull and humerus. Other common sites for metastasis include lymph nodes, lung, liver and brain. Metastatic prostate cancer is typically diagnosed by open or laparoscopic pelvic lymphadenectomy, whole body radionuclide scans, skeletal radiography, and/or bone lesion biopsy.
  • The term “modulator” or “test compound” or “drug candidate” or grammatical equivalents as used herein describe any molecule, e.g., protein, oligopeptide, small organic molecule, polysaccharide, polynucleotide, etc., to be tested for the capacity to directly or indirectly alter the cancer phenotype or the expression of a cancer sequence, e.g., a nucleic acid or protein sequences, or effects of cancer sequences (e.g., signaling, gene expression, protein interaction, etc.) In one aspect, a modulator will neutralize the effect of a cancer protein of the invention. By “neutralize” is meant that an activity of a protein is inhibited or blocked, along with the consequent effect on the cell. In another aspect, a modulator will neutralize the effect of a gene, and its corresponding protein, of the invention by normalizing levels of said protein. In preferred embodiments, modulators alter expression profiles, or expression profile nucleic acids or proteins provided herein, or downstream effector pathways. In one embodiment, the modulator suppresses a cancer phenotype, e.g. to a normal tissue fingerprint. In another embodiment, a modulator induced a cancer phenotype. Generally, a plurality of assay mixtures is run in parallel with different agent concentrations to obtain a differential response to the various concentrations. Typically, one of these concentrations serves as a negative control, i.e., at zero concentration or below the level of detection.
  • Modulators, drug candidates or test compounds encompass numerous chemical classes, though typically they are organic molecules, preferably small organic compounds having a molecular weight of more than 100 and less than about 2,500 Daltons. Preferred small molecules are less than 2000, or less than 1500 or less than 1000 or less than 500 D. Candidate agents comprise functional groups necessary for structural interaction with proteins, particularly hydrogen bonding, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups. The candidate agents often comprise cyclical carbon or heterocyclic structures and/or aromatic or polyaromatic structures substituted with one or more of the above functional groups. Modulators also comprise biomolecules such as peptides, saccharides, fatty acids, steroids, purines, pyrimidines, derivatives, structural analogs or combinations thereof. Particularly preferred are peptides. One class of modulators are peptides, for example of from about five to about 35 amino acids, with from about five to about 20 amino acids being preferred, and from about 7 to about 15 being particularly preferred. Preferably, the cancer modulatory protein is soluble, includes a non-transmembrane region, and/or, has an N-terminal Cys to aid in solubility. In one embodiment, the C-terminus of the fragment is kept as a free acid and the N-terminus is a free amine to aid in coupling, i.e., to cysteine. In one embodiment, a cancer protein of the invention is conjugated to an immunogenic agent as discussed herein. In one embodiment, the cancer protein is conjugated to BSA. The peptides of the invention, e.g., of preferred lengths, can be linked to each other or to other amino acids to create a longer peptide/protein. The modulatory peptides can be digests of naturally occurring proteins as is outlined above, random peptides, or “biased” random peptides. In a preferred embodiment, peptide/protein-based modulators are antibodies, and fragments thereof, as defined herein.
  • Modulators of cancer can also be nucleic acids. Nucleic acid modulating agents can be naturally occurring nucleic acids, random nucleic acids, or “biased” random nucleic acids. For example, digests of prokaryotic or eukaryotic genomes can be used in an approach analogous to that outlined above for proteins.
  • The term “monoclonal antibody” refers to an antibody obtained from a population of substantially homogeneous antibodies, i.e., the antibodies comprising the population are identical except for possible naturally occurring mutations that are present in minor amounts.
  • A “motif”, as in biological motif of a 58P1D12-related protein, refers to any pattern of amino acids forming part of the primary sequence of a protein, that is associated with a particular function (e.g. protein-protein interaction, protein-DNA interaction, etc) or modification (e.g. that is phosphorylated, glycosylated or amidated), or localization (e.g. secretory sequence, nuclear localization sequence, etc.) or a sequence that is correlated with being immunogenic, either humorally or cellularly. A motif can be either contiguous or capable of being aligned to certain positions that are generally correlated with a certain function or property. In the context of HLA motifs, “motif” refers to the pattern of residues in a peptide of defined length, usually a peptide of from about 8 to about 13 amino acids for a class I HLA motif and from about 6 to about 25 amino acids for a class II HLA motif, which is recognized by a particular HLA molecule. Peptide motifs for HLA binding are typically different for each protein encoded by each human HLA allele and differ in the pattern of the primary and secondary anchor residues.
  • A “pharmaceutical excipient” comprises a material such as an adjuvant, a carrier, pH-adjusting and buffering agents, tonicity adjusting agents, wetting agents, preservative, and the like.
  • “Pharmaceutically acceptable” refers to a non-toxic, inert, and/or composition that is physiologically compatible with humans or other mammals.
  • The term “polynucleotide” means a polymeric form of nucleotides of at least 10 bases or base pairs in length, either ribonucleotides or deoxynucleotides or a modified form of either type of nucleotide, and is meant to include single and double stranded forms of DNA and/or RNA. In the art, this term if often used interchangeably with “oligonucleotide”. A polynucleotide can comprise a nucleotide sequence disclosed herein wherein thymidine (T), as shown for example in FIG. 2, can also be uracil (U); this definition pertains to the differences between the chemical structures of DNA and RNA, in particular the observation that one of the four major bases in RNA is uracil (U) instead of thymidine (T).
  • The term “polypeptide” means a polymer of at least about 4, 5, 6, 7, or 8 amino acids. Throughout the specification, standard three letter or single letter designations for amino acids are used. In the art, this term is often used interchangeably with “peptide” or “protein”.
  • An HLA “primary anchor residue” is an amino acid at a specific position along a peptide sequence which is understood to provide a contact point between the immunogenic peptide and the HLA molecule. One to three, usually two, primary anchor residues within a peptide of defined length generally defines a “motif” for an immunogenic peptide. These residues are understood to fit in close contact with peptide binding groove of an HLA molecule, with their side chains buried in specific pockets of the binding groove. In one embodiment, for example, the primary anchor residues for an HLA class I molecule are located at position 2 (from the amino terminal position) and at the carboxyl terminal position of a 8, 9, 10, 11, or 12 residue peptide epitope in accordance with the invention. Alternatively, in another embodiment, the primary anchor residues of a peptide binds an HLA class II molecule are spaced relative to each other, rather than to the termini of a peptide, where the peptide is generally of at least 9 amino acids in length. The primary anchor positions for each motif and supermotif are set forth in Table IV. For example, analog peptides can be created by altering the presence or absence of particular residues in the primary and/or secondary anchor positions shown in Table IV. Such analogs are used to modulate the binding affinity and/or population coverage of a peptide comprising a particular HLA motif or supermotif.
  • “Radioisotopes” include, but are not limited to the following (non-limiting exemplary uses are also set forth):
  • Examples of Medical Isotopes
  • Isotope Description of use
    Actinium-225 See Thorium-229 (Th-229)
    (AC-225)
    Actinium-227 Parent of Radium-223 (Ra-223) which is an alpha emitter used to treat
    (AC-227) metastases in the skeleton resulting from cancer (i.e., breast and prostate
    cancers), and cancer radioimmunotherapy
    Bismuth-212 See Thorium-228 (Th-228)
    (Bi-212)
    Bismuth-213 See Thorium-229 (Th-229)
    (Bi-213)
    Cadmium-109 Cancer detection
    (Cd-109)
    Cobalt-60 Radiation source for radiotherapy of cancer, for food irradiators, and for
    (Co-60) sterilization of medical supplies
    Copper-64 A positron emitter used for cancer therapy and SPECT imaging
    (Cu-64)
    Copper-67 Beta/gamma emitter used in cancer radioimmunotherapy and diagnostic
    (Cu-67) studies (i.e., breast and colon cancers, and lymphoma)
    Dysprosium-166 Cancer radioimmunotherapy
    (Dy-166)
    Erbium-169 Rheumatoid arthritis treatment, particularly for the small joints associated
    (Er-169) with fingers and toes
    Europium-152 Radiation source for food irradiation and for sterilization of medical
    (Eu-152) supplies
    Europium-154 Radiation source for food irradiation and for sterilization of medical
    (Eu-154) supplies
    Gadolinium-153 Osteoporosis detection and nuclear medical quality assurance devices
    (Gd-153)
    Gold-198 Implant and intracavity therapy of ovarian, prostate, and brain cancers
    (Au-198)
    Holmium-166 Multiple myeloma treatment in targeted skeletal therapy, cancer
    (Ho-166) radioimmunotherapy, bone marrow ablation, and rheumatoid arthritis
    treatment
    Iodine-125 Osteoporosis detection, diagnostic imaging, tracer drugs, brain cancer
    (I-125) treatment, radiolabeling, tumor imaging, mapping of receptors in the
    brain, interstitial radiation therapy, brachytherapy for treatment of prostate
    cancer, determination of glomerular filtration rate (GFR), determination of
    plasma volume, detection of deep vein thrombosis of the legs
    Iodine-131 Thyroid function evaluation, thyroid disease detection, treatment of
    (I-131) thyroid cancer as well as other non-malignant thyroid diseases (i.e.,
    Graves disease, goiters, and hyperthyroidism), treatment of leukemia,
    lymphoma, and other forms of cancer (e.g., breast cancer) using
    radioimmunotherapy
    Iridium-192 Brachytherapy, brain and spinal cord tumor treatment, treatment of
    (Ir-192) blocked arteries (i.e., arteriosclerosis and restenosis), and implants for
    breast and prostate tumors
    Lutetium-177 Cancer radioimmunotherapy and treatment of blocked arteries (i.e.,
    (Lu-177) arteriosclerosis and restenosis)
    Molybdenum-99 Parent of Technetium-99m (Tc-99m) which is used for imaging the brain,
    (Mo-99) liver, lungs, heart, and other organs. Currently, Tc-99m is the most widely
    used radioisotope used for diagnostic imaging of various cancers and
    diseases involving the brain, heart, liver, lungs; also used in detection of
    deep vein thrombosis of the legs
    Osmium-194 Cancer radioimmunotherapy
    (Os-194)
    Palladium-103 Prostate cancer treatment
    (Pd-103)
    Platinum-195m Studies on biodistribution and metabolism of cisplatin, a chemotherapeutic
    (Pt-195m) drug
    Phosphorus-32 Polycythemia rubra vera (blood cell disease) and leukemia treatment, bone
    (P-32) cancer diagnosis/treatment; colon, pancreatic, and liver cancer treatment;
    radiolabeling nucleic acids for in vitro research, diagnosis of superficial
    tumors, treatment of blocked arteries (i.e., arteriosclerosis and restenosis),
    and intracavity therapy
    Phosphorus-33 Leukemia treatment, bone disease diagnosis/treatment, radiolabeling, and
    (P-33) treatment of blocked arteries (i.e., arteriosclerosis and restenosis)
    Radium-223 See Actinium-227 (Ac-227)
    (Ra-223)
    Rhenium-186 Bone cancer pain relief, rheumatoid arthritis treatment, and diagnosis and
    (Re-186) treatment of lymphoma and bone, breast, colon, and liver cancers using
    radioimmunotherapy
    Rhenium-188 Cancer diagnosis and treatment using radioimmunotherapy, bone cancer
    (Re-188) pain relief, treatment of rheumatoid arthritis, and treatment of prostate
    cancer
    Rhodium-105 Cancer radioimmunotherapy
    (Rh-105)
    Samarium-145 Ocular cancer treatment
    (Sm-145)
    Samarium-153 Cancer radioimmunotherapy and bone cancer pain relief
    (Sm-153)
    Scandium-47 Cancer radioimmunotherapy and bone cancer pain relief
    (Sc-47)
    Selenium-75 Radiotracer used in brain studies, imaging of adrenal cortex by gamma-
    (Se-75) scintigraphy, lateral locations of steroid secreting tumors, pancreatic
    scanning, detection of hyperactive parathyroid glands, measure rate of bile
    acid loss from the endogenous pool
    Strontium-85 Bone cancer detection and brain scans
    (Sr-85)
    Strontium-89 Bone cancer pain relief, multiple myeloma treatment, and osteoblastic
    (Sr-89) therapy
    Technetium-99m See Molybdenum-99 (Mo-99)
    (Tc-99m)
    Thorium-228 Parent of Bismuth-212 (Bi-212) which is an alpha emitter used in cancer
    (Th-228) radioimmunotherapy
    Thorium-229 Parent of Actinium-225 (Ac-225) and grandparent of Bismuth-213 (Bi-
    (Th-229) 213) which are alpha emitters used in cancer radioimmunotherapy
    Thulium-170 Gamma source for blood irradiators, energy source for implanted medical
    (Tm-170) devices
    Tin-117m Cancer immunotherapy and bone cancer pain relief
    (Sn-117m)
    Tungsten-188 Parent for Rhenium-188 (Re-188) which is used for cancer
    (W-188) diagnostics/treatment, bone cancer pain relief, rheumatoid arthritis
    treatment, and treatment of blocked arteries (i.e., arteriosclerosis and
    restenosis)
    Xenon-127 Neuroimaging of brain disorders, high resolution SPECT studies,
    (Xe-127) pulmonary function tests, and cerebral blood flow studies
    Ytterbium-175 Cancer radioimmunotherapy
    (Yb-175)
    Yttrium-90 Microseeds obtained from irradiating Yttrium-89 (Y-89) for liver cancer
    (Y-90) treatment
    Yttrium-91 A gamma-emitting label for Yttrium-90 (Y-90) which is used for cancer
    (Y-91) radioimmunotherapy (i.e., lymphoma, breast, colon, kidney, lung, ovarian,
    prostate, pancreatic, and inoperable liver cancers)
  • By “randomized” or grammatical equivalents as herein applied to nucleic acids and proteins is meant that each nucleic acid and peptide consists of essentially random nucleotides and amino acids, respectively. These random peptides (or nucleic acids, discussed herein) can incorporate any nucleotide or amino acid at any position. The synthetic process can be designed to generate randomized proteins or nucleic acids, to allow the formation of all or most of the possible combinations over the length of the sequence, thus forming a library of randomized candidate bioactive proteinaceous agents.
  • In one embodiment, a library is “fully randomized,” with no sequence preferences or constants at any position. In another embodiment, the library is a “biased random” library. That is, some positions within the sequence either are held constant, or are selected from a limited number of possibilities. For example, the nucleotides or amino acid residues are randomized within a defined class, e.g., of hydrophobic amino acids, hydrophilic residues, sterically biased (either small or large) residues, towards the creation of nucleic acid binding domains, the creation of cysteines, for cross-linking, prolines for SH-3 domains, serines, threonines, tyrosines or histidines for phosphorylation sites, etc., or to purines, etc.
  • A “recombinant” DNA or RNA molecule is a DNA or RNA molecule that has been subjected to molecular manipulation in vitro.
  • Non-limiting examples of small molecules include compounds that bind or interact with 58P1D12, ligands including hormones, neuropeptides, chemokines, odorants, phospholipids, and functional equivalents thereof that bind and preferably inhibit 58P1D12 protein function. Such non-limiting small molecules preferably have a molecular weight of less than about 10 kDa, more preferably below about 9, about 8, about 7, about 6, about 5 or about 4 kDa. In certain embodiments, small molecules physically associate with, or bind, 58P1D12 protein; are not found in naturally occurring metabolic pathways; and/or are more soluble in aqueous than non-aqueous solutions
  • “Stringency” of hybridization reactions is readily determinable by one of ordinary skill in the art, and generally is an empirical calculation dependent upon probe length, washing temperature, and salt concentration. In general, longer probes require higher temperatures for proper annealing, while shorter probes need lower temperatures. Hybridization generally depends on the ability of denatured nucleic acid sequences to reanneal when complementary strands are present in an environment below their melting temperature. The higher the degree of desired homology between the probe and hybridizable sequence, the higher the relative temperature that can be used. As a result, it follows that higher relative temperatures would tend to make the reaction conditions more stringent, while lower temperatures less so. For additional details and explanation of stringency of hybridization reactions, see Ausubel et al., Current Protocols in Molecular Biology, Wiley Interscience Publishers, (1995).
  • “Stringent conditions” or “high stringency conditions”, as defined herein, are identified by, but not limited to, those that: (1) employ low ionic strength and high temperature for washing, for example 0.015 M sodium chloride/0.0015 M sodium citrate/0.1% sodium dodecyl sulfate at 50° C.; (2) employ during hybridization a denaturing agent, such as formamide, for example, 50% (v/v) formamide with 0.1% bovine serum albumin/0.1% Ficoll/0.1% polyvinylpyrrolidone/50 mM sodium phosphate buffer at pH 6.5 with 750 mM sodium chloride, 75 mM sodium citrate at 42° C.; or (3) employ 50% formamide, 5×SSC (0.75 M NaCl, 0.075 M sodium citrate), 50 mM sodium phosphate (pH 6.8), 0.1% sodium pyrophosphate, 5×Denhardt's solution, sonicated salmon sperm DNA (50 μg/ml), 0.1% SDS, and 10% dextran sulfate at 42° C., with washes at 42° C. in 0.2×SSC (sodium chloride/sodium. citrate) and 50% formamide at 55° C., followed by a high-stringency wash consisting of 0.1×SSC containing EDTA at 55° C. “Moderately stringent conditions” are described by, but not limited to, those in Sambrook et al., Molecular Cloning: A Laboratory Manual, New York: Cold Spring Harbor Press, 1989, and include the use of washing solution and hybridization conditions (e.g., temperature, ionic strength and % SDS) less stringent than those described above. An example of moderately stringent conditions is overnight incubation at 37° C. in a solution comprising: 20% formamide, 5×SSC (150 mM NaCl, 15 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5×Denhardt's solution, 10% dextran sulfate, and 20 mg/mL denatured sheared salmon sperm DNA, followed by washing the filters in 1×SSC at about 37-50° C. The skilled artisan will recognize how to adjust the temperature, ionic strength, etc. as necessary to accommodate factors such as probe length and the like.
  • An HLA “supermotif” is a peptide binding specificity shared by HLA molecules encoded by two or more HLA alleles. Overall phenotypic frequencies of HLA-supertypes in different ethnic populations are set forth in Table IV (F). The non-limiting constituents of various supetypes are as follows:
  • A2: A*0201, A*0202, A*0203, A*0204, A*0205, A*0206, A*6802, A*6901, A*0207
  • A3: A3, A11, A31, A*3301, A*6801, A*0301, A*1101, A*3101
  • B7: B7, B*3501-03, B*51, B*5301, B*5401, B*5501, B*5502, B*5601, B*6701, B*7801, B*0702, B*5101, B*5602
  • B44: B*3701, B*4402, B*4403, B*60 (B*4001), B61 (B*4006)
  • A1: A*0102, A*2604, A*3601, A*4301, A*8001
  • A24: A*24, A*30, A*2403, A*2404, A*3002, A*3003
  • B27: B*1401-02, B*1503, B*1509, B*1510, B*1518, B*3801-02, B*3901, B*3902, B*3903-04, B*4801-02, B*7301, B*2701-08
  • B58: B*1516, B*1517, B*5701, B*5702, B58
  • B62: B*4601, B52, B*1501 (B62), B*1502 (B75), B*1513 (B77)
  • Calculated population coverage afforded by different HLA-supertype combinations are set forth in Table IV (G).
  • As used herein “to treat” or “therapeutic” and grammatically related terms, refer to any improvement of any consequence of disease, such as prolonged survival, less morbidity, and/or a lessening of side effects which are the byproducts of an alternative therapeutic modality; full eradication of disease is not required.
  • A “transgenic animal” (e.g., a mouse or rat) is an animal having cells that contain a transgene, which transgene was introduced into the animal or an ancestor of the animal at a prenatal, e.g., an embryonic stage. A “transgene” is a DNA that is integrated into the genome of a cell from which a transgenic animal develops.
  • As used herein, an HLA or cellular immune response “vaccine” is a composition that contains or encodes one or more peptides of the invention. There are numerous embodiments of such vaccines, such as a cocktail of one or more individual peptides; one or more peptides of the invention comprised by a polyepitopic peptide; or nucleic acids that encode such individual peptides or polypeptides, e.g., a minigene that encodes a polyepitopic peptide. The “one or more peptides” can include any whole unit integer from 1-150 or more, e.g., at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, or 150 or more peptides of the invention. The peptides or polypeptides can optionally be modified, such as by lipidation, addition of targeting or other sequences. HLA class I peptides of the invention can be admixed with, or linked to, HLA class II peptides, to facilitate activation of both cytotoxic T lymphocytes and helper T lymphocytes. HLA vaccines can also comprise peptide-pulsed antigen presenting cells, e.g., dendritic cells.
  • The term “variant” refers to a molecule that exhibits a variation from a described type or norm, such as a protein that has one or more different amino acid residues in the corresponding position(s) of a specifically described protein (e.g. the 58P1D12 protein shown in FIG. 2 or FIG. 3. An analog is an example of a variant protein. Splice isoforms and single nucleotides polymorphisms (SNPs) are further examples of variants.
  • The “58P1D12-related proteins” of the invention include those specifically identified herein, as well as allelic variants, conservative substitution variants, analogs and homologs that can be isolated/generated and characterized without undue experimentation following the methods outlined herein or readily available in the art. Fusion proteins that combine parts of different 58P1D12 proteins or fragments thereof, as well as fusion proteins of a 58P1D12 protein and a heterologous polypeptide are also included. Such 58P1D12 proteins are collectively referred to as the 58P1D12-related proteins, the proteins of the invention, or 58P1D12. The term “58P1D12-related protein” refers to a polypeptide fragment or a 58P1D12 protein sequence of 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, or more than 25 amino acids; or, at least 30, 35, 40, 45, 50, 55, 60, 65, 70, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 275, 300, 325, 350, 375, 400, 425, 450, 475, 500, 525, 550, 575, or 576 or more amino acids.
  • II.) 58P1D12 POLYNUCLEOTIDES
  • One aspect of the invention provides polynucleotides corresponding or complementary to all or part of a 58P1D12 gene, mRNA, and/or coding sequence, preferably in isolated form, including polynucleotides encoding a 58P1D12-related protein and fragments thereof, DNA, RNA, DNA/RNA hybrid, and related molecules, polynucleotides or oligonucleotides complementary to a 58P1D12 gene or mRNA sequence or a part thereof, and polynucleotides or oligonucleotides that hybridize to a 58P1D12 gene, mRNA, or to a 58P1D12 encoding polynucleotide (collectively, “58P1D12 polynucleotides”). In all instances when referred to in this section, T can also be U in FIG. 2.
  • Embodiments of a 58P1D12 polynucleotide include: a 58P1D12 polynucleotide having the sequence shown in FIG. 2, the nucleotide sequence of 58P1D12 as shown in FIG. 2 wherein T is U; at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in FIG. 2; or, at least 10 contiguous nucleotides of a polynucleotide having the sequence as shown in FIG. 2 where T is U. For example, embodiments of 58P1D12 nucleotides comprise, without limitation:
  • (I) a polynucleotide comprising, consisting essentially of, or consisting of a sequence as shown in FIG. 2, wherein T can also be U;
  • (II) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2A, from nucleotide residue number 380 through nucleotide residue number 1201, including the stop codon, wherein T can also be U;
  • (III) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2B, from nucleotide residue number 388 through nucleotide residue number 1086, including the stop codon, wherein T can also be U;
  • (IV) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2C, from nucleotide residue number 206 through nucleotide residue number 904, including the a stop codon, wherein T can also be U;
  • (V) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2D, from nucleotide residue number 206 through nucleotide residue number 916, including the stop codon, wherein T can also be U;
  • (VI) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as shown in FIG. 2E, from nucleotide residue number 106 through nucleotide residue number 816, including the stop codon, wherein T can also be U;
  • (VII) a polynucleotide comprising, consisting essentially of, or consisting of the sequence as set forth in FIG. 2F, wherein T can also be U;
  • (VIII) a polynucleotide that encodes a 58P1D12-related protein that is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% homologous to an entire amino acid sequence shown in FIG. 2A-F;
  • (IX) a polynucleotide that encodes a 58P1D12-related protein that is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% identical to an entire amino acid sequence shown in FIG. 2A-F;
  • (X) a polynucleotide that encodes at least one peptide set forth in Tables VIII-XXI and XXII-XLIX;
  • (XI) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3A in any whole number increment up to 273 that includes at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of FIG. 5;
  • (XII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3A in any whole number increment up to 273 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of FIG. 6;
  • (XIII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3A in any whole number increment up to 273 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of FIG. 7;
  • (XIV) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3A in any whole number increment up to 273 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of FIG. 8;
  • (XV) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3A in any whole number increment up to 273 that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of FIG. 9;
  • (XVI) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3B, and/or 3C in any whole number increment up to 232 and/or 236 respectively that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of FIG. 5;
  • (XVII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3B and/or 3C in any whole number increment up to 232 and/or 236 respectively that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of FIG. 6;
  • (XVIII) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3B and/or 3C in any whole number increment up to 232 and/or 236 respectively that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of FIG. 7;
  • (XIX) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3B and/or 3C in any whole number increment up to 232 and/or 236 respectively that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of FIG. 8;
  • (XX) a polynucleotide that encodes a peptide region of at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a peptide of FIG. 3B and/or 3C in any whole number increment up to 232 and/or 236 respectively that includes 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of FIG. 9;
  • (XXI) a polynucleotide that is fully complementary to a polynucleotide of any one of (I)-(XX);
  • (XXII) a polynucleotide that is fully complementary to a polynucleotide of any one of (I)-(XXI);
  • (XXIII) a peptide that is encoded by any of (I) to (XXII); and;
  • (XXIV) a composition comprising a polynucleotide of any of (I)-(XXII) or peptide of (XXIII) together with a pharmaceutical excipient and/or in a human unit dose form;
  • (XXV) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to modulate a cell expressing 58P1D12;
  • (XXVI) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing 58P1D12;
  • (XXVII) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing 58P1D12, said cell from a cancer of a tissue listed in Table I;
  • (XXVIII) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to diagnose, prophylax, prognose, or treat a a cancer;
  • (XXIX) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to diagnose, prophylax, prognose, or treat a a cancer of a tissue listed in Table I; and;
  • (XXX) a method of using a polynucleotide of any (I)-(XXII) or peptide of (XXIII) or a composition of (XXIV) in a method to identify or characterize a modulator of a cell expressing 58P1D12.
  • As used herein, a range is understood to disclose specifically all whole unit positions thereof.
  • Typical embodiments of the invention disclosed herein include 58P1D12 polynucleotides that encode specific portions of 58P1D12 mRNA sequences (and those which are complementary to such sequences) such as those that encode the proteins and/or fragments thereof, for example: [!!! revise to include up to the appropriate length].
  • (a) 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 255, 260, 265, 270, and 273 or more contiguous amino acids of 58P1D12 variant 1; the maximal lengths relevant for other variants are: variant 2, 232 amino acids; and variant 3, 236 amino acids.
  • For example, representative embodiments of the invention disclosed herein include: polynucleotides and their encoded peptides themselves encoding about amino acid 1 to about amino acid 10 of the 58P1D12 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 10 to about amino acid 20 of the 58P1D12 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 20 to about amino acid 30 of the 58P1D12 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 30 to about amino acid 40 of the 58P1D12 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 40 to about amino acid 50 of the 58P1D12 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 50 to about amino acid 60 of the 58P1D12 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 60 to about amino acid 70 of the 58P1D12 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 70 to about amino acid 80 of the 58P1D12 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 80 to about amino acid 90 of the 58P1D12 protein shown in FIG. 2 or FIG. 3, polynucleotides encoding about amino acid 90 to about amino acid 100 of the 58P1D12 protein shown in FIG. 2 or FIG. 3, in increments of about 10 amino acids, ending at the carboxyl terminal amino acid set forth in FIG. 2 or FIG. 3. Accordingly, polynucleotides encoding portions of the amino acid sequence (of about 10 amino acids), of amino acids, 100 through the carboxyl terminal amino acid of the 58P1D12 protein are embodiments of the invention. Wherein it is understood that each particular amino acid position discloses that position plus or minus five amino acid residues.
  • Polynucleotides encoding relatively long portions of a 58P1D12 protein are also within the scope of the invention. For example, polynucleotides encoding from about amino acid 1 (or 20 or 30 or 40 etc.) to about amino acid 20, (or 30, or 40 or 50 etc.) of the 58P1D12 protein “or variant” shown in FIG. 2 or FIG. 3 can be generated by a variety of techniques well known in the art. These polynucleotide fragments can include any portion of the 58P1D12 sequence as shown in FIG. 2.
  • Additional illustrative embodiments of the invention disclosed herein include 58P1D12 polynucleotide fragments encoding one or more of the biological motifs contained within a 58P1D12 protein “or variant” sequence, including one or more of the motif-bearing subsequences of a 58P1D12 protein “or variant” set forth in Tables VIII-XXI and XXII-XLIX. In another embodiment, typical polynucleotide fragments of the invention encode one or more of the regions of 58P1D12 protein or variant that exhibit homology to a known molecule. In another embodiment of the invention, typical polynucleotide fragments can encode one or more of the 58P1D12 protein or variant N-glycosylation sites, cAMP and cGMP-dependent protein kinase phosphorylation sites, casein kinase II phosphorylation sites or N-myristoylation site and amidation sites.
  • Note that to determine the starting position of any peptide set forth in Tables VIII-XXI and Tables XXII to XLIX (collectively HLA Peptide Tables) respective to its parental protein, e.g., variant 1, variant 2, etc., reference is made to three factors: the particular variant, the length of the peptide in an HLA Peptide Table, and the Search Peptides listed in Table VII. Generally, a unique Search Peptide is used to obtain HLA peptides for a particular variant. The position of each Search Peptide relative to its respective parent molecule is listed in Table VII. Accordingly, if a Search Peptide begins at position “X”, one must add the value “X minus 1” to each position in Tables VIII-XXI and Tables XXII-IL to obtain the actual position of the HLA peptides in their parental molecule. For example if a particular Search Peptide begins at position 150 of its parental molecule, one must add 150-1, i.e., 149 to each HLA peptide amino acid position to calculate the position of that amino acid in the parent molecule.
  • II.A.) Uses of 58P1D12 Polynucleotides
  • II.A.1.) Monitoring of Genetic Abnormalities
  • The polynucleotides of the preceding paragraphs have a number of different specific uses. The human 58P1D12 gene maps to the chromosomal location set forth in the Example entitled “Chromosomal Mapping of 58P1D12.” For example, because the 58P1D12 gene maps to this chromosome, polynucleotides that encode different regions of the 58P1D12 proteins are used to characterize cytogenetic abnormalities of this chromosomal locale, such as abnormalities that are identified as being associated with various cancers. In certain genes, a variety of chromosomal abnormalities including rearrangements have been identified as frequent cytogenetic abnormalities in a number of different cancers (see e.g. Krajinovic et al., Mutat. Res. 382(3-4): 81-83 (1998); Johansson et al., Blood 86(10): 3905-3914 (1995) and Finger et al., P.N.A.S. 85(23): 9158-9162 (1988)). Thus, polynucleotides encoding specific regions of the 58P1D12 proteins provide new tools that can be used to delineate, with greater precision than previously possible, cytogenetic abnormalities in the chromosomal region that encodes 58P1D12 that may contribute to the malignant phenotype. In this context, these polynucleotides satisfy a need in the art for expanding the sensitivity of chromosomal screening in order to identify more subtle and less common chromosomal abnormalities (see e.g. Evans et al., Am. J. Obstet. Gynecol 171(4): 1055-1057 (1994)).
  • Furthermore, as 58P1D12 was shown to be highly expressed in prostate and other cancers, 58P1D12 polynucleotides are used in methods assessing the status of 58P1D12 gene products in normal versus cancerous tissues. Typically, polynucleotides that encode specific regions of the 58P1D12 proteins are used to assess the presence of perturbations (such as deletions, insertions, point mutations, or alterations resulting in a loss of an antigen etc.) in specific regions of the 58P1D12 gene, such as regions containing one or more motifs. Exemplary assays include both RT-PCR assays as well as single-strand conformation polymorphism (SSCP) analysis (see, e.g., Marrogi et al., J. Cutan. Pathol. 26(8): 369-378 (1999), both of which utilize polynucleotides encoding specific regions of a protein to examine these regions within the protein.
  • II.A.2.) Antisense Embodiments
  • Other specifically contemplated nucleic acid related embodiments of the invention disclosed herein are genomic DNA, cDNAs, ribozymes, and antisense molecules, as well as nucleic acid molecules based on an alternative backbone, or including alternative bases, whether derived from natural sources or synthesized, and include molecules capable of inhibiting the RNA or protein expression of 58P1D12. For example, antisense molecules can be RNAs or other molecules, including peptide nucleic acids (PNAs) or non-nucleic acid molecules such as phosphorothioate derivatives that specifically bind DNA or RNA in a base pair-dependent manner. A skilled artisan can readily obtain these classes of nucleic acid molecules using the 58P1D12 polynucleotides and polynucleotide sequences disclosed herein.
  • Antisense technology entails the administration of exogenous oligonucleotides that bind to a target polynucleotide located within the cells. The term “antisense” refers to the fact that such oligonucleotides are complementary to their intracellular targets, e.g., 58P1D12. See for example, Jack Cohen, Oligodeoxynucleotides, Antisense Inhibitors of Gene Expression, CRC Press, 1989; and Synthesis 1:1-5 (1988). The 58P1D12 antisense oligonucleotides of the present invention include derivatives such as S-oligonucleotides (phosphorothioate derivatives or S-oligos, see, Jack Cohen, supra), which exhibit enhanced cancer cell growth inhibitory action. S-oligos (nucleoside phosphorothioates) are isoelectronic analogs of an oligonucleotide (O-oligo) in which a nonbridging oxygen atom of the phosphate group is replaced by a sulfur atom. The S-oligos of the present invention can be prepared by treatment of the corresponding O-oligos with 3H-1,2-benzodithiol-3-one-1,1-dioxide, which is a sulfur transfer reagent. See, e.g., Iyer, R. P. et al., J. Org. Chem. 55:4693-4698 (1990); and Iyer, R. P. et al., J. Am. Chem. Soc. 112:1253-1254 (1990). Additional 58P1D12 antisense oligonucleotides of the present invention include morpholino antisense oligonucleotides known in the art (see, e.g., Partridge et al., 1996, Antisense & Nucleic Acid Drug Development 6: 169-175).
  • The 58P1D12 antisense oligonucleotides of the present invention typically can be RNA or DNA that is complementary to and stably hybridizes with the first 100 5′ codons or last 100 3′ codons of a 58P1D12 genomic sequence or the corresponding mRNA. Absolute complementarity is not required, although high degrees of complementarity are preferred. Use of an oligonucleotide complementary to this region allows for the selective hybridization to 58P1D12 mRNA and not to mRNA specifying other regulatory subunits of protein kinase. In one embodiment, 58P1D12 antisense oligonucleotides of the present invention are 15 to 30-mer fragments of the antisense DNA molecule that have a sequence that hybridizes to 58P1D12 mRNA. Optionally, 58P1D12 antisense oligonucleotide is a 30-mer oligonucleotide that is complementary to a region in the first 10 5′ codons or last 10 3′ codons of 58P1D12. Alternatively, the antisense molecules are modified to employ ribozymes in the inhibition of 58P1D12 expression, see, e.g., L. A. Couture & D. T. Stinchcomb; Trends Genet. 12: 510-515 (1996).
  • II.A.3.) Primers and Primer Pairs
  • Further specific embodiments of these nucleotides of the invention include primers and primer pairs, which allow the specific amplification of polynucleotides of the invention or of any specific parts thereof, and probes that selectively or specifically hybridize to nucleic acid molecules of the invention or to any part thereof. Probes can be labeled with a detectable marker, such as, for example, a radioisotope, fluorescent compound, bioluminescent compound, a chemiluminescent compound, metal chelator or enzyme. Such probes and primers are used to detect the presence of a 58P1D12 polynucleotide in a sample and as a means for detecting a cell expressing a 58P1D12 protein.
  • Examples of such probes include polypeptides comprising all or part of the human 58P1D12 cDNA sequence shown in FIG. 2. Examples of primer pairs capable of specifically amplifying 58P1D12 mRNAs are also described in the Examples. As will be understood by the skilled artisan, a great many different primers and probes can be prepared based on the sequences provided herein and used effectively to amplify and/or detect a 58P1D12 mRNA.
  • The 58P1D12 polynucleotides of the invention are useful for a variety of purposes, including but not limited to their use as probes and primers for the amplification and/or detection of the 58P1D12 gene(s), mRNA(s), or fragments thereof; as reagents for the diagnosis and/or prognosis of prostate cancer and other cancers; as coding sequences capable of directing the expression of 58P1D12 polypeptides; as tools for modulating or inhibiting the expression of the 58P1D12 gene(s) and/or translation of the 58P1D12 transcript(s); and as therapeutic agents.
  • The present invention includes the use of any probe as described herein to identify and isolate a 58P1D12 or 58P1D12 related nucleic acid sequence from a naturally occurring source, such as humans or other mammals, as well as the isolated nucleic acid sequence per se, which would comprise all or most of the sequences found in the probe used.
  • II.A.4.) Isolation of 58P1D12-Encoding Nucleic Acid Molecules
  • The 58P1D12 cDNA sequences described herein enable the isolation of other polynucleotides encoding 58P1D12 gene product(s), as well as the isolation of polynucleotides encoding 58P1D12 gene product homologs, alternatively spliced isoforms, allelic variants, and mutant forms of a 58P1D12 gene product as well as polynucleotides that encode analogs of 58P1D12-related proteins. Various molecular cloning methods that can be employed to isolate full length cDNAs encoding a 58P1D12 gene are well known (see, for example, Sambrook, J. et al., Molecular Cloning: A Laboratory Manual, 2d edition, Cold Spring Harbor Press, New York, 1989; Current Protocols in Molecular Biology. Ausubel et al., Eds., Wiley and Sons, 1995). For example, lambda phage cloning methodologies can be conveniently employed, using commercially available cloning systems (e.g., Lambda ZAP Express, Stratagene). Phage clones containing 58P1D12 gene cDNAs can be identified by probing with a labeled 58P1D12 cDNA or a fragment thereof. For example, in one embodiment, a 58P1D12 cDNA (e.g., FIG. 2) or a portion thereof can be synthesized and used as a probe to retrieve overlapping and full-length cDNAs corresponding to a 58P1D12 gene. A 58P1D12 gene itself can be isolated by screening genomic DNA libraries, bacterial artificial chromosome libraries (BACs), yeast artificial chromosome libraries (YACs), and the like, with 58P1D12 DNA probes or primers.
  • II.A.5.) Recombinant Nucleic Acid Molecules and Host-Vector Systems
  • The invention also provides recombinant DNA or RNA molecules containing a 58P1D12 polynucleotide, a fragment, analog or homologue thereof, including but not limited to phages, plasmids, phagemids, cosmids, YACs, BACs, as well as various viral and non-viral vectors well known in the art, and cells transformed or transfected with such recombinant DNA or RNA molecules. Methods for generating such molecules are well known (see, for example, Sambrook et al., 1989, supra).
  • The invention further provides a host-vector system comprising a recombinant DNA molecule containing a 58P1D12 polynucleotide, fragment, analog or homologue thereof within a suitable prokaryotic or eukaryotic host cell. Examples of suitable eukaryotic host cells include a yeast cell, a plant cell, or an animal cell, such as a mammalian cell or an insect cell (e.g., a baculovirus-infectible cell such as an Sf9 or HighFive cell). Examples of suitable mammalian cells include various prostate cancer cell lines such as DU145 and TsuPrl, other transfectable or transducible prostate cancer cell lines, primary cells (PrEC), as well as a number of mammalian cells routinely used for the expression of recombinant proteins (e.g., COS, CHO, 293, 293T cells). More particularly, a polynucleotide comprising the coding sequence of 58P1D12 or a fragment, analog or homolog thereof can be used to generate 58P1D12 proteins or fragments thereof using any number of host-vector systems routinely used and widely known in the art.
  • A wide range of host-vector systems suitable for the expression of 58P1D12 proteins or fragments thereof are available, see for example, Sambrook et al., 1989, supra; Current Protocols in Molecular Biology, 1995, supra). Preferred vectors for mammalian expression include but are not limited to pcDNA 3.1 myc-His-tag (Invitrogen) and the retroviral vector pSRatkneo (Muller et al., 1991, MCB 11:1785). Using these expression vectors, 58P1D12 can be expressed in several prostate cancer and non-prostate cell lines, including for example 293, 293T, rat-1, NIH 3T3 and TsuPr1. The host-vector systems of the invention are useful for the production of a 58P1D12 protein or fragment thereof. Such host-vector systems can be employed to study the functional properties of 58P1D12 and 58P1D12 mutations or analogs.
  • Recombinant human 58P1D12 protein or an analog or homolog or fragment thereof can be produced by mammalian cells transfected with a construct encoding a 58P1D12-related nucleotide. For example, 293T cells can be transfected with an expression plasmid encoding 58P1D12 or fragment, analog or homolog thereof, a 58P1D12-related protein is expressed in the 293T cells, and the recombinant 58P1D12 protein is isolated using standard purification methods (e.g., affinity purification using anti-58P1D12 antibodies). In another embodiment, a 58P1D12 coding sequence is subcloned into the retroviral vector pSRαMSVtkneo and used to infect various mammalian cell lines, such as NIH 3T3, TsuPr1, 293 and rat-1 in order to establish 58P1D12 expressing cell lines. Various other expression systems well known in the art can also be employed. Expression constructs encoding a leader peptide joined in frame to a 58P1D12 coding sequence can be used for the generation of a secreted form of recombinant 58P1D12 protein.
  • As discussed herein, redundancy in the genetic code permits variation in 58P1D12 gene sequences. In particular, it is known in the art that specific host species often have specific codon preferences, and thus one can adapt the disclosed sequence as preferred for a desired host. For example, preferred analog codon sequences typically have rare codons (i.e., codons having a usage frequency of less than about 20% in known sequences of the desired host) replaced with higher frequency codons. Codon preferences for a specific species are calculated, for example, by utilizing codon usage tables available on the INTERNET such as at URL dna.affrc.go.jp/˜nakamura/codon.html.
  • Additional sequence modifications are known to enhance protein expression in a cellular host. These include elimination of sequences encoding spurious polyadenylation signals, exon/intron splice site signals, transposon-like repeats, and/or other such well-characterized sequences that are deleterious to gene expression. The GC content of the sequence is adjusted to levels average for a given cellular host, as calculated by reference to known genes expressed in the host cell. Where possible, the sequence is modified to avoid predicted hairpin secondary mRNA structures. Other useful modifications include the addition of a translational initiation consensus sequence at the start of the open reading frame, as described in Kozak, Mol. Cell Biol., 9:5073-5080 (1989). Skilled artisans understand that the general rule that eukaryotic ribosomes initiate translation exclusively at the 5′ proximal AUG codon is abrogated only under rare conditions (see, e.g., Kozak PNAS 92(7): 2662-2666, (1995) and Kozak NAR 15(20): 8125-8148 (1987)).
  • III.) 58P1D12-Related Proteins
  • Another aspect of the present invention provides 58P1D12-related proteins. Specific embodiments of 58P1D12 proteins comprise a polypeptide having all or part of the amino acid sequence of human 58P1D12 as shown in FIG. 2 or FIG. 3. Alternatively, embodiments of 58P1D12 proteins comprise variant, homolog or analog polypeptides that have alterations in the amino acid sequence of 58P1D12 shown in FIG. 2 or FIG. 3.
  • Embodiments of a 58P1D12 polypeptide include: a 58P1D12 polypeptide having a sequence shown in FIG. 2, a peptide sequence of a 58P1D12 as shown in FIG. 2 wherein T is U; at least 10 contiguous nucleotides of a polypeptide having the sequence as shown in FIG. 2; or, at least 10 contiguous peptides of a polypeptide having the sequence as shown in FIG. 2 where T is U. For example, embodiments of 58P1D12 peptides comprise, without limitation:
  • (I) a protein comprising, consisting essentially of, or consisting of an amino acid sequence as shown in FIG. 2A-F or FIG. 3A-C;
  • (II) a 58P1D12-related protein that is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% homologous to an entire amino acid sequence shown in FIG. 2A-F or 3A-C;
  • (III) a 58P1D12-related protein that is at least 90, 91, 92, 93, 94, 95, 96, 97, 98, 99 or 100% identical to an entire amino acid sequence shown in FIG. 2A-F or 3A-C;
  • (IV) a protein that comprises at least one peptide set forth in Tables VIII to XLIX, optionally with a proviso that it is not an entire protein of FIG. 2;
  • (V) a protein that comprises at least one peptide set forth in Tables VIII-XXI, collectively, which peptide is also set forth in Tables XXII to XLIX, collectively, optionally with a proviso that it is not an entire protein of FIG. 2;
  • (VI) a protein that comprises at least two peptides selected from the peptides set forth in Tables VIII-XLIX, optionally with a proviso that it is not an entire protein of FIG. 2;
  • (VII) a protein that comprises at least two peptides selected from the peptides set forth in Tables VIII to XLIX collectively, with a proviso that the protein is not a contiguous sequence from an amino acid sequence of FIG. 2;
  • (VIII) a protein that comprises at least one peptide selected from the peptides set forth in Tables VIII-XXI; and at least one peptide selected from the peptides set forth in Tables XXII to XLIX, with a proviso that the protein is not a contiguous sequence from an amino acid sequence of FIG. 2;
  • (IX) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of FIG. 3A, 3B, and/or 3C in any whole number increment up to 273, 232, and/or 236 respectively that includes at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Hydrophilicity profile of FIG. 5;
  • (X) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of FIG. 3A, 3B, and/or 3C, in any whole number increment up to 273, 236, and/or 232 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value less than 0.5 in the Hydropathicity profile of FIG. 6;
  • (XI) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of FIG. 3A, 3B and/or 3C, in any whole number increment up to 273, 232 and/or 236 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Percent Accessible Residues profile of FIG. 7;
  • (XII) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acids of a protein of FIG. 3A, 3B and/or 3C, in any whole number increment up to 273, 232 and/or 236 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Average Flexibility profile of FIG. 8;
  • (XIII) a polypeptide comprising at least 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, amino acids of a protein of FIG. 3A, 3B, and/or 3C in any whole number increment up to 272, 232 and/or 236 respectively that includes at least at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 amino acid position(s) having a value greater than 0.5 in the Beta-turn profile of FIG. 9;
  • (XIV) a peptide that occurs at least twice in Tables VIII-XXI and XXII to XLIX, collectively;
  • (XV) a peptide that occurs at least three times in Tables VIII-XXI and XXII to XLIX, collectively;
  • (XVI) a peptide that occurs at least four times in Tables VIII-XXI and XXII to XLIX, collectively;
  • (XVII) a peptide that occurs at least five times in Tables VIII-XXI and XXII to XLIX, collectively;
  • (XVIII) a peptide that occurs at least once in Tables VIII-XXI, and at least once in tables XXII to XLIX;
  • (XIX) a peptide that occurs at least once in Tables VIII-XXI, and at least twice in tables XXII to XLIX;
  • (XX) a peptide that occurs at least twice in Tables VIII-XXI, and at least once in tables XXII to XLIX;
  • (XXI) a peptide that occurs at least twice in Tables VIII-XXI, and at least twice in tables XXII to XLIX;
  • (XXII) a peptide which comprises one two, three, four, or five of the following characteristics, or an oligonucleotide encoding such peptide:
  • i) a region of at least 5 amino acids of a particular peptide of FIG. 3, in any whole number increment up to the full length of that protein in FIG. 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Hydrophilicity profile of FIG. 5;
  • ii) a region of at least 5 amino acids of a particular peptide of FIG. 3, in any whole number increment up to the full length of that protein in FIG. 3, that includes an amino acid position having a value equal to or less than 0.5, 0.4, 0.3, 0.2, 0.1, or having a value equal to 0.0, in the Hydropathicity profile of FIG. 6;
  • iii) a region of at least 5 amino acids of a particular peptide of FIG. 3, in any whole number increment up to the full length of that protein in FIG. 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Percent Accessible Residues profile of FIG. 7;
  • iv) a region of at least 5 amino acids of a particular peptide of FIG. 3, in any whole number increment up to the full length of that protein in FIG. 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Average Flexibility profile of FIG. 8; or,
  • v) a region of at least 5 amino acids of a particular peptide of FIG. 3, in any whole number increment up to the full length of that protein in FIG. 3, that includes an amino acid position having a value equal to or greater than 0.5, 0.6, 0.7, 0.8, 0.9, or having a value equal to 1.0, in the Beta-turn profile of FIG. 9;
  • (XXIII) a composition comprising a peptide of (I)-(XXII) or an antibody or binding region thereof together with a pharmaceutical excipient and/or in a human unit dose form;
  • (XXIV) a method of using a peptide of (I)-(XXII), or an antibody or binding region thereof or a composition of (XXIII) in a method to modulate a cell expressing 58P1D12;
  • (XXV) a method of using a peptide of (I)-(XXII) or an antibody or binding region thereof or a composition of (XXIII) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing 58P1D12;
  • (XXVI) a method of using a peptide of (I)-(XXII) or an antibody or binding region thereof or a composition (XXIII) in a method to diagnose, prophylax, prognose, or treat an individual who bears a cell expressing 58P1D12, said cell from a cancer of a tissue listed in Table I;
  • (XXVII) a method of using a peptide of (I)-(XXII) or an antibody or binding region thereof or a composition of (XXIII) in a method to diagnose, prophylax, prognose, or treat a cancer;
  • (XXVIII) a method of using a peptide of (I)-(XXII) or an antibody or binding region thereof or a composition of (XXIII) in a method to diagnose, prophylax, prognose, or treat a a cancer of a tissue listed in Table I;
  • (XXIX) a method of using a a peptide of (I)-(XXII) or an antibody or binding region thereof or a composition and;
  • (XXIII) in a method to identify or characterize a modulator of a cell expressing 58P1D12.
  • As used herein, a range is understood to specifically disclose all whole unit positions thereof.
  • Typical embodiments of the invention disclosed herein include 58P1D12 polynucleotides that encode specific portions of 58P1D12 mRNA sequences (and those which are complementary to such sequences) such as those that encode the proteins and/or fragments thereof, for example:
  • (a) 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 105, 110, 115, 120, 125, 130, 135, 140, 145, 150, 155, 160, 165, 170, 175, 180, 185, 190, 195, 200, 225, 250, 255, 260, 265, 270 and 273 or more contiguous amino acids of 58P1D12 variant 1; the maximal lengths relevant for other variants are: variant 2, 232 amino acids; and variant 4, 236 amino acids.
  • In general, naturally occurring allelic variants of human 58P1D12 share a high degree of structural identity and homology (e.g., 90% or more homology). Typically, allelic variants of a 58P1D12 protein contain conservative amino acid substitutions within the 58P1D12 sequences described herein or contain a substitution of an amino acid from a corresponding position in a homologue of 58P1D12. One class of 58P1D12 allelic variants are proteins that share a high degree of homology with at least a small region of a particular 58P1D12 amino acid sequence, but further contain a radical departure from the sequence, such as a non-conservative substitution, truncation, insertion or frame shift. In comparisons of protein sequences, the terms, similarity, identity, and homology each have a distinct meaning as appreciated in the field of genetics. Moreover, orthology and paralogy can be important concepts describing the relationship of members of a given protein family in one organism to the members of the same family in other organisms.
  • Amino acid abbreviations are provided in Table II. Conservative amino acid substitutions can frequently be made in a protein without altering either the conformation or the function of the protein. Proteins of the invention can comprise 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 conservative substitutions. Such changes include substituting any of isoleucine (I), valine (V), and leucine (L) for any other of these hydrophobic amino acids; aspartic acid (D) for glutamic acid (E) and vice versa; glutamine (Q) for asparagine (N) and vice versa; and serine (S) for threonine (T) and vice versa. Other substitutions can also be considered conservative, depending on the environment of the particular amino acid and its role in the three-dimensional structure of the protein. For example, glycine (G) and alanine (A) can frequently be interchangeable, as can alanine (A) and valine (V). Methionine (M), which is relatively hydrophobic, can frequently be interchanged with leucine and isoleucine, and sometimes with valine. Lysine (K) and arginine (R) are frequently interchangeable in locations in which the significant feature of the amino acid residue is its charge and the differing pK's of these two amino acid residues are not significant. Still other changes can be considered “conservative” in particular environments (see, e.g. Table III herein; pages 13-15 “Biochemistry” 2nd ED. Lubert Stryer ed (Stanford University); Henikoff et al., PNAS 1992 Vol 89 10915-10919; Lei et al., J Biol Chem 1995 May 19; 270(20):11882-6).
  • Embodiments of the invention disclosed herein include a wide variety of art-accepted variants or analogs of 58P1D12 proteins such as polypeptides having amino acid insertions, deletions and substitutions. 58P1D12 variants can be made using methods known in the art such as site-directed mutagenesis, alanine scanning, and PCR mutagenesis. Site-directed mutagenesis (Carter et al., Nucl. Acids Res., 13:4331 (1986); Zoller et al., Nucl. Acids Res., 10:6487 (1987)), cassette mutagenesis (Wells et al., Gene, 34:315 (1985)), restriction selection mutagenesis (Wells et al., Philos. Trans. R. Soc. London SerA, 317:415 (1986)) or other known techniques can be performed on the cloned DNA to produce the 58P1D12 variant DNA.
  • Scanning amino acid analysis can also be employed to identify one or more amino acids along a contiguous sequence that is involved in a specific biological activity such as a protein-protein interaction. Among the preferred scanning amino acids are relatively small, neutral amino acids. Such amino acids include alanine, glycine, serine, and cysteine. Alanine is typically a preferred scanning amino acid among this group because it eliminates the side-chain beyond the beta-carbon and is less likely to alter the main-chain conformation of the variant. Alanine is also typically preferred because it is the most common amino acid. Further, it is frequently found in both buried and exposed positions (Creighton, The Proteins, (W.H. Freeman & Co., N.Y.); Chothia, J. Mol. Biol., 150:1 (1976)). If alanine substitution does not yield adequate amounts of variant, an isosteric amino acid can be used.
  • As defined herein, 58P1D12 variants, analogs or homologs, have the distinguishing attribute of having at least one epitope that is “cross reactive” with a 58P1D12 protein having an amino acid sequence of FIG. 3. As used in this sentence, “cross reactive” means that an antibody or T cell that specifically binds to a 58P1D12 variant also specifically binds to a 58P1D12 protein having an amino acid sequence set forth in FIG. 3. A polypeptide ceases to be a variant of a protein shown in FIG. 3, when it no longer contains any epitope capable of being recognized by an antibody or T cell that specifically binds to the starting 58P1D12 protein. Those skilled in the art understand that antibodies that recognize proteins bind to epitopes of varying size, and a grouping of the order of about four or five amino acids, contiguous or not, is regarded as a typical number of amino acids in a minimal epitope. See, e.g., Nair et al., J. Immunol 2000 165(12): 6949-6955; Hebbes et al., Mol Immunol (1989) 26(9):865-73; Schwartz et al., J Immunol (1985) 135(4):2598-608.
  • Other classes of 58P1D12-related protein variants share 70%, 75%, 80%, 85% or 90% or more similarity with an amino acid sequence of FIG. 3, or a fragment thereof. Another specific class of 58P1D12 protein variants or analogs comprises one or more of the 58P1D12 biological motifs described herein or presently known in the art. Thus, encompassed by the present invention are analogs of 58P1D12 fragments (nucleic or amino acid) that have altered functional (e.g. immunogenic) properties relative to the starting fragment. It is to be appreciated that motifs now or which become part of the art are to be applied to the nucleic or amino acid sequences of FIG. 2 or FIG. 3.
  • As discussed herein, embodiments of the claimed invention include polypeptides containing less than the full amino acid sequence of a 58P1D12 protein shown in FIG. 2 or FIG. 3. For example, representative embodiments of the invention comprise peptides/proteins having any 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15 or more contiguous amino acids of a 58P1D12 protein shown in FIG. 2 or FIG. 3.
  • Moreover, representative embodiments of the invention disclosed herein include polypeptides consisting of about amino acid 1 to about amino acid 10 of a 58P1D12 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 10 to about amino acid 20 of a 58P1D12 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 20 to about amino acid 30 of a 58P1D12 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 30 to about amino acid 40 of a 58P1D12 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 40 to about amino acid 50 of a 58P1D12 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 50 to about amino acid 60 of a 58P1D12 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 60 to about amino acid 70 of a 58P1D12 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 70 to about amino acid 80 of a 58P1D12 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 80 to about amino acid 90 of a 58P1D12 protein shown in FIG. 2 or FIG. 3, polypeptides consisting of about amino acid 90 to about amino acid 100 of a 58P1D12 protein shown in FIG. 2 or FIG. 3, etc. throughout the entirety of a 58P1D12 amino acid sequence. Moreover, polypeptides consisting of about amino acid 1 (or 20 or 30 or 40 etc.) to about amino acid 20, (or 130, or 140 or 150 etc.) of a 58P1D12 protein shown in FIG. 2 or FIG. 3 are embodiments of the invention. It is to be appreciated that the starting and stopping positions in this paragraph refer to the specified position as well as that position plus or minus 5 residues.
  • 58P1D12-related proteins are generated using standard peptide synthesis technology or using chemical cleavage methods well known in the art. Alternatively, recombinant methods can be used to generate nucleic acid molecules that encode a 58P1D12-related protein. In one embodiment, nucleic acid molecules provide a means to generate defined fragments of a 58P1D12 protein (or variants, homologs or analogs thereof).
  • III.A.) Motif-bearing Protein Embodiments
  • Additional illustrative embodiments of the invention disclosed herein include 58P1D12 polypeptides comprising the amino acid residues of one or more of the biological motifs contained within a 58P1D12 polypeptide sequence set forth in FIG. 2 or FIG. 3. Various motifs are known in the art, and a protein can be evaluated for the presence of such motifs by a number of publicly available Internet sites (see, e.g., URL addresses: pfam.wustl.edu/; searchlauncher.bcm.tmc.edu/seq-search/struc-predict.html; psort.ims.u-tokyo.ac.jp/; cbs.dtu.dk/; ebi.ac.uk/interpro/scan.html; expasy.ch/tools/scnpsitl.html; Epimatrix™ and Epimer™, Brown University, brown.edu/Research/TB-HIV_Lab/epimatrix/epimatrix.html; and BIMAS, bimas.dcrt.nih.gov/.).
  • Motif bearing subsequences of all 58P1D12 variant proteins are set forth and identified in Tables VIII-XXI and XXII-XLIX.
  • Table V sets forth several frequently occurring motifs based on pfam searches (see URL address pfam.wustl.edu/). The columns of Table V list (1) motif name abbreviation, (2) percent identity found amongst the different member of the motif family, (3) motif name or description and (4) most common function; location information is included if the motif is relevant for location.
  • Polypeptides comprising one or more of the 58P1D12 motifs discussed above are useful in elucidating the specific characteristics of a malignant phenotype in view of the observation that the 58P1D12 motifs discussed above are associated with growth dysregulation and because 58P1D12 is overexpressed in certain cancers (See, e.g., Table I). Casein kinase II, cAMP and camp-dependent protein kinase, and Protein Kinase C, for example, are enzymes known to be associated with the development of the malignant phenotype (see e.g. Chen et al., Lab Invest., 78(2): 165-174 (1998); Gaiddon et al., Endocrinology 136(10): 4331-4338 (1995); Hall et al., Nucleic Acids Research 24(6): 1119-1126 (1996); Peterziel et al., Oncogene 18(46): 6322-6329 (1999) and O'Brian, Oncol. Rep. 5(2): 305-309 (1998)). Moreover, both glycosylation and myristoylation are protein modifications also associated with cancer and cancer progression (see e.g. Dennis et al., Biochem. Biophys. Acta 1473(1):21-34 (1999); Raju et al., Exp. Cell Res. 235(1): 145-154 (1997)). Amidation is another protein modification also associated with cancer and cancer progression (see e.g. Treston et al., J. Natl. Cancer Inst. Monogr. (13): 169-175 (1992)).
  • In another embodiment, proteins of the invention comprise one or more of the immunoreactive epitopes identified in accordance with art-accepted methods, such as the peptides set forth in Tables VIII-XXI and XXII-XLIX. CTL epitopes can be determined using specific algorithms to identify peptides within a 58P1D12 protein that are capable of optimally binding to specified HLA alleles (e.g., Table IV; Epimatrix™ and Epimer™, Brown University, URL brown.edu/Research/TB-HIV_Lab/epimatrix/epimatrix.html; and BIMAS, URL bimas.dcrt nih gov/.) Moreover, processes for identifying peptides that have sufficient binding affinity for HLA molecules and which are correlated with being immunogenic epitopes, are well known in the art, and are carried out without undue experimentation. In addition, processes for identifying peptides that are immunogenic epitopes, are well known in the art, and are carried out without undue experimentation either in vitro or in vivo.
  • Also known in the art are principles for creating analogs of such epitopes in order to modulate immunogenicity. For example, one begins with an epitope that bears a CTL or HTL motif (see, e.g., the HLA Class I and HLA Class II motifs/supermotifs of Table IV). The epitope is analoged by substituting out an amino acid at one of the specified positions, and replacing it with another amino acid specified for that position. For example, on the basis of residues defined in Table IV, one can substitute out a deleterious residue in favor of any other residue, such as a preferred residue; substitute a less-preferred residue with a preferred residue; or substitute an originally-occurring preferred residue with another preferred residue. Substitutions can occur at primary anchor positions or at other positions in a peptide; see, e.g., Table IV.
  • A variety of references reflect the art regarding the identification and generation of epitopes in a protein of interest as well as analogs thereof. See, for example, WO 97/33602 to Chesnut et al.; Sette, Immunogenetics 1999 50(3-4): 201-212; Sette et al., J. Immunol. 2001 166(2): 1389-1397; Sidney et al., Hum. Immunol. 1997 58(1): 12-20; Kondo et al., Immunogenetics 1997 45(4): 249-258; Sidney et al., J. Immunol. 1996 157(8): 3480-90; and Falk et al., Nature 351: 290-6 (1991); Hunt et al., Science 255:1261-3 (1992); Parker et al., J. Immunol. 149:3580-7 (1992); Parker et al., J. Immunol. 152:163-75 (1994)); Kast et al., 1994 152(8): 3904-12; Borras-Cuesta et al., Hum. Immunol. 2000 61(3): 266-278; Alexander et al., J. Immunol. 2000 164(3); 164(3): 1625-1633; Alexander et al., PMID: 7895164, UI: 95202582; O'Sullivan et al., J. Immunol. 1991 147(8): 2663-2669; Alexander et al., Immunity 1994 1(9): 751-761 and Alexander et al., Immunol. Res. 1998 18(2): 79-92.
  • Related embodiments of the invention include polypeptides comprising combinations of the different motifs set forth in Table VI, and/or, one or more of the predicted CTL epitopes of Tables VIII-XXI and XXII-XLIX, and/or, one or more of the predicted HTL epitopes of Tables XLVI-XLIX, and/or, one or more of the T cell binding motifs known in the art. Preferred embodiments contain no insertions, deletions or substitutions either within the motifs or within the intervening sequences of the polypeptides. In addition, embodiments which include a number of either N-terminal and/or C-terminal amino acid residues on either side of these motifs may be desirable (to, for example, include a greater portion of the polypeptide architecture in which the motif is located). Typically, the number of N-terminal and/or C-terminal amino acid residues on either side of a motif is between about 1 to about 100 amino acid residues, preferably 5 to about 50 amino acid residues.
  • 58P1D12-related proteins are embodied in many forms, preferably in isolated form. A purified 58P1D12 protein molecule will be substantially free of other proteins or molecules that impair the binding of 58P1D12 to antibody, T cell or other ligand. The nature and degree of isolation and purification will depend on the intended use. Embodiments of a 58P1D12-related proteins include purified 58P1D12-related proteins and functional, soluble 58P1D12-related proteins. In one embodiment, a functional, soluble 58P1D12 protein or fragment thereof retains the ability to be bound by antibody, T cell or other ligand.
  • The invention also provides 58P1D12 proteins comprising biologically active fragments of a 58P1D12 amino acid sequence shown in FIG. 2 or FIG. 3. Such proteins exhibit properties of the starting 58P1D12 protein, such as the ability to elicit the generation of antibodies that specifically bind an epitope associated with the starting 58P1D12 protein; to be bound by such antibodies; to elicit the activation of HTL or CTL; and/or, to be recognized by HTL or CTL that also specifically bind to the starting protein.
  • 58P1D12-related polypeptides that contain particularly interesting structures can be predicted and/or identified using various analytical techniques well known in the art, including, for example, the methods of Chou-Fasman, Garnier-Robson, Kyte-Doolittle, Eisenberg, Karplus-Schultz or Jameson-Wolf analysis, or based on immunogenicity. Fragments that contain such structures are particularly useful in generating subunit-specific anti-58P1D12 antibodies or T cells or in identifying cellular factors that bind to 58P1D12. For example, hydrophilicity profiles can be generated, and immunogenic peptide fragments identified, using the method of Hopp, T. P. and Woods, K. R., 1981, Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828. Hydropathicity profiles can be generated, and immunogenic peptide fragments identified, using the method of Kyte, J. and Doolittle, R. F., 1982, J. Mol. Biol. 157:105-132. Percent (%) Accessible Residues profiles can be generated, and immunogenic peptide fragments identified, using the method of Janin J., 1979, Nature 277:491-492. Average Flexibility profiles can be generated, and immunogenic peptide fragments identified, using the method of Bhaskaran R., Ponnuswamy P.K., 1988, Int. J. Pept. Protein Res. 32:242-255. Beta-turn profiles can be generated, and immunogenic peptide fragments identified, using the method of Deleage, G., Roux B., 1987, Protein Engineering 1:289-294.
  • CTL epitopes can be determined using specific algorithms to identify peptides within a 58P1D12 protein that are capable of optimally binding to specified HLA alleles (e.g., by using the SYFPEITHI site at World Wide Web URL syfpeithi.bmi-heidelberg.com/; the listings in Table IV(A)-(E); Epimatrix™ and Epimer™, Brown University, URL (brown.edu/Research/TB-HIV_Lab/epimatrix/epimatrix.html); and BIMAS, URL bimas.dcrt.nih gov/). Illustrating this, peptide epitopes from 58P1D12 that are presented in the context of human MHC Class I molecules, e.g., HLA-A1, A2, A3, A11, A24, B7 and B35 were predicted (see, e.g., Tables VIII-XXI, XXII-XLIX). Specifically, the complete amino acid sequence of the 58P1D12 protein and relevant portions of other variants, i.e., for HLA Class I predictions 9 flanking residues on either side of a point mutation or exon junction, and for HLA Class II predictions 14 flanking residues on either side of a point mutation or exon junction corresponding to that variant, were entered into the HLA Peptide Motif Search algorithm found in the Bioinformatics and Molecular Analysis Section (BIMAS) web site listed above; in addition to the site SYFPEITHI, at URL syfpeithi.bmi-heidelberg.com/.
  • The HLA peptide motif search algorithm was developed by Dr. Ken Parker based on binding of specific peptide sequences in the groove of HLA Class I molecules, in particular HLA-A2 (see, e.g., Falk et al., Nature 351: 290-6 (1991); Hunt et al., Science 255:1261-3 (1992); Parker et al., J. Immunol. 149:3580-7 (1992); Parker et al., J. Immunol. 152:163-75 (1994)). This algorithm allows location and ranking of 8-mer, 9-mer, and 10-mer peptides from a complete protein sequence for predicted binding to HLA-A2 as well as numerous other HLA Class I molecules. Many HLA class I binding peptides are 8-, 9-, 10 or 11-mers. For example, for Class I HLA-A2, the epitopes preferably contain a leucine (L) or methionine (M) at position 2 and a valine (V) or leucine (L) at the C-terminus (see, e.g., Parker et al., J. Immunol. 149:3580-7 (1992)). Selected results of 58P1D12 predicted binding peptides are shown in Tables VIII-XXI and XXII-XLIX herein. In Tables VIII-XXI and XXII-XLVII, selected candidates, 9-mers and 10-mers, for each family member are shown along with their location, the amino acid sequence of each specific peptide, and an estimated binding score. In Tables XLVI-XLIX, selected candidates, 15-mers, for each family member are shown along with their location, the amino acid sequence of each specific peptide, and an estimated binding score. The binding score corresponds to the estimated half time of dissociation of complexes containing the peptide at 37° C. at pH 6.5. Peptides with the highest binding score are predicted to be the most tightly bound to HLA Class I on the cell surface for the greatest period of time and thus represent the best immunogenic targets for T-cell recognition.
  • Actual binding of peptides to an HLA allele can be evaluated by stabilization of HLA expression on the antigen-processing defective cell line T2 (see, e.g., Xue et al., Prostate 30:73-8 (1997) and Peshwa et al., Prostate 36:129-38 (1998)) Immunogenicity of specific peptides can be evaluated in vitro by stimulation of CD8+ cytotoxic T lymphocytes (CTL) in the presence of antigen presenting cells such as dendritic cells.
  • It is to be appreciated that every epitope predicted by the BIMAS site, Epimer™ and Epimatrix™ sites, or specified by the HLA class I or class II motifs available in the art or which become part of the art such as set forth in Table IV (or determined using World Wide Web site URL syfpeithi.bmi-heidelberg.com/, or BIMAS, bimas.dcrt.nih.gov/) are to be “applied” to a 58P1D12 protein in accordance with the invention. As used in this context “applied” means that a 58P1D12 protein is evaluated, e.g., visually or by computer-based patterns finding methods, as appreciated by those of skill in the relevant art. Every subsequence of a 58P1D12 protein of 8, 9, 10, or 11 amino acid residues that bears an HLA Class I motif, or a subsequence of 9 or more amino acid residues that bear an HLA Class II motif are within the scope of the invention.
  • III.B.) Expression of 58P1D12-Related Proteins
  • In an embodiment described in the examples that follow, 58P1D12 can be conveniently expressed in cells (such as 293T cells) transfected with a commercially available expression vector such as a CMV-driven expression vector encoding 58P1D12 with a C-terminal 6× His and MYC tag (pcDNA3.1/mycHIS, Invitrogen or Tag5, GenHunter Corporation, Nashville Tenn.). The Tag5 vector provides an IgGK secretion signal that can be used to facilitate the production of a secreted 58P1D12 protein in transfected cells. The secreted HIS-tagged 58P1D12 in the culture media can be purified, e.g., using a nickel column using standard techniques.
  • III.C.) Modifications of 58P1D12-Related Proteins
  • Modifications of 58P1D12-related proteins such as covalent modifications are included within the scope of this invention. One type of covalent modification includes reacting targeted amino acid residues of a 58P1D12 polypeptide with an organic derivatizing agent that is capable of reacting with selected side chains or the N- or C-terminal residues of a 58P1D12 protein. Another type of covalent modification of a 58P1D12 polypeptide included within the scope of this invention comprises altering the native glycosylation pattern of a protein of the invention. Another type of covalent modification of 58P1D12 comprises linking a 58P1D12 polypeptide to one of a variety of nonproteinaceous polymers, e.g., polyethylene glycol (PEG), polypropylene glycol, or polyoxyalkylenes, in the manner set forth in U.S. Pat. Nos. 4,640,835; 4,496,689; 4,301,144; 4,670,417; 4,791,192 or 4,179,337.
  • The 58P1D12-related proteins of the present invention can also be modified to form a chimeric molecule comprising 58P1D12 fused to another, heterologous polypeptide or amino acid sequence. Such a chimeric molecule can be synthesized chemically or recombinantly. A chimeric molecule can have a protein of the invention fused to another tumor-associated antigen or fragment thereof. Alternatively, a protein in accordance with the invention can comprise a fusion of fragments of a 58P1D12 sequence (amino or nucleic acid) such that a molecule is created that is not, through its length, directly homologous to the amino or nucleic acid sequences shown in FIG. 2 or FIG. 3. Such a chimeric molecule can comprise multiples of the same subsequence of 58P1D12. A chimeric molecule can comprise a fusion of a 58P1D12-related protein with a polyhistidine epitope tag, which provides an epitope to which immobilized nickel can selectively bind, with cytokines or with growth factors. The epitope tag is generally placed at the amino- or carboxyl-terminus of a 58P1D12 protein. In an alternative embodiment, the chimeric molecule can comprise a fusion of a 58P1D12-related protein with an immunoglobulin or a particular region of an immunoglobulin. For a bivalent form of the chimeric molecule (also referred to as an “immunoadhesin”), such a fusion could be to the Fc region of an IgG molecule. The Ig fusions preferably include the substitution of a soluble (transmembrane domain deleted or inactivated) form of a 58P1D12 polypeptide in place of at least one variable region within an Ig molecule. In a preferred embodiment, the immunoglobulin fusion includes the hinge, CH2 and CH3, or the hinge, CH1, CH2 and CH3 regions of an IgGI molecule. For the production of immunoglobulin fusions see, e.g., U.S. Pat. No. 5,428,130 issued Jun. 27, 1995.
  • III.D.) Uses of 58P1D12-Related Proteins
  • The proteins of the invention have a number of different specific uses. As 58P1D12 is highly expressed in prostate and other cancers, 58P1D12-related proteins are used in methods that assess the status of 58P1D12 gene products in normal versus cancerous tissues, thereby elucidating the malignant phenotype. Typically, polypeptides from specific regions of a 58P1D12 protein are used to assess the presence of perturbations (such as deletions, insertions, point mutations etc.) in those regions (such as regions containing one or more motifs). Exemplary assays utilize antibodies or T cells targeting 58P1D12-related proteins comprising the amino acid residues of one or more of the biological motifs contained within a 58P1D12 polypeptide sequence in order to evaluate the characteristics of this region in normal versus cancerous tissues or to elicit an immune response to the epitope. Alternatively, 58P1D12-related proteins that contain the amino acid residues of one or more of the biological motifs in a 58P1D12 protein are used to screen for factors that interact with that region of 58P1D12.
  • 58P1D12 protein fragments/subsequences are particularly useful in generating and characterizing domain-specific antibodies (e.g., antibodies recognizing an extracellular or intracellular epitope of a 58P1D12 protein), for identifying agents or cellular factors that bind to 58P1D12 or a particular structural domain thereof, and in various therapeutic and diagnostic contexts, including but not limited to diagnostic assays, cancer vaccines and methods of preparing such vaccines.
  • Proteins encoded by the 58P1D12 genes, or by analogs, homologs or fragments thereof, have a variety of uses, including but not limited to generating antibodies and in methods for identifying ligands and other agents and cellular constituents that bind to a 58P1D12 gene product. Antibodies raised against a 58P1D12 protein or fragment thereof are useful in diagnostic and prognostic assays, and imaging methodologies in the management of human cancers characterized by expression of 58P1D12 protein, such as those listed in Table I. Such antibodies can be expressed intracellularly and used in methods of treating patients with such cancers. 58P1D12-related nucleic acids or proteins are also used in generating HTL or CTL responses.
  • Various immunological assays useful for the detection of 58P1D12 proteins are used, including but not limited to various types of radioimmunoassays, enzyme-linked immunosorbent assays (ELISA), enzyme-linked immunofluorescent assays (ELIFA), immunocytochemical methods, and the like. Antibodies can be labeled and used as immunological imaging reagents capable of detecting 58P1D12-expressing cells (e.g., in radioscintigraphic imaging methods). 58P1D12 proteins are also particularly useful in generating cancer vaccines, as further described herein.
  • IV.) 58P1D12 ANTIBODIES
  • Another aspect of the invention provides antibodies that bind to 58P1D12-related proteins. Preferred antibodies specifically bind to a 58P1D12-related protein and do not bind (or bind weakly) to peptides or proteins that are not 58P1D12-related proteins under physiological conditions. In this context, examples of physiological conditions include: 1) phosphate buffered saline; 2) Tris-buffered saline containing 25 mM Tris and 150 mM NaCl; or normal saline (0.9% NaCl); 4) animal serum such as human serum; or, 5) a combination of any of 1) through 4); these reactions preferably taking place at pH 7.5, alternatively in a range of pH 7.0 to 8.0, or alternatively in a range of pH 6.5 to 8.5; also, these reactions taking place at a temperature between 4° C. to 37° C. For example, antibodies that bind 58P1D12 can bind 58P1D12-related proteins such as the homologs or analogs thereof.
  • 58P1D12 antibodies of the invention are particularly useful in cancer (see, e.g., Table I) diagnostic and prognostic assays, and imaging methodologies. Similarly, such antibodies are useful in the treatment, diagnosis, and/or prognosis of other cancers, to the extent 58P1D12 is also expressed or overexpressed in these other cancers. Moreover, intracellularly expressed antibodies (e.g., single chain antibodies) are therapeutically useful in treating cancers in which the expression of 58P1D12 is involved, such as advanced or metastatic prostate cancers.
  • The invention also provides various immunological assays useful for the detection and quantification of 58P1D12 and mutant 58P1D12-related proteins. Such assays can comprise one or more 58P1D12 antibodies capable of recognizing and binding a 58P1D12-related protein, as appropriate. These assays are performed within various immunological assay formats well known in the art, including but not limited to various types of radioimmunoassays, enzyme-linked immunosorbent assays (ELISA), enzyme-linked immunofluorescent assays (ELIFA), and the like.
  • Immunological non-antibody assays of the invention also comprise T cell immunogenicity assays (inhibitory or stimulatory) as well as major histocompatibility complex (MHC) binding assays.
  • In addition, immunological imaging methods capable of detecting prostate cancer and other cancers expressing 58P1D12 are also provided by the invention, including but not limited to radioscintigraphic imaging methods using labeled 58P1D12 antibodies. Such assays are clinically useful in the detection, monitoring, and prognosis of 58P1D12 expressing cancers such as prostate cancer.
  • 58P1D12 antibodies are also used in methods for purifying a 58P1D12-related protein and for isolating 58P1D12 homologues and related molecules. For example, a method of purifying a 58P1D12-related protein comprises incubating a 58P1D12 antibody, which has been coupled to a solid matrix, with a lysate or other solution containing a 58P1D12-related protein under conditions that permit the 58P1D12 antibody to bind to the 58P1D12-related protein; washing the solid matrix to eliminate impurities; and eluting the 58P1D12-related protein from the coupled antibody. Other uses of 58P1D12 antibodies in accordance with the invention include generating anti-idiotypic antibodies that mimic a 58P1D12 protein.
  • Various methods for the preparation of antibodies are well known in the art. For example, antibodies can be prepared by immunizing a suitable mammalian host using a 58P1D12-related protein, peptide, or fragment, in isolated or immunoconjugated form (Antibodies: A Laboratory Manual, CSH Press, Eds., Harlow, and Lane (1988); Harlow, Antibodies, Cold Spring Harbor Press, NY (1989)). In addition, fusion proteins of 58P1D12 can also be used, such as a 58P1D12 GST-fusion protein. In a particular embodiment, a GST fusion protein comprising all or most of the amino acid sequence of FIG. 2 or FIG. 3 is produced, then used as an immunogen to generate appropriate antibodies. In another embodiment, a 58P1D12-related protein is synthesized and used as an immunogen.
  • In addition, naked DNA immunization techniques known in the art are used (with or without purified 58P1D12-related protein or 58P1D12 expressing cells) to generate an immune response to the encoded immunogen (for review, see Donnelly et al., 1997, Ann. Rev. Immunol. 15: 617-648).
  • The amino acid sequence of a 58P1D12 protein as shown in FIG. 2 or FIG. 3 can be analyzed to select specific regions of the 58P1D12 protein for generating antibodies. For example, hydrophobicity and hydrophilicity analyses of a 58P1D12 amino acid sequence are used to identify hydrophilic regions in the 58P1D12 structure. Regions of a 58P1D12 protein that show immunogenic structure, as well as other regions and domains, can readily be identified using various other methods known in the art, such as Chou-Fasman, Garnier-Robson, Kyte-Doolittle, Eisenberg, Karplus-Schultz or Jameson-Wolf analysis. Hydrophilicity profiles can be generated using the method of Hopp, T. P. and Woods, K. R., 1981, Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828. Hydropathicity profiles can be generated using the method of Kyte, J. and Doolittle, R.F., 1982, J. Mol. Biol. 157:105-132. Percent (%) Accessible Residues profiles can be generated using the method of Janin J., 1979, Nature 277:491-492. Average Flexibility profiles can be generated using the method of Bhaskaran R., Ponnuswamy P. K., 1988, Int. J. Pept. Protein Res. 32:242-255. Beta-turn profiles can be generated using the method of Deleage, G., Roux B., 1987, Protein Engineering 1:289-294. Thus, each region identified by any of these programs or methods is within the scope of the present invention. Methods for the generation of 58P1D12 antibodies are further illustrated by way of the examples provided herein. Methods for preparing a protein or polypeptide for use as an immunogen are well known in the art. Also well known in the art are methods for preparing immunogenic conjugates of a protein with a carrier, such as BSA, KLH or other carrier protein. In some circumstances, direct conjugation using, for example, carbodiimide reagents are used; in other instances linking reagents such as those supplied by Pierce Chemical Co., Rockford, Ill., are effective. Administration of a 58P1D12 immunogen is often conducted by injection over a suitable time period and with use of a suitable adjuvant, as is understood in the art. During the immunization schedule, titers of antibodies can be taken to determine adequacy of antibody formation.
  • 58P1D12 monoclonal antibodies can be produced by various means well known in the art. For example, immortalized cell lines that secrete a desired monoclonal antibody are prepared using the standard hybridoma technology of Kohler and Milstein or modifications that immortalize antibody-producing B cells, as is generally known. Immortalized cell lines that secrete the desired antibodies are screened by immunoassay in which the antigen is a 58P1D12-related protein. When the appropriate immortalized cell culture is identified, the cells can be expanded and antibodies produced either from in vitro cultures or from ascites fluid.
  • The antibodies or fragments of the invention can also be produced, by recombinant means. Regions that bind specifically to the desired regions of a 58P1D12 protein can also be produced in the context of chimeric or complementarity-determining region (CDR) grafted antibodies of multiple species origin. Humanized or human 58P1D12 antibodies can also be produced, and are preferred for use in therapeutic contexts. Methods for humanizing murine and other non-human antibodies, by substituting one or more of the non-human antibody CDRs for corresponding human antibody sequences, are well known (see for example, Jones et al., 1986, Nature 321: 522-525; Riechmann et al., 1988, Nature 332: 323-327; Verhoeyen et al., 1988, Science 239: 1534-1536). See also, Carter et al., 1993, Proc. Natl. Acad. Sci. USA 89: 4285 and Sims et al., 1993, J. Immunol. 151: 2296.
  • Methods for producing fully human monoclonal antibodies include phage display and transgenic methods (for review, see Vaughan et al., 1998, Nature Biotechnology 16: 535-539). Fully human 58P1D12 monoclonal antibodies can be generated using cloning technologies employing large human Ig gene combinatorial libraries (i.e., phage display) (Griffiths and Hoogenboom, Building an in vitro immune system: human antibodies from phage display libraries. In: Protein Engineering of Antibody Molecules for Prophylactic and Therapeutic Applications in Man, Clark, M. (Ed.), Nottingham Academic, pp 45-64 (1993); Burton and Barbas, Human Antibodies from combinatorial libraries. Id., pp 65-82). Fully human 58P1D12 monoclonal antibodies can also be produced using transgenic mice engineered to contain human immunoglobulin gene loci as described in PCT Patent Application WO98/24893, Kucherlapati and Jakobovits et al., published Dec. 3, 1997 (see also, Jakobovits, 1998, Exp. Opin. Invest. Drugs 7(4): 607-614; U.S. Pat. Nos. 6,162,963 issued 19 Dec. 2000; 6,150,584 issued 12 Nov. 2000; and, 6,114,598 issued 5 Sep. 2000). This method avoids the in vitro manipulation required with phage display technology and efficiently produces high affinity authentic human antibodies.
  • Reactivity of 58P1D12 antibodies with a 58P1D12-related protein can be established by a number of well known means, including Western blot, immunoprecipitation, ELISA, and FACS analyses using, as appropriate, 58P1D12-related proteins, 58P1D12-expressing cells or extracts thereof. A 58P1D12 antibody or fragment thereof can be labeled with a detectable marker or conjugated to a second molecule. Suitable detectable markers include, but are not limited to, a radioisotope, a fluorescent compound, a bioluminescent compound, chemiluminescent compound, a metal chelator or an enzyme. Further, bi-specific antibodies specific for two or more 58P1D12 epitopes are generated using methods generally known in the art. Homodimeric antibodies can also be generated by cross-linking techniques known in the art (e.g., Wolff et al., Cancer Res. 53: 2560-2565).
  • V.) 58P1D12 CELLULAR IMMUNE RESPONSES
  • The mechanism by which T cells recognize antigens has been delineated. Efficacious peptide epitope vaccine compositions of the invention induce a therapeutic or prophylactic immune responses in very broad segments of the world-wide population. For an understanding of the value and efficacy of compositions of the invention that induce cellular immune responses, a brief review of immunology-related technology is provided.
  • A complex of an HLA molecule and a peptidic antigen acts as the ligand recognized by HLA-restricted T cells (Buus, S. et al., Cell 47:1071, 1986; Babbitt, B. P. et al., Nature 317:359, 1985; Townsend, A. and Bodmer, H., Annu. Rev. Immunol. 7:601, 1989; Germain, R. N., Annu. Rev. Immunol. 11:403, 1993). Through the study of single amino acid substituted antigen analogs and the sequencing of endogenously bound, naturally processed peptides, critical residues that correspond to motifs required for specific binding to HLA antigen molecules have been identified and are set forth in Table IV (see also, e.g., Southwood, et al., J. Immunol. 160:3363, 1998; Rammensee, et al., Immunogenetics 41:178, 1995; Rammensee et al., SYFPEITHI, access via World Wide Web at URL (134.2.96.221/scripts.hlaserver.dll/home.htm); Sette, A. and Sidney, J. Curr. Opin. Immunol. 10:478, 1998; Engelhard, V. H., Curr. Opin. Immunol. 6:13, 1994; Sette, A. and Grey, H. M., Curr. Opin. Immunol. 4:79, 1992; Sinigaglia, F. and Hammer, J. Curr. Biol. 6:52, 1994; Ruppert et al., Cell 74:929-937, 1993; Kondo et al., J. Immunol. 155:4307-4312, 1995; Sidney et al., J. Immunol. 157:3480-3490, 1996; Sidney et al., Human Immunol. 45:79-93, 1996; Sette, A. and Sidney, J. Immunogenetics 1999 November; 50(3-4):201-12, Review).
  • Furthermore, x-ray crystallographic analyses of HLA-peptide complexes have revealed pockets within the peptide binding cleft/groove of HLA molecules which accommodate, in an allele-specific mode, residues borne by peptide ligands; these residues in turn determine the HLA binding capacity of the peptides in which they are present. (See, e.g., Madden, D. R. Annu. Rev. Immunol. 13:587, 1995; Smith, et al., Immunity 4:203, 1996; Fremont et al., Immunity 8:305, 1998; Stern et al., Structure 2:245, 1994; Jones, E. Y. Curr. Opin. Immunol. 9:75, 1997; Brown, J. H. et al., Nature 364:33, 1993; Guo, H. C. et al., Proc. Natl. Acad. Sci. USA 90:8053, 1993; Guo, H. C. et al., Nature 360:364, 1992; Silver, M. L. et al., Nature 360:367, 1992; Matsumura, M. et al., Science 257:927, 1992; Madden et al., Cell 70:1035, 1992; Fremont, D. H. et al., Science 257:919, 1992; Saper, M. A., Bjorkman, P. J. and Wiley, D. C., J. Mol. Biol. 219:277, 1991.)
  • Accordingly, the definition of class I and class II allele-specific HLA binding motifs, or class I or class II supermotifs allows identification of regions within a protein that are correlated with binding to particular HLA antigen(s).
  • Thus, by a process of HLA motif identification, candidates for epitope-based vaccines have been identified; such candidates can be further evaluated by HLA-peptide binding assays to determine binding affinity and/or the time period of association of the epitope and its corresponding HLA molecule. Additional confirmatory work can be performed to select, amongst these vaccine candidates, epitopes with preferred characteristics in terms of population coverage, and/or immunogenicity.
  • Various strategies can be utilized to evaluate cellular immunogenicity, including:
  • 1) Evaluation of primary T cell cultures from normal individuals (see, e.g., Wentworth, P. A. et al., Mol. Immunol. 32:603, 1995; Celis, E. et al., Proc. Natl. Acad. Sci. USA 91:2105, 1994; Tsai, V. et al., J. Immunol. 158:1796, 1997; Kawashima, I. et al., Human Immunol. 59:1, 1998). This procedure involves the stimulation of peripheral blood lymphocytes (PBL) from normal subjects with a test peptide in the presence of antigen presenting cells in vitro over a period of several weeks. T cells specific for the peptide become activated during this time and are detected using, e.g., a lymphokine- or 51Cr-release assay involving peptide sensitized target cells.
  • 2) Immunization of HLA transgenic mice (see, e.g., Wentworth, P. A. et al., J. Immunol. 26:97, 1996; Wentworth, P. A. et al., Int. Immunol. 8:651, 1996; Alexander, J. et al., J. Immunol. 159:4753, 1997). For example, in such methods peptides in incomplete Freund's adjuvant are administered subcutaneously to HLA transgenic mice. Several weeks following immunization, splenocytes are removed and cultured in vitro in the presence of test peptide for approximately one week. Peptide-specific T cells are detected using, e.g., a 51Cr-release assay involving peptide sensitized target cells and target cells expressing endogenously generated antigen.
  • 3) Demonstration of recall T cell responses from immune individuals who have been either effectively vaccinated and/or from chronically ill patients (see, e.g., Rehermann, B. et al., J. Exp. Med. 181:1047, 1995; Doolan, D. L. et al., Immunity 7:97, 1997; Bertoni, R. et al., J. Clin. Invest. 100:503, 1997; Threlkeld, S. C. et al., J. Immunol. 159:1648, 1997; Diepolder, H. M. et al., J. Virol. 71:6011, 1997). Accordingly, recall responses are detected by culturing PBL from subjects that have been exposed to the antigen due to disease and thus have generated an immune response “naturally”, or from patients who were vaccinated against the antigen. PBL from subjects are cultured in vitro for 1-2 weeks in the presence of test peptide plus antigen presenting cells (APC) to allow activation of “memory” T cells, as compared to “naive” T cells. At the end of the culture period, T cell activity is detected using assays including 51Cr release involving peptide-sensitized targets, T cell proliferation, or lymphokine release.
  • VI.) 58P1D12 TRANSGENIC ANIMALS
  • Nucleic acids that encode a 58P1D12-related protein can also be used to generate either transgenic animals or “knock out” animals that, in turn, are useful in the development and screening of therapeutically useful reagents. In accordance with established techniques, cDNA encoding 58P1D12 can be used to clone genomic DNA that encodes 58P1D12. The cloned genomic sequences can then be used to generate transgenic animals containing cells that express DNA that encode 58P1D12. Methods for generating transgenic animals, particularly animals such as mice or rats, have become conventional in the art and are described, for example, in U.S. Pat. Nos. 4,736,866 issued 12 Apr. 1988, and 4,870,009 issued 26 Sep. 1989. Typically, particular cells would be targeted for 58P1D12 transgene incorporation with tissue-specific enhancers.
  • Transgenic animals that include a copy of a transgene encoding 58P1D12 can be used to examine the effect of increased expression of DNA that encodes 58P1D12. Such animals can be used as tester animals for reagents thought to confer protection from, for example, pathological conditions associated with its overexpression. In accordance with this aspect of the invention, an animal is treated with a reagent and a reduced incidence of a pathological condition, compared to untreated animals that bear the transgene, would indicate a potential therapeutic intervention for the pathological condition.
  • Alternatively, non-human homologues of 58P1D12 can be used to construct a 58P1D12 “knock out” animal that has a defective or altered gene encoding 58P1D12 as a result of homologous recombination between the endogenous gene encoding 58P1D12 and altered genomic DNA encoding 58P1D12 introduced into an embryonic cell of the animal. For example, cDNA that encodes 58P1D12 can be used to clone genomic DNA encoding 58P1D12 in accordance with established techniques. A portion of the genomic DNA encoding 58P1D12 can be deleted or replaced with another gene, such as a gene encoding a selectable marker that can be used to monitor integration. Typically, several kilobases of unaltered flanking DNA (both at the 5′ and 3′ ends) are included in the vector (see, e.g., Thomas and Capecchi, Cell, 51:503 (1987) for a description of homologous recombination vectors). The vector is introduced into an embryonic stem cell line (e.g., by electroporation) and cells in which the introduced DNA has homologously recombined with the endogenous DNA are selected (see, e.g., Li et al., Cell, 69:915 (1992)). The selected cells are then injected into a blastocyst of an animal (e.g., a mouse or rat) to form aggregation chimeras (see, e.g., Bradley, in Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, E. J. Robertson, ed. (IRL, Oxford, 1987), pp. 113-152). A chimeric embryo can then be implanted into a suitable pseudopregnant female foster animal, and the embryo brought to term to create a “knock out” animal Progeny harboring the homologously recombined DNA in their germ cells can be identified by standard techniques and used to breed animals in which all cells of the animal contain the homologously recombined DNA. Knock out animals can be characterized, for example, for their ability to defend against certain pathological conditions or for their development of pathological conditions due to absence of a 58P1D12 polypeptide.
  • VII.) METHODS FOR THE DETECTION OF 58P1D12
  • Another aspect of the present invention relates to methods for detecting 58P1D12 polynucleotides and 58P1D12-related proteins, as well as methods for identifying a cell that expresses 58P1D12. The expression profile of 58P1D12 makes it a diagnostic marker for metastasized disease. Accordingly, the status of 58P1D12 gene products provides information useful for predicting a variety of factors including susceptibility to advanced stage disease, rate of progression, and/or tumor aggressiveness. As discussed in detail herein, the status of 58P1D12 gene products in patient samples can be analyzed by a variety protocols that are well known in the art including immunohistochemical analysis, the variety of Northern blotting techniques including in situ hybridization, RT-PCR analysis (for example on laser capture micro-dissected samples), Western blot analysis and tissue array analysis.
  • More particularly, the invention provides assays for the detection of 58P1D12 polynucleotides in a biological sample, such as serum, bone, prostate, and other tissues, urine, semen, cell preparations, and the like. Detectable 58P1D12 polynucleotides include, for example, a 58P1D12 gene or fragment thereof, 58P1D12 mRNA, alternative splice variant 58P1D12 mRNAs, and recombinant DNA or RNA molecules that contain a 58P1D12 polynucleotide. A number of methods for amplifying and/or detecting the presence of 58P1D12 polynucleotides are well known in the art and can be employed in the practice of this aspect of the invention.
  • In one embodiment, a method for detecting a 58P1D12 mRNA in a biological sample comprises producing cDNA from the sample by reverse transcription using at least one primer; amplifying the cDNA so produced using a 58P1D12 polynucleotides as sense and antisense primers to amplify 58P1D12 cDNAs therein; and detecting the presence of the amplified 58P1D12 cDNA. Optionally, the sequence of the amplified 58P1D12 cDNA can be determined.
  • In another embodiment, a method of detecting a 58P1D12 gene in a biological sample comprises first isolating genomic DNA from the sample; amplifying the isolated genomic DNA using 58P1D12 polynucleotides as sense and antisense primers; and detecting the presence of the amplified 58P1D12 gene. Any number of appropriate sense and antisense probe combinations can be designed from a 58P1D12 nucleotide sequence (see, e.g., FIG. 2) and used for this purpose.
  • The invention also provides assays for detecting the presence of a 58P1D12 protein in a tissue or other biological sample such as serum, semen, bone, prostate, urine, cell preparations, and the like. Methods for detecting a 58P1D12-related protein are also well known and include, for example, immunoprecipitation, immunohistochemical analysis, Western blot analysis, molecular binding assays, ELISA, ELIFA and the like. For example, a method of detecting the presence of a 58P1D12-related protein in a biological sample comprises first contacting the sample with a 58P1D12 antibody, a 58P1D12-reactive fragment thereof, or a recombinant protein containing an antigen-binding region of a 58P1D12 antibody; and then detecting the binding of 58P1D12-related protein in the sample.
  • Methods for identifying a cell that expresses 58P1D12 are also within the scope of the invention. In one embodiment, an assay for identifying a cell that expresses a 58P1D12 gene comprises detecting the presence of 58P1D12 mRNA in the cell. Methods for the detection of particular mRNAs in cells are well known and include, for example, hybridization assays using complementary DNA probes (such as in situ hybridization using labeled 58P1D12 riboprobes, Northern blot and related techniques) and various nucleic acid amplification assays (such as RT-PCR using complementary primers specific for 58P1D12, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like). Alternatively, an assay for identifying a cell that expresses a 58P1D12 gene comprises detecting the presence of 58P1D12-related protein in the cell or secreted by the cell. Various methods for the detection of proteins are well known in the art and are employed for the detection of 58P1D12-related proteins and cells that express 58P1D12-related proteins.
  • 58P1D12 expression analysis is also useful as a tool for identifying and evaluating agents that modulate 58P1D12 gene expression. For example, 58P1D12 expression is significantly upregulated in prostate cancer, and is expressed in cancers of the tissues listed in Table I. Identification of a molecule or biological agent that inhibits 58P1D12 expression or over-expression in cancer cells is of therapeutic value. For example, such an agent can be identified by using a screen that quantifies 58P1D12 expression by RT-PCR, nucleic acid hybridization or antibody binding.
  • VIII.) METHODS FOR MONITORING THE STATUS OF 58P1D12-RELATED GENES AND THEIR PRODUCTS
  • Oncogenesis is known to be a multistep process where cellular growth becomes progressively dysregulated and cells progress from a normal physiological state to precancerous and then cancerous states (see, e.g., Alers et al., Lab Invest. 77(5): 437-438 (1997) and Isaacs et al., Cancer Surv. 23: 19-32 (1995)). In this context, examining a biological sample for evidence of dysregulated cell growth (such as aberrant 58P1D12 expression in cancers) allows for early detection of such aberrant physiology, before a pathologic state such as cancer has progressed to a stage that therapeutic options are more limited and or the prognosis is worse. In such examinations, the status of 58P1D12 in a biological sample of interest can be compared, for example, to the status of 58P1D12 in a corresponding normal sample (e.g. a sample from that individual or alternatively another individual that is not affected by a pathology). An alteration in the status of 58P1D12 in the biological sample (as compared to the normal sample) provides evidence of dysregulated cellular growth. In addition to using a biological sample that is not affected by a pathology as a normal sample, one can also use a predetermined normative value such as a predetermined normal level of mRNA expression (see, e.g., Greyer et al., J. Comp. Neurol. 1996 Dec. 9; 376(2): 306-14 and U.S. Pat. No. 5,837,501) to compare 58P1D12 status in a sample.
  • The term “status” in this context is used according to its art accepted meaning and refers to the condition or state of a gene and its products. Typically, skilled artisans use a number of parameters to evaluate the condition or state of a gene and its products. These include, but are not limited to the location of expressed gene products (including the location of 58P1D12 expressing cells) as well as the level, and biological activity of expressed gene products (such as 58P1D12 mRNA, polynucleotides and polypeptides). Typically, an alteration in the status of 58P1D12 comprises a change in the location of 58P1D12 and/or 58P1D12 expressing cells and/or an increase in 58P1D12 mRNA and/or protein expression.
  • 58P1D12 status in a sample can be analyzed by a number of means well known in the art, including without limitation, immunohistochemical analysis, in situ hybridization, RT-PCR analysis on laser capture micro-dissected samples, Western blot analysis, and tissue array analysis. Typical protocols for evaluating the status of a 58P1D12 gene and gene products are found, for example in Ausubel et al. eds., 1995, Current Protocols In Molecular Biology, Units 2 (Northern Blotting), 4 (Southern Blotting), 15 (Immunoblotting) and 18 (PCR Analysis). Thus, the status of 58P1D12 in a biological sample is evaluated by various methods utilized by skilled artisans including, but not limited to genomic Southern analysis (to examine, for example perturbations in a 58P1D12 gene), Northern analysis and/or PCR analysis of 58P1D12 mRNA (to examine, for example alterations in the polynucleotide sequences or expression levels of 58P1D12 mRNAs), and, Western and/or immunohistochemical analysis (to examine, for example alterations in polypeptide sequences, alterations in polypeptide localization within a sample, alterations in expression levels of 58P1D12 proteins and/or associations of 58P1D12 proteins with polypeptide binding partners). Detectable 58P1D12 polynucleotides include, for example, a 58P1D12 gene or fragment thereof, 58P1D12 mRNA, alternative splice variants, 58P1D12 mRNAs, and recombinant DNA or RNA molecules containing a 58P1D12 polynucleotide.
  • The expression profile of 58P1D12 makes it a diagnostic marker for local and/or metastasized disease, and provides information on the growth or oncogenic potential of a biological sample. In particular, the status of 58P1D12 provides information useful for predicting susceptibility to particular disease stages, progression, and/or tumor aggressiveness. The invention provides methods and assays for determining 58P1D12 status and diagnosing cancers that express 58P1D12, such as cancers of the tissues listed in Table I. For example, because 58P1D12 mRNA is so highly expressed in prostate and other cancers relative to normal prostate tissue, assays that evaluate the levels of 58P1D12 mRNA transcripts or proteins in a biological sample can be used to diagnose a disease associated with 58P1D12 dysregulation, and can provide prognostic information useful in defining appropriate therapeutic options.
  • The expression status of 58P1D12 provides information including the presence, stage and location of dysplastic, precancerous and cancerous cells, predicting susceptibility to various stages of disease, and/or for gauging tumor aggressiveness. Moreover, the expression profile makes it useful as an imaging reagent for metastasized disease. Consequently, an aspect of the invention is directed to the various molecular prognostic and diagnostic methods for examining the status of 58P1D12 in biological samples such as those from individuals suffering from, or suspected of suffering from a pathology characterized by dysregulated cellular growth, such as cancer.
  • As described above, the status of 58P1D12 in a biological sample can be examined by a number of well-known procedures in the art. For example, the status of 58P1D12 in a biological sample taken from a specific location in the body can be examined by evaluating the sample for the presence or absence of 58P1D12 expressing cells (e.g. those that express 58P1D12 mRNAs or proteins). This examination can provide evidence of dysregulated cellular growth, for example, when 58P1D12-expressing cells are found in a biological sample that does not normally contain such cells (such as a lymph node), because such alterations in the status of 58P1D12 in a biological sample are often associated with dysregulated cellular growth. Specifically, one indicator of dysregulated cellular growth is the metastases of cancer cells from an organ of origin (such as the prostate) to a different area of the body (such as a lymph node). In this context, evidence of dysregulated cellular growth is important for example because occult lymph node metastases can be detected in a substantial proportion of patients with prostate cancer, and such metastases are associated with known predictors of disease progression (see, e.g., Murphy et al., Prostate 42(4): 315-317 (2000);Su et al., Semin Surg. Oncol. 18(1): 17-28 (2000) and Freeman et al., J Urol 1995 August 154(2 Pt 1):474-8).
  • In one aspect, the invention provides methods for monitoring 58P1D12 gene products by determining the status of 58P1D12 gene products expressed by cells from an individual suspected of having a disease associated with dysregulated cell growth (such as hyperplasia or cancer) and then comparing the status so determined to the status of 58P1D12 gene products in a corresponding normal sample. The presence of aberrant 58P1D12 gene products in the test sample relative to the normal sample provides an indication of the presence of dysregulated cell growth within the cells of the individual.
  • In another aspect, the invention provides assays useful in determining the presence of cancer in an individual, comprising detecting a significant increase in 58P1D12 mRNA or protein expression in a test cell or tissue sample relative to expression levels in the corresponding normal cell or tissue. The presence of 58P1D12 mRNA can, for example, be evaluated in tissues including but not limited to those listed in Table I. The presence of significant 58P1D12 expression in any of these tissues is useful to indicate the emergence, presence and/or severity of a cancer, since the corresponding normal tissues do not express 58P1D12 mRNA or express it at lower levels.
  • In a related embodiment, 58P1D12 status is determined at the protein level rather than at the nucleic acid level. For example, such a method comprises determining the level of 58P1D12 protein expressed by cells in a test tissue sample and comparing the level so determined to the level of 58P1D12 expressed in a corresponding normal sample. In one embodiment, the presence of 58P1D12 protein is evaluated, for example, using immunohistochemical methods. 58P1D12 antibodies or binding partners capable of detecting 58P1D12 protein expression are used in a variety of assay formats well known in the art for this purpose.
  • In a further embodiment, one can evaluate the status of 58P1D12 nucleotide and amino acid sequences in a biological sample in order to identify perturbations in the structure of these molecules. These perturbations can include insertions, deletions, substitutions and the like. Such evaluations are useful because perturbations in the nucleotide and amino acid sequences are observed in a large number of proteins associated with a growth dysregulated phenotype (see, e.g., Marrogi et al., 1999, J. Cutan. Pathol. 26(8):369-378). For example, a mutation in the sequence of 58P1D12 may be indicative of the presence or promotion of a tumor. Such assays therefore have diagnostic and predictive value where a mutation in 58P1D12 indicates a potential loss of function or increase in tumor growth.
  • A wide variety of assays for observing perturbations in nucleotide and amino acid sequences are well known in the art. For example, the size and structure of nucleic acid or amino acid sequences of 58P1D12 gene products are observed by the Northern, Southern, Western, PCR and DNA sequencing protocols discussed herein. In addition, other methods for observing perturbations in nucleotide and amino acid sequences such as single strand conformation polymorphism analysis are well known in the art (see, e.g., U.S. Pat. Nos. 5,382,510 issued 7 Sep. 1999, and 5,952,170 issued 17 Jan. 1995).
  • Additionally, one can examine the methylation status of a 58P1D12 gene in a biological sample. Aberrant demethylation and/or hypermethylation of CpG islands in gene 5′ regulatory regions frequently occurs in immortalized and transformed cells, and can result in altered expression of various genes. For example, promoter hypermethylation of the pi-class glutathione S-transferase (a protein expressed in normal prostate but not expressed in >90% of prostate carcinomas) appears to permanently silence transcription of this gene and is the most frequently detected genomic alteration in prostate carcinomas (De Marzo et al., Am. J. Pathol. 155(6): 1985-1992 (1999)). In addition, this alteration is present in at least 70% of cases of high-grade prostatic intraepithelial neoplasia (PIN) (Brooks et al., Cancer Epidemiol. Biomarkers Prey., 1998, 7:531-536). In another example, expression of the LAGE-I tumor specific gene (which is not expressed in normal prostate but is expressed in 25-50% of prostate cancers) is induced by deoxy-azacytidine in lymphoblastoid cells, suggesting that tumoral expression is due to demethylation (Lethe et al., Int. J. Cancer 76(6): 903-908 (1998)). A variety of assays for examining methylation status of a gene are well known in the art. For example, one can utilize, in Southern hybridization approaches, methylation-sensitive restriction enzymes that cannot cleave sequences that contain methylated CpG sites to assess the methylation status of CpG islands. In addition, MSP (methylation specific PCR) can rapidly profile the methylation status of all the CpG sites present in a CpG island of a given gene. This procedure involves initial modification of DNA by sodium bisulfite (which will convert all unmethylated cytosines to uracil) followed by amplification using primers specific for methylated versus unmethylated DNA. Protocols involving methylation interference can also be found for example in Current Protocols In Molecular Biology, Unit 12, Frederick M. Ausubel et al. eds., 1995.
  • Gene amplification is an additional method for assessing the status of 58P1D12. Gene amplification is measured in a sample directly, for example, by conventional Southern blotting or Northern blotting to quantitate the transcription of mRNA (Thomas, 1980, Proc. Natl. Acad. Sci. USA, 77:5201-5205), dot blotting (DNA analysis), or in situ hybridization, using an appropriately labeled probe, based on the sequences provided herein. Alternatively, antibodies are employed that recognize specific duplexes, including DNA duplexes, RNA duplexes, and DNA-RNA hybrid duplexes or DNA-protein duplexes. The antibodies in turn are labeled and the assay carried out where the duplex is bound to a surface, so that upon the formation of duplex on the surface, the presence of antibody bound to the duplex can be detected.
  • Biopsied tissue or peripheral blood can be conveniently assayed for the presence of cancer cells using for example, Northern, dot blot or RT-PCR analysis to detect 58P1D12 expression. The presence of RT-PCR amplifiable 58P1D12 mRNA provides an indication of the presence of cancer. RT-PCR assays are well known in the art. RT-PCR detection assays for tumor cells in peripheral blood are currently being evaluated for use in the diagnosis and management of a number of human solid tumors. In the prostate cancer field, these include RT-PCR assays for the detection of cells expressing PSA and PSM (Verkaik et al., 1997, Urol. Res. 25:373-384; Ghossein et al., 1995, J. Clin. Oncol. 13:1195-2000; Heston et al., 1995, Clin. Chem. 41:1687-1688).
  • A further aspect of the invention is an assessment of the susceptibility that an individual has for developing cancer. In one embodiment, a method for predicting susceptibility to cancer comprises detecting 58P1D12 mRNA or 58P1D12 protein in a tissue sample, its presence indicating susceptibility to cancer, wherein the degree of 58P1D12 mRNA expression correlates to the degree of susceptibility. In a specific embodiment, the presence of 58P1D12 in prostate or other tissue is examined, with the presence of 58P1D12 in the sample providing an indication of prostate cancer susceptibility (or the emergence or existence of a prostate tumor). Similarly, one can evaluate the integrity 58P1D12 nucleotide and amino acid sequences in a biological sample, in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like. The presence of one or more perturbations in 58P1D12 gene products in the sample is an indication of cancer susceptibility (or the emergence or existence of a tumor).
  • The invention also comprises methods for gauging tumor aggressiveness. In one embodiment, a method for gauging aggressiveness of a tumor comprises determining the level of 58P1D12 mRNA or 58P1D12 protein expressed by tumor cells, comparing the level so determined to the level of 58P1D12 mRNA or 58P1D12 protein expressed in a corresponding normal tissue taken from the same individual or a normal tissue reference sample, wherein the degree of 58P1D12 mRNA or 58P1D12 protein expression in the tumor sample relative to the normal sample indicates the degree of aggressiveness. In a specific embodiment, aggressiveness of a tumor is evaluated by determining the extent to which 58P1D12 is expressed in the tumor cells, with higher expression levels indicating more aggressive tumors. Another embodiment is the evaluation of the integrity of 58P1D12 nucleotide and amino acid sequences in a biological sample, in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like. The presence of one or more perturbations indicates more aggressive tumors.
  • Another embodiment of the invention is directed to methods for observing the progression of a malignancy in an individual over time. In one embodiment, methods for observing the progression of a malignancy in an individual over time comprise determining the level of 58P1D12 mRNA or 58P1D12 protein expressed by cells in a sample of the tumor, comparing the level so determined to the level of 58P1D12 mRNA or 58P1D12 protein expressed in an equivalent tissue sample taken from the same individual at a different time, wherein the degree of 58P1D12 mRNA or 58P1D12 protein expression in the tumor sample over time provides information on the progression of the cancer. In a specific embodiment, the progression of a cancer is evaluated by determining 58P1D12 expression in the tumor cells over time, where increased expression over time indicates a progression of the cancer. Also, one can evaluate the integrity 58P1D12 nucleotide and amino acid sequences in a biological sample in order to identify perturbations in the structure of these molecules such as insertions, deletions, substitutions and the like, where the presence of one or more perturbations indicates a progression of the cancer.
  • The above diagnostic approaches can be combined with any one of a wide variety of prognostic and diagnostic protocols known in the art. For example, another embodiment of the invention is directed to methods for observing a coincidence between the expression of 58P1D12 gene and 58P1D12 gene products (or perturbations in 58P1D12 gene and 58P1D12 gene products) and a factor that is associated with malignancy, as a means for diagnosing and prognosticating the status of a tissue sample. A wide variety of factors associated with malignancy can be utilized, such as the expression of genes associated with malignancy (e.g. PSA, PSCA and PSM expression for prostate cancer etc.) as well as gross cytological observations (see, e.g., Bocking et al., 1984, Anal. Quant. Cytol. 6(2):74-88; Epstein, 1995, Hum. Pathol. 26(2):223-9; Thorson et al., 1998, Mod. Pathol. 11(6):543-51; Baisden et al., 1999, Am. J. Surg. Pathol. 23(8):918-24). Methods for observing a coincidence between the expression of 58P1D12 gene and 58P1D12 gene products (or perturbations in 58P1D12 gene and 58P1D12 gene products) and another factor that is associated with malignancy are useful, for example, because the presence of a set of specific factors that coincide with disease provides information crucial for diagnosing and prognosticating the status of a tissue sample.
  • In one embodiment, methods for observing a coincidence between the expression of 58P1D12 gene and 58P1D12 gene products (or perturbations in 58P1D12 gene and 58P1D12 gene products) and another factor associated with malignancy entails detecting the overexpression of 58P1D12 mRNA or protein in a tissue sample, detecting the overexpression of PSA mRNA or protein in a tissue sample (or PSCA or PSM expression), and observing a coincidence of 58P1D12 mRNA or protein and PSA mRNA or protein overexpression (or PSCA or PSM expression). In a specific embodiment, the expression of 58P1D12 and PSA mRNA in prostate tissue is examined, where the coincidence of 58P1D12 and PSA mRNA overexpression in the sample indicates the existence of prostate cancer, prostate cancer susceptibility or the emergence or status of a prostate tumor.
  • Methods for detecting and quantifying the expression of 58P1D12 mRNA or protein are described herein, and standard nucleic acid and protein detection and quantification technologies are well known in the art. Standard methods for the detection and quantification of 58P1D12 mRNA include in situ hybridization using labeled 58P1D12 riboprobes, Northern blot and related techniques using 58P1D12 polynucleotide probes, RT-PCR analysis using primers specific for 58P1D12, and other amplification type detection methods, such as, for example, branched DNA, SISBA, TMA and the like. In a specific embodiment, semi-quantitative RT-PCR is used to detect and quantify 58P1D12 mRNA expression. Any number of primers capable of amplifying 58P1D12 can be used for this purpose, including but not limited to the various primer sets specifically described herein. In a specific embodiment, polyclonal or monoclonal antibodies specifically reactive with the wild-type 58P1D12 protein can be used in an immunohistochemical assay of biopsied tissue.
  • IX.) IDENTIFICATION OF MOLECULES THAT INTERACT WITH 58P1D12
  • The 58P1D12 protein and nucleic acid sequences disclosed herein allow a skilled artisan to identify proteins, small molecules and other agents that interact with 58P1D12, as well as pathways activated by 58P1D12 via any one of a variety of art accepted protocols. For example, one can utilize one of the so-called interaction trap systems (also referred to as the “two-hybrid assay”). In such systems, molecules interact and reconstitute a transcription factor which directs expression of a reporter gene, whereupon the expression of the reporter gene is assayed. Other systems identify protein-protein interactions in vivo through reconstitution of a eukaryotic transcriptional activator, see, e.g., U.S. Pat. Nos. 5,955,280 issued 21 Sep. 1999, 5,925,523 issued 20 Jul. 1999, 5,846,722 issued 8 Dec. 1998 and 6,004,746 issued 21 Dec. 1999. Algorithms are also available in the art for genome-based predictions of protein function (see, e.g., Marcotte, et al., Nature 402: 4 Nov. 1999, 83-86).
  • Alternatively one can screen peptide libraries to identify molecules that interact with 58P1D12 protein sequences. In such methods, peptides that bind to 58P1D12 are identified by screening libraries that encode a random or controlled collection of amino acids. Peptides encoded by the libraries are expressed as fusion proteins of bacteriophage coat proteins, the bacteriophage particles are then screened against the 58P1D12 protein(s).
  • Accordingly, peptides having a wide variety of uses, such as therapeutic, prognostic or diagnostic reagents, are thus identified without any prior information on the structure of the expected ligand or receptor molecule. Typical peptide libraries and screening methods that can be used to identify molecules that interact with 58P1D12 protein sequences are disclosed for example in U.S. Pat. Nos. 5,723,286 issued 3 Mar. 1998 and 5,733,731 issued 31 Mar. 1998.
  • Alternatively, cell lines that express 58P1D12 are used to identify protein-protein interactions mediated by 58P1D12. Such interactions can be examined using immunoprecipitation techniques (see, e.g., Hamilton B. J., et al. Biochem. Biophys. Res. Commun 1999, 261:646-51). 58P1D12 protein can be immunoprecipitated from 58P1D12-expressing cell lines using anti-58P1D12 antibodies. Alternatively, antibodies against His-tag can be used in a cell line engineered to express fusions of 58P1D12 and a His-tag (vectors mentioned above). The immunoprecipitated complex can be examined for protein association by procedures such as Western blotting, 35S-methionine labeling of proteins, protein microsequencing, silver staining and two-dimensional gel electrophoresis.
  • Small molecules and ligands that interact with 58P1D12 can be identified through related embodiments of such screening assays. For example, small molecules can be identified that interfere with protein function, including molecules that interfere with 58P1D12's ability to mediate phosphorylation and de-phosphorylation, interaction with DNA or RNA molecules as an indication of regulation of cell cycles, second messenger signaling or tumorigenesis. Similarly, small molecules that modulate 58P1D12-related ion channel, protein pump, or cell communication functions are identified and used to treat patients that have a cancer that expresses 58P1D12 (see, e.g., Hille, B., Ionic Channels of Excitable Membranes 2nd Ed., Sinauer Assoc., Sunderland, Mass., 1992). Moreover, ligands that regulate 58P1D12 function can be identified based on their ability to bind 58P1D12 and activate a reporter construct. Typical methods are discussed for example in U.S. Pat. No. 5,928,868 issued 27 Jul. 1999, and include methods for forming hybrid ligands in which at least one ligand is a small molecule. In an illustrative embodiment, cells engineered to express a fusion protein of 58P1D12 and a DNA-binding protein are used to co-express a fusion protein of a hybrid ligand/small molecule and a cDNA library transcriptional activator protein. The cells further contain a reporter gene, the expression of which is conditioned on the proximity of the first and second fusion proteins to each other, an event that occurs only if the hybrid ligand binds to target sites on both hybrid proteins. Those cells that express the reporter gene are selected and the unknown small molecule or the unknown ligand is identified. This method provides a means of identifying modulators, which activate or inhibit 58P1D12.
  • An embodiment of this invention comprises a method of screening for a molecule that interacts with a 58P1D12 amino acid sequence shown in FIG. 2 or FIG. 3, comprising the steps of contacting a population of molecules with a 58P1D12 amino acid sequence, allowing the population of molecules and the 58P1D12 amino acid sequence to interact under conditions that facilitate an interaction, determining the presence of a molecule that interacts with the 58P1D12 amino acid sequence, and then separating molecules that do not interact with the 58P1D12 amino acid sequence from molecules that do. In a specific embodiment, the method further comprises purifying, characterizing and identifying a molecule that interacts with the 58P1D12 amino acid sequence. The identified molecule can be used to modulate a function performed by 58P1D12. In a preferred embodiment, the 58P1D12 amino acid sequence is contacted with a library of peptides.
  • X.) THERAPEUTIC METHODS AND COMPOSITIONS
  • The identification of 58P1D12 as a protein that is normally expressed in a restricted set of tissues, but which is also expressed in cancers such as those listed in Table I, opens a number of therapeutic approaches to the treatment of such cancers.
  • Of note, targeted antitumor therapies have been useful even when the targeted protein is expressed on normal tissues, even vital normal organ tissues. A vital organ is one that is necessary to sustain life, such as the heart or colon. A non-vital organ is one that can be removed whereupon the individual is still able to survive. Examples of non-vital organs are ovary, breast, and prostate.
  • For example, Herceptin® is an FDA approved pharmaceutical that has as its active ingredient an antibody which is immunoreactive with the protein variously known as HER2, HER2/neu, and erb-b-2. It is marketed by Genentech and has been a commercially successful antitumor agent. Herceptin sales reached almost $400 million in 2002. Herceptin is a treatment for HER2 positive metastatic breast cancer. However, the expression of HER2 is not limited to such tumors. The same protein is expressed in a number of normal tissues. In particular, it is known that HER2/neu is present in normal kidney and heart, thus these tissues are present in all human recipients of Herceptin. The presence of HER2/neu in normal kidney is also confirmed by Latif, Z., et al., B.J.U. International (2002) 89:5-9. As shown in this article (which evaluated whether renal cell carcinoma should be a preferred indication for anti-HER2 antibodies such as Herceptin) both protein and mRNA are produced in benign renal tissues. Notably, HER2/neu protein was strongly overexpressed in benign renal tissue.
  • Despite the fact that HER2/neu is expressed in such vital tissues as heart and kidney, Herceptin is a very useful, FDA approved, and commercially successful drug. The effect of Herceptin on cardiac tissue, i.e., “cardiotoxicity,” has merely been a side effect to treatment. When patients were treated with Herceptin alone, significant cardiotoxicity occurred in a very low percentage of patients.
  • Of particular note, although kidney tissue is indicated to exhibit normal expression, possibly even higher expression than cardiac tissue, kidney has no appreciable Herceptin side effect whatsoever. Moreover, of the diverse array of normal tissues in which HER2 is expressed, there is very little occurrence of any side effect. Only cardiac tissue has manifested any appreciable side effect at all. A tissue such as kidney, where HER2/neu expression is especially notable, has not been the basis for any side effect.
  • Furthermore, favorable therapeutic effects have been found for antitumor therapies that target epidermal growth factor receptor (EGFR). EGFR is also expressed in numerous normal tissues. There have been very limited side effects in normal tissues following use of anti-EGFR therapeutics.
  • Thus, expression of a target protein in normal tissue, even vital normal tissue, does not defeat the utility of a targeting agent for the protein as a therapeutic for certain tumors in which the protein is also overexpressed.
  • Accordingly, therapeutic approaches that inhibit the activity of a 58P1D12 protein are useful for patients suffering from a cancer that expresses 58P1D12. These therapeutic approaches generally fall into two classes. One class comprises various methods for inhibiting the binding or association of a 58P1D12 protein with its binding partner or with other proteins. Another class comprises a variety of methods for inhibiting the transcription of a 58P1D12 gene or translation of 58P1D12 mRNA.
  • X.A.) Anti-Cancer Vaccines
  • The invention provides cancer vaccines comprising a 58P1D12-related protein or 58P1D12-related nucleic acid. In view of the expression of 58P1D12, cancer vaccines prevent and/or treat 58P1D12-expressing cancers with minimal or no effects on non-target tissues. The use of a tumor antigen in a vaccine that generates humoral and/or cell-mediated immune responses as anti-cancer therapy is well known in the art and has been employed in prostate cancer using human PSMA and rodent PAP immunogens (Hodge et al., 1995, Int. J. Cancer 63:231-237; Fong et al., 1997, J. Immunol. 159:3113-3117).
  • Such methods can be readily practiced by employing a 58P1D12-related protein, or a 58P1D12-encoding nucleic acid molecule and recombinant vectors capable of expressing and presenting the 58P1D12 immunogen (which typically comprises a number of antibody or T cell epitopes). Skilled artisans understand that a wide variety of vaccine systems for delivery of immunoreactive epitopes are known in the art (see, e.g., Heryln et al., Ann Med 1999 February 31(1):66-78; Maruyama et al., Cancer Immunol Immunother 2000 June 49(3):123-32) Briefly, such methods of generating an immune response (e.g. humoral and/or cell-mediated) in a mammal, comprise the steps of: exposing the mammal's immune system to an immunoreactive epitope (e.g. an epitope present in a 58P1D12 protein shown in FIG. 3 or analog or homolog thereof) so that the mammal generates an immune response that is specific for that epitope (e.g. generates antibodies that specifically recognize that epitope). In a preferred method, a 58P1D12 immunogen contains a biological motif, see e.g., Tables VIII-XXI and XXII-XLIX, or a peptide of a size range from 58P1D12 indicated in FIG. 5, FIG. 6, FIG. 7, FIG. 8, and FIG. 9.
  • The entire 58P1D12 protein, immunogenic regions or epitopes thereof can be combined and delivered by various means. Such vaccine compositions can include, for example, lipopeptides (e.g.,Vitiello, A. et al., J. Clin. Invest. 95:341, 1995), peptide compositions encapsulated in poly(DL-lactide-co-glycolide) (“PLG”) microspheres (see, e.g., Eldridge, et al., Molec. Immunol. 28:287-294, 1991: Alonso et al., Vaccine 12:299-306, 1994; Jones et al., Vaccine 13:675-681, 1995), peptide compositions contained in immune stimulating complexes (ISCOMS) (see, e.g., Takahashi et al., Nature 344:873-875, 1990; Hu et al., Clin Exp Immunol. 113:235-243, 1998), multiple antigen peptide systems (MAPs) (see e.g., Tam, J. P., Proc. Natl. Acad. Sci. U.S.A. 85:5409-5413, 1988; Tam, J.P., J. Immunol. Methods 196:17-32, 1996), peptides formulated as multivalent peptides; peptides for use in ballistic delivery systems, typically crystallized peptides, viral delivery vectors (Perkus, M. E. et al., In: Concepts in vaccine development, Kaufmann, S. H. E., ed., p. 379, 1996; Chakrabarti, S. et al., Nature 320:535, 1986; Hu, S. L. et al., Nature 320:537, 1986; Kieny, M.-P. et al., AIDS Bio/Technology 4:790, 1986; Top, F. H. et al., J. Infect. Dis. 124:148, 1971; Chanda, P. K. et al., Virology 175:535, 1990), particles of viral or synthetic origin (e.g., Kotler, N. et al., J. Immunol. Methods. 192:25, 1996; Eldridge, J. H. et al., Sem. Hematol. 30:16, 1993; Falo, L. D., Jr. et al., Nature Med. 7:649, 1995), adjuvants (Warren, H. S., Vogel, F. R., and Chedid, L. A. Annu. Rev. Immunol. 4:369, 1986; Gupta, R. K. et al., Vaccine 11:293, 1993), liposomes (Reddy, R. et al., J. Immunol. 148:1585, 1992; Rock, K. L., Immunol. Today 17:131, 1996), or, naked or particle absorbed cDNA (Ulmer, J. B. et al., Science 259:1745, 1993; Robinson, H. L., Hunt, L. A., and Webster, R. G., Vaccine 11:957, 1993; Shiver, J. W. et al., In: Concepts in vaccine development, Kaufmann, S. H. E., ed., p. 423, 1996; Cease, K. B., and Berzofsky, J. A., Annu. Rev. Immunol. 12:923, 1994 and Eldridge, J. H. et al., Sem. Hematol. 30:16, 1993). Toxin-targeted delivery technologies, also known as receptor mediated targeting, such as those of Avant Immunotherapeutics, Inc. (Needham, Mass.) may also be used.
  • In patients with 58P1D12-associated cancer, the vaccine compositions of the invention can also be used in conjunction with other treatments used for cancer, e.g., surgery, chemotherapy, drug therapies, radiation therapies, etc. including use in combination with immune adjuvants such as IL-2, IL-12, GM-CSF, and the like.
  • Cellular Vaccines:
  • CTL epitopes can be determined using specific algorithms to identify peptides within 58P1D12 protein that bind corresponding HLA alleles (see e.g., Table IV; Epimer™ and Epimatrix™, Brown University (URL brown.edu/Research/TB-HIV_Lab/epimatrix/epimatrix.html); and, BIMAS, (URL bimas.dcrt.nih gov/; SYFPEITHI at URL syfpeithi.bmi-heidelberg.com/). In a preferred embodiment, a 58P1D12 immunogen contains one or more amino acid sequences identified using techniques well known in the art, such as the sequences shown in Tables VIII-XXI and XXII-XLIX or a peptide of 8, 9, 10 or 11 amino acids specified by an HLA Class I motif/supermotif (e.g., Table IV (A), Table IV (D), or Table IV (E)) and/or a peptide of at least 9 amino acids that comprises an HLA Class II motif/supermotif (e.g., Table IV (B) or Table IV (C)). As is appreciated in the art, the HLA Class I binding groove is essentially closed ended so that peptides of only a particular size range can fit into the groove and be bound, generally HLA Class I epitopes are 8, 9, 10, or 11 amino acids long. In contrast, the HLA Class II binding groove is essentially open ended; therefore a peptide of about 9 or more amino acids can be bound by an HLA Class II molecule. Due to the binding groove differences between HLA Class I and II, HLA Class I motifs are length specific, i.e., position two of a Class I motif is the second amino acid in an amino to carboxyl direction of the peptide. The amino acid positions in a Class II motif are relative only to each other, not the overall peptide, i.e., additional amino acids can be attached to the amino and/or carboxyl termini of a motif-bearing sequence. HLA Class II epitopes are often 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 amino acids long, or longer than 25 amino acids.
  • Antibody-Based Vaccines
  • A wide variety of methods for generating an immune response in a mammal are known in the art (for example as the first step in the generation of hybridomas). Methods of generating an immune response in a mammal comprise exposing the mammal's immune system to an immunogenic epitope on a protein (e.g. a 58P1D12 protein) so that an immune response is generated. A typical embodiment consists of a method for generating an immune response to 58P1D12 in a host, by contacting the host with a sufficient amount of at least one 58P1D12 B cell or cytotoxic T-cell epitope or analog thereof; and at least one periodic interval thereafter re-contacting the host with the 58P1D12 B cell or cytotoxic T-cell epitope or analog thereof. A specific embodiment consists of a method of generating an immune response against a 58P1D12-related protein or a man-made multiepitopic peptide comprising: administering 58P1D12 immunogen (e.g. a 58P1D12 protein or a peptide fragment thereof, a 58P1D12 fusion protein or analog etc.) in a vaccine preparation to a human or another mammal. Typically, such vaccine preparations further contain a suitable adjuvant (see, e.g., U.S. Pat. No. 6,146,635) or a universal helper epitope such as a PADRE™peptide (Epimmune Inc., San Diego, Calif.; see, e.g., Alexander et al., J. Immunol. 2000 164(3); 164(3): 1625-1633; Alexander et al., Immunity 1994 1(9): 751-761 and Alexander et al., Immunol. Res. 1998 18(2): 79-92). An alternative method comprises generating an immune response in an individual against a 58P1D12 immunogen by: administering in vivo to muscle or skin of the individual's body a DNA molecule that comprises a DNA sequence that encodes a 58P1D12 immunogen, the DNA sequence operatively linked to regulatory sequences which control the expression of the DNA sequence; wherein the DNA molecule is taken up by cells, the DNA sequence is expressed in the cells and an immune response is generated against the immunogen (see, e.g., U.S. Pat. No. 5,962,428). Optionally a genetic vaccine facilitator such as anionic lipids; saponins; lectins; estrogenic compounds; hydroxylated lower alkyls; dimethyl sulfoxide; and urea is also administered. In addition, an antiidiotypic antibody can be administered that mimics 58P1D12, in order to generate a response to the target antigen.
  • Nucleic Acid Vaccines:
  • Vaccine compositions of the invention include nucleic acid-mediated modalities. DNA or RNA that encode protein(s) of the invention can be administered to a patient. Genetic immunization methods can be employed to generate prophylactic or therapeutic humoral and cellular immune responses directed against cancer cells expressing 58P1D12. Constructs comprising DNA encoding a 58P1D12-related protein/immunogen and appropriate regulatory sequences can be injected directly into muscle or skin of an individual, such that the cells of the muscle or skin take-up the construct and express the encoded 58P1D12 protein/immunogen. Alternatively, a vaccine comprises a 58P1D12-related protein. Expression of the 58P1D12-related protein immunogen results in the generation of prophylactic or therapeutic humoral and cellular immunity against cells that bear a 58P1D12 protein. Various prophylactic and therapeutic genetic immunization techniques known in the art can be used (for review, see information and references published at Internet address genweb.com). Nucleic acid-based delivery is described, for instance, in Wolff et. al., Science 247:1465 (1990) as well as U.S. Pat. Nos. 5,580,859; 5,589,466; 5,804,566; 5,739,118; 5,736,524; 5,679,647; WO 98/04720. Examples of DNA-based delivery technologies include “naked DNA”, facilitated (bupivicaine, polymers, peptide-mediated) delivery, cationic lipid complexes, and particle-mediated (“gene gun”) or pressure-mediated delivery (see, e.g., U.S. Pat. No. 5,922,687).
  • For therapeutic or prophylactic immunization purposes, proteins of the invention can be expressed via viral or bacterial vectors. Various viral gene delivery systems that can be used in the practice of the invention include, but are not limited to, vaccinia, fowlpox, canarypox, adenovirus, influenza, poliovirus, adeno-associated virus, lentivirus, and sindbis virus (see, e.g., Restifo, 1996, Curr. Opin. Immunol. 8:658-663; Tsang et al. J. Natl. Cancer Inst. 87:982-990 (1995)). Non-viral delivery systems can also be employed by introducing naked DNA encoding a 58P1D12-related protein into the patient (e.g., intramuscularly or intradermally) to induce an anti-tumor response.
  • Vaccinia virus is used, for example, as a vector to express nucleotide sequences that encode the peptides of the invention. Upon introduction into a host, the recombinant vaccinia virus expresses the protein immunogenic peptide, and thereby elicits a host immune response. Vaccinia vectors and methods useful in immunization protocols are described in, e.g., U.S. Pat. No. 4,722,848. Another vector is BCG (Bacille Calmette Guerin). BCG vectors are described in Stover et al., Nature 351:456-460 (1991). A wide variety of other vectors useful for therapeutic administration or immunization of the peptides of the invention, e.g. adeno and adeno-associated virus vectors, retroviral vectors, Salmonella typhi vectors, detoxified anthrax toxin vectors, and the like, will be apparent to those skilled in the art from the description herein.
  • Thus, gene delivery systems are used to deliver a 58P1D12-related nucleic acid molecule. In one embodiment, the full-length human 58P1D12 cDNA is employed. In another embodiment, 58P1D12 nucleic acid molecules encoding specific cytotoxic T lymphocyte (CTL) and/or antibody epitopes are employed.
  • Ex Vivo Vaccines
  • Various ex vivo strategies can also be employed to generate an immune response. One approach involves the use of antigen presenting cells (APCs) such as dendritic cells (DC) to present 58P1D12 antigen to a patient's immune system. Dendritic cells express MHC class I and II molecules, B7 co-stimulator, and IL-12, and are thus highly specialized antigen presenting cells. In prostate cancer, autologous dendritic cells pulsed with peptides of the prostate-specific membrane antigen (PSMA) are being used in a Phase I clinical trial to stimulate prostate cancer patients' immune systems (Tjoa et al., 1996, Prostate 28:65-69; Murphy et al., 1996, Prostate 29:371-380). Thus, dendritic cells can be used to present 58P1D12 peptides to T cells in the context of MHC class I or II molecules. In one embodiment, autologous dendritic cells are pulsed with 58P1D12 peptides capable of binding to MHC class I and/or class II molecules. In another embodiment, dendritic cells are pulsed with the complete 58P1D12 protein. Yet another embodiment involves engineering the overexpression of a 58P1D12 gene in dendritic cells using various implementing vectors known in the art, such as adenovirus (Arthur et al., 1997, Cancer Gene Ther. 4:17-25), retrovirus (Henderson et al., 1996, Cancer Res. 56:3763-3770), lentivirus, adeno-associated virus, DNA transfection (Ribas et al., 1997, Cancer Res. 57:2865-2869), or tumor-derived RNA transfection (Ashley et al., 1997, J. Exp. Med. 186:1177-1182). Cells that express 58P1D12 can also be engineered to express immune modulators, such as GM-CSF, and used as immunizing agents.
  • X.B.) 58P1D12 as a Target for Antibody-Based Therapy
  • 58P1D12 is an attractive target for antibody-based therapeutic strategies. A number of antibody strategies are known in the art for targeting both extracellular and intracellular molecules (see, e.g., complement and ADCC mediated killing as well as the use of intrabodies). Because 58P1D12 is expressed by cancer cells of various lineages relative to corresponding normal cells, systemic administration of 58P1D12-immunoreactive compositions are prepared that exhibit excellent sensitivity without toxic, non-specific and/or non-target effects caused by binding of the immunoreactive composition to non-target organs and tissues. Antibodies specifically reactive with domains of 58P1D12 are useful to treat 58P1D12-expressing cancers systemically, either as conjugates with a toxin or therapeutic agent, or as naked antibodies capable of inhibiting cell proliferation or function.
  • 58P1D12 antibodies can be introduced into a patient such that the antibody binds to 58P1D12 and modulates a function, such as an interaction with a binding partner, and consequently mediates destruction of the tumor cells and/or inhibits the growth of the tumor cells. Mechanisms by which such antibodies exert a therapeutic effect can include complement-mediated cytolysis, antibody-dependent cellular cytotoxicity, modulation of the physiological function of 58P1D12, inhibition of ligand binding or signal transduction pathways, modulation of tumor cell differentiation, alteration of tumor angiogenesis factor profiles, and/or apoptosis.
  • Those skilled in the art understand that antibodies can be used to specifically target and bind immunogenic molecules such as an immunogenic region of a 58P1D12 sequence shown in FIG. 2 or FIG. 3. In addition, skilled artisans understand that it is routine to conjugate antibodies to cytotoxic agents (see, e.g., Slevers et al. Blood 93:11 3678-3684 (Jun. 1, 1999)). When cytotoxic and/or therapeutic agents are delivered directly to cells, such as by conjugating them to antibodies specific for a molecule expressed by that cell (e.g. 58P1D12), the cytotoxic agent will exert its known biological effect (i.e. cytotoxicity) on those cells.
  • A wide variety of compositions and methods for using antibody-cytotoxic agent conjugates to kill cells are known in the art. In the context of cancers, typical methods entail administering to an animal having a tumor a biologically effective amount of a conjugate comprising a selected cytotoxic and/or therapeutic agent linked to a targeting agent (e.g. an anti-58P1D12 antibody) that binds to a marker (e.g. 58P1D12) expressed, accessible to binding or localized on the cell surfaces. A typical embodiment is a method of delivering a cytotoxic and/or therapeutic agent to a cell expressing 58P1D12, comprising conjugating the cytotoxic agent to an antibody that immunospecifically binds to a 58P1D12 epitope, and, exposing the cell to the antibody-agent conjugate. Another illustrative embodiment is a method of treating an individual suspected of suffering from metastasized cancer, comprising a step of administering parenterally to said individual a pharmaceutical composition comprising a therapeutically effective amount of an antibody conjugated to a cytotoxic and/or therapeutic agent.
  • Cancer immunotherapy using anti-58P1D12 antibodies can be done in accordance with various approaches that have been successfully employed in the treatment of other types of cancer, including but not limited to colon cancer (Arlen et al., 1998, Crit. Rev. Immunol. 18:133-138), multiple myeloma (Ozaki et al., 1997, Blood 90:3179-3186, Tsunenari et al., 1997, Blood 90:2437-2444), gastric cancer (Kasprzyk et al., 1992, Cancer Res. 52:2771-2776), B-cell lymphoma (Funakoshi et al., 1996, J. Immunother. Emphasis Tumor Immunol. 19:93-101), leukemia (Zhong et al., 1996, Leuk. Res. 20:581-589), colorectal cancer (Moun et al., 1994, Cancer Res. 54:6160-6166; Velders et al., 1995, Cancer Res. 55:4398-4403), and breast cancer (Shepard et al., 1991, J. Clin. Immunol. 11:117-127). Some therapeutic approaches involve conjugation of naked antibody to a toxin or radioisotope, such as the conjugation of Y91 or I131 to anti-CD20 antibodies (e.g., Zevalin™, IDEC Pharmaceuticals Corp. or Bexxar™, Coulter Pharmaceuticals), while others involve co-administration of antibodies and other therapeutic agents, such as Herceptin™ (trastuzumab) with paclitaxel (Genentech, Inc.). The antibodies can be conjugated to a therapeutic agent. To treat prostate cancer, for example, 58P1D12 antibodies can be administered in conjunction with radiation, chemotherapy or hormone ablation. Also, antibodies can be conjugated to a toxin such as calicheamicin (e.g., Mylotarg™, Wyeth-Ayerst, Madison, N J, a recombinant humanized IgG4 kappa antibody conjugated to antitumor antibiotic calicheamicin) or a maytansinoid (e.g., taxane-based Tumor-Activated Prodrug, TAP, platform, ImmunoGen, Cambridge, Mass., also see e.g., U.S. Pat. No. 5,416,064).
  • Although 58P1D12 antibody therapy is useful for all stages of cancer, antibody therapy can be particularly appropriate in advanced or metastatic cancers. Treatment with the antibody therapy of the invention is indicated for patients who have received one or more rounds of chemotherapy. Alternatively, antibody therapy of the invention is combined with a chemotherapeutic or radiation regimen for patients who have not received chemotherapeutic treatment. Additionally, antibody therapy can enable the use of reduced dosages of concomitant chemotherapy, particularly for patients who do not tolerate the toxicity of the chemotherapeutic agent very well. Fan et al. (Cancer Res. 53:4637-4642, 1993), Prewett et al. (International J. of Onco. 9:217-224, 1996), and Hancock et al. (Cancer Res. 51:4575-4580, 1991) describe the use of various antibodies together with chemotherapeutic agents.
  • Although 58P1D12 antibody therapy is useful for all stages of cancer, antibody therapy can be particularly appropriate in advanced or metastatic cancers. Treatment with the antibody therapy of the invention is indicated for patients who have received one or more rounds of chemotherapy. Alternatively, antibody therapy of the invention is combined with a chemotherapeutic or radiation regimen for patients who have not received chemotherapeutic treatment. Additionally, antibody therapy can enable the use of reduced dosages of concomitant chemotherapy, particularly for patients who do not tolerate the toxicity of the chemotherapeutic agent very well.
  • Cancer patients can be evaluated for the presence and level of 58P1D12 expression, preferably using immunohistochemical assessments of tumor tissue, quantitative 58P1D12 imaging, or other techniques that reliably indicate the presence and degree of 58P1D12 expression. Immunohistochemical analysis of tumor biopsies or surgical specimens is preferred for this purpose. Methods for immunohistochemical analysis of tumor tissues are well known in the art.
  • Anti-58P1D12 monoclonal antibodies that treat prostate and other cancers include those that initiate a potent immune response against the tumor or those that are directly cytotoxic. In this regard, anti-58P1D12 monoclonal antibodies (mAbs) can elicit tumor cell lysis by either complement-mediated or antibody-dependent cell cytotoxicity (ADCC) mechanisms, both of which require an intact Fc portion of the immunoglobulin molecule for interaction with effector cell Fc receptor sites on complement proteins. In addition, anti-58P1D12 mAbs that exert a direct biological effect on tumor growth are useful to treat cancers that express 58P1D12. Mechanisms by which directly cytotoxic mAbs act include: inhibition of cell growth, modulation of cellular differentiation, modulation of tumor angiogenesis factor profiles, and the induction of apoptosis. The mechanism(s) by which a particular anti-58P1D12 mAb exerts an anti-tumor effect is evaluated using any number of in vitro assays that evaluate cell death such as ADCC, ADMMC, complement-mediated cell lysis, and so forth, as is generally known in the art.
  • In some patients, the use of murine or other non-human monoclonal antibodies, or human/mouse chimeric mAbs can induce moderate to strong immune responses against the non-human antibody. This can result in clearance of the antibody from circulation and reduced efficacy. In the most severe cases, such an immune response can lead to the extensive formation of immune complexes which, potentially, can cause renal failure. Accordingly, preferred monoclonal antibodies used in the therapeutic methods of the invention are those that are either fully human or humanized and that bind specifically to the target 58P1D12 antigen with high affinity but exhibit low or no antigenicity in the patient.
  • Therapeutic methods of the invention contemplate the administration of single anti-58P1D12 mAbs as well as combinations, or cocktails, of different mAbs. Such mAb cocktails can have certain advantages inasmuch as they contain mAbs that target different epitopes, exploit different effector mechanisms or combine directly cytotoxic mAbs with mAbs that rely on immune effector functionality. Such mAbs in combination can exhibit synergistic therapeutic effects. In addition, anti-58P1D12 mAbs can be administered concomitantly with other therapeutic modalities, including but not limited to various chemotherapeutic agents, androgen-blockers, immune modulators (e.g., IL-2, GM-CSF), surgery or radiation. The anti-58P1D12 mAbs are administered in their “naked” or unconjugated form, or can have a therapeutic agent(s) conjugated to them.
  • Anti-58P1D12 antibody formulations are administered via any route capable of delivering the antibodies to a tumor cell. Routes of administration include, but are not limited to, intravenous, intraperitoneal, intramuscular, intratumor, intradermal, and the like. Treatment generally involves repeated administration of the anti-58P1D12 antibody preparation, via an acceptable route of administration such as intravenous injection (IV), typically at a dose in the range of about 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, or 25 mg/kg body weight. In general, doses in the range of 10-1000 mg mAb per week are effective and well tolerated.
  • Based on clinical experience with the Herceptin™ mAb in the treatment of metastatic breast cancer, an initial loading dose of approximately 4 mg/kg patient body weight IV, followed by weekly doses of about 2 mg/kg IV of the anti-58P1D12 mAb preparation represents an acceptable dosing regimen. Preferably, the initial loading dose is administered as a 90-minute or longer infusion. The periodic maintenance dose is administered as a 30 minute or longer infusion, provided the initial dose was well tolerated. As appreciated by those of skill in the art, various factors can influence the ideal dose regimen in a particular case. Such factors include, for example, the binding affinity and half life of the Ab or mAbs used, the degree of 58P1D12 expression in the patient, the extent of circulating shed 58P1D12 antigen, the desired steady-state antibody concentration level, frequency of treatment, and the influence of chemotherapeutic or other agents used in combination with the treatment method of the invention, as well as the health status of a particular patient.
  • Optionally, patients should be evaluated for the levels of 58P1D12 in a given sample (e.g. the levels of circulating 58P1D12 antigen and/or 58P1D12 expressing cells) in order to assist in the determination of the most effective dosing regimen, etc. Such evaluations are also used for monitoring purposes throughout therapy, and are useful to gauge therapeutic success in combination with the evaluation of other parameters (for example, urine cytology and/or ImmunoCyt levels in bladder cancer therapy, or by analogy, serum PSA levels in prostate cancer therapy).
  • Anti-idiotypic anti-58P1D12 antibodies can also be used in anti-cancer therapy as a vaccine for inducing an immune response to cells expressing a 58P1D12-related protein. In particular, the generation of anti-idiotypic antibodies is well known in the art; this methodology can readily be adapted to generate anti-idiotypic anti-58P1D12 antibodies that mimic an epitope on a 58P1D12-related protein (see, for example, Wagner et al., 1997, Hybridoma 16: 33-40; Foon et al., 1995, J. Clin. Invest. 96:334-342; Herlyn et al., 1996, Cancer Immunol. Immunother. 43:65-76). Such an anti-idiotypic antibody can be used in cancer vaccine strategies.
  • X.C.) 58P1D12 as a Target for Cellular Immune Responses
  • Vaccines and methods of preparing vaccines that contain an immunogenically effective amount of one or more HLA-binding peptides as described herein are further embodiments of the invention. Furthermore, vaccines in accordance with the invention encompass compositions of one or more of the claimed peptides. A peptide can be present in a vaccine individually. Alternatively, the peptide can exist as a homopolymer comprising multiple copies of the same peptide, or as a heteropolymer of various peptides. Polymers have the advantage of increased immunological reaction and, where different peptide epitopes are used to make up the polymer, the additional ability to induce antibodies and/or CTLs that react with different antigenic determinants of the pathogenic organism or tumor-related peptide targeted for an immune response. The composition can be a naturally occurring region of an antigen or can be prepared, e.g., recombinantly or by chemical synthesis.
  • Carriers that can be used with vaccines of the invention are well known in the art, and include, e.g., thyroglobulin, albumins such as human serum albumin, tetanus toxoid, polyamino acids such as poly 1-lysine, poly 1-glutamic acid, influenza, hepatitis B virus core protein, and the like. The vaccines can contain a physiologically tolerable (i.e., acceptable) diluent such as water, or saline, preferably phosphate buffered saline. The vaccines also typically include an adjuvant. Adjuvants such as incomplete Freund's adjuvant, aluminum phosphate, aluminum hydroxide, or alum are examples of materials well known in the art. Additionally, as disclosed herein, CTL responses can be primed by conjugating peptides of the invention to lipids, such as tripalmitoyl-S-glycerylcysteinlyseryl-serine (P3CSS). Moreover, an adjuvant such as a synthetic cytosine-phosphorothiolated-guanine-containing (CpG) oligonucleotides has been found to increase CTL responses 10- to 100-fold. (see, e.g. Davila and Celis, J. Immunol. 165:539-547 (2000)).
  • Upon immunization with a peptide composition in accordance with the invention, via injection, aerosol, oral, transdermal, transmucosal, intrapleural, intrathecal, or other suitable routes, the immune system of the host responds to the vaccine by producing large amounts of CTLs and/or HTLs specific for the desired antigen. Consequently, the host becomes at least partially immune to later development of cells that express or overexpress 58P1D12 antigen, or derives at least some therapeutic benefit when the antigen was tumor-associated.
  • In some embodiments, it may be desirable to combine the class I peptide components with components that induce or facilitate neutralizing antibody and or helper T cell responses directed to the target antigen. A preferred embodiment of such a composition comprises class I and class II epitopes in accordance with the invention. An alternative embodiment of such a composition comprises a class I and/or class II epitope in accordance with the invention, along with a cross reactive HTL epitope such as PADRE™(Epimmune, San Diego, Calif.) molecule (described e.g., in U.S. Pat. No. 5,736,142).
  • A vaccine of the invention can also include antigen-presenting cells (APC), such as dendritic cells (DC), as a vehicle to present peptides of the invention. Vaccine compositions can be created in vitro, following dendritic cell mobilization and harvesting, whereby loading of dendritic cells occurs in vitro. For example, dendritic cells are transfected, e.g., with a minigene in accordance with the invention, or are pulsed with peptides. The dendritic cell can then be administered to a patient to elicit immune responses in vivo. Vaccine compositions, either DNA- or peptide-based, can also be administered in vivo in combination with dendritic cell mobilization whereby loading of dendritic cells occurs in vivo.
  • Preferably, the following principles are utilized when selecting an array of epitopes for inclusion in a polyepitopic composition for use in a vaccine, or for selecting discrete epitopes to be included in a vaccine and/or to be encoded by nucleic acids such as a minigene. It is preferred that each of the following principles be balanced in order to make the selection. The multiple epitopes to be incorporated in a given vaccine composition may be, but need not be, contiguous in sequence in the native antigen from which the epitopes are derived.
  • 1.) Epitopes are selected which, upon administration, mimic immune responses that have been observed to be correlated with tumor clearance. For HLA Class I this includes 3-4 epitopes that come from at least one tumor associated antigen (TAA). For HLA Class II a similar rationale is employed; again 3-4 epitopes are selected from at least one TAA (see, e.g., Rosenberg et al., Science 278:1447-1450). Epitopes from one TAA may be used in combination with epitopes from one or more additional TAAs to produce a vaccine that targets tumors with varying expression patterns of frequently-expressed TAAs.
  • 2.) Epitopes are selected that have the requisite binding affinity established to be correlated with immunogenicity: for HLA Class I an IC50 of 500 nM or less, often 200 nM or less; and for Class II an IC50 of 1000 nM or less.
  • 3.) Sufficient supermotif bearing-peptides, or a sufficient array of allele-specific motif-bearing peptides, are selected to give broad population coverage. For example, it is preferable to have at least 80% population coverage. A Monte Carlo analysis, a statistical evaluation known in the art, can be employed to assess the breadth, or redundancy of, population coverage.
  • 4.) When selecting epitopes from cancer-related antigens it is often useful to select analogs because the patient may have developed tolerance to the native epitope.
  • 5.) Of particular relevance are epitopes referred to as “nested epitopes.” Nested epitopes occur where at least two epitopes overlap in a given peptide sequence. A nested peptide sequence can comprise B cell, HLA class I and/or HLA class II epitopes. When providing nested epitopes, a general objective is to provide the greatest number of epitopes per sequence. Thus, an aspect is to avoid providing a peptide that is any longer than the amino terminus of the amino terminal epitope and the carboxyl terminus of the carboxyl terminal epitope in the peptide. When providing a multi-epitopic sequence, such as a sequence comprising nested epitopes, it is generally important to screen the sequence in order to insure that it does not have pathological or other deleterious biological properties.
  • 6.) If a polyepitopic protein is created, or when creating a minigene, an objective is to generate the smallest peptide that encompasses the epitopes of interest. This principle is similar, if not the same as that employed when selecting a peptide comprising nested epitopes. However, with an artificial polyepitopic peptide, the size minimization objective is balanced against the need to integrate any spacer sequences between epitopes in the polyepitopic protein. Spacer amino acid residues can, for example, be introduced to avoid junctional epitopes (an epitope recognized by the immune system, not present in the target antigen, and only created by the man-made juxtaposition of epitopes), or to facilitate cleavage between epitopes and thereby enhance epitope presentation. Junctional epitopes are generally to be avoided because the recipient may generate an immune response to that non-native epitope. Of particular concern is a junctional epitope that is a “dominant epitope.” A dominant epitope may lead to such a zealous response that immune responses to other epitopes are diminished or suppressed.
  • 7.) Where the sequences of multiple variants of the same target protein are present, potential peptide epitopes can also be selected on the basis of their conservancy. For example, a criterion for conservancy may define that the entire sequence of an HLA class I binding peptide or the entire 9-mer core of a class II binding peptide be conserved in a designated percentage of the sequences evaluated for a specific protein antigen.
  • X.C.1. Minigene Vaccines
  • A number of different approaches are available which allow simultaneous delivery of multiple epitopes. Nucleic acids encoding the peptides of the invention are a particularly useful embodiment of the invention. Epitopes for inclusion in a minigene are preferably selected according to the guidelines set forth in the previous section. A preferred means of administering nucleic acids encoding the peptides of the invention uses minigene constructs encoding a peptide comprising one or multiple epitopes of the invention.
  • The use of multi-epitope minigenes is described below and in, Ishioka et al., J. Immunol. 162:3915-3925, 1999; An, L. and Whitton, J. L., J. Virol. 71:2292, 1997; Thomson, S. A. et al., J. Immunol. 157:822, 1996; Whitton, J. L. et al., J. Virol. 67:348, 1993; Hanke, R. et al., Vaccine 16:426, 1998. For example, a multi-epitope DNA plasmid encoding supermotif- and/or motif-bearing epitopes derived 58P1D12, the PADRE® universal helper T cell epitope or multiple HTL epitopes from 58P1D12 (see e.g., Tables VIII-XXI and XXII to XLIX), and an endoplasmic reticulum-translocating signal sequence can be engineered. A vaccine may also comprise epitopes that are derived from other TAAs.
  • The immunogenicity of a multi-epitopic minigene can be confirmed in transgenic mice to evaluate the magnitude of CTL induction responses against the epitopes tested. Further, the immunogenicity of DNA-encoded epitopes in vivo can be correlated with the in vitro responses of specific CTL lines against target cells transfected with the DNA plasmid. Thus, these experiments can show that the minigene serves to both: 1.) generate a CTL response and 2.) that the induced CTLs recognized cells expressing the encoded epitopes.
  • For example, to create a DNA sequence encoding the selected epitopes (minigene) for expression in human cells, the amino acid sequences of the epitopes may be reverse translated. A human codon usage table can be used to guide the codon choice for each amino acid. These epitope-encoding DNA sequences may be directly adjoined, so that when translated, a continuous polypeptide sequence is created. To optimize expression and/or immunogenicity, additional elements can be incorporated into the minigene design. Examples of amino acid sequences that can be reverse translated and included in the minigene sequence include: HLA class I epitopes, HLA class II epitopes, antibody epitopes, a ubiquitination signal sequence, and/or an endoplasmic reticulum targeting signal. In addition, HLA presentation of CTL and HTL epitopes may be improved by including synthetic (e.g. poly-alanine) or naturally-occurring flanking sequences adjacent to the CTL or HTL epitopes; these larger peptides comprising the epitope(s) are within the scope of the invention.
  • The minigene sequence may be converted to DNA by assembling oligonucleotides that encode the plus and minus strands of the minigene. Overlapping oligonucleotides (30-100 bases long) may be synthesized, phosphorylated, purified and annealed under appropriate conditions using well known techniques. The ends of the oligonucleotides can be joined, for example, using T4 DNA ligase. This synthetic minigene, encoding the epitope polypeptide, can then be cloned into a desired expression vector.
  • Standard regulatory sequences well known to those of skill in the art are preferably included in the vector to ensure expression in the target cells. Several vector elements are desirable: a promoter with a down-stream cloning site for minigene insertion; a polyadenylation signal for efficient transcription termination; an E. coli origin of replication; and an E. coli selectable marker (e.g. ampicillin or kanamycin resistance). Numerous promoters can be used for this purpose, e.g., the human cytomegalovirus (hCMV) promoter. See, e.g., U.S. Pat. Nos. 5,580,859 and 5,589,466 for other suitable promoter sequences.
  • Additional vector modifications may be desired to optimize minigene expression and immunogenicity. In some cases, introns are required for efficient gene expression, and one or more synthetic or naturally-occurring introns could be incorporated into the transcribed region of the minigene. The inclusion of mRNA stabilization sequences and sequences for replication in mammalian cells may also be considered for increasing minigene expression.
  • Once an expression vector is selected, the minigene is cloned into the polylinker region downstream of the promoter. This plasmid is transformed into an appropriate E. coli strain, and DNA is prepared using standard techniques. The orientation and DNA sequence of the minigene, as well as all other elements included in the vector, are confirmed using restriction mapping and DNA sequence analysis. Bacterial cells harboring the correct plasmid can be stored as a master cell bank and a working cell bank.
  • In addition, immunostimulatory sequences (ISSs or CpGs) appear to play a role in the immunogenicity of DNA vaccines. These sequences may be included in the vector, outside the minigene coding sequence, if desired to enhance immunogenicity.
  • In some embodiments, a bi-cistronic expression vector which allows production of both the minigene-encoded epitopes and a second protein (included to enhance or decrease immunogenicity) can be used. Examples of proteins or polypeptides that could beneficially enhance the immune response if co-expressed include cytokines (e.g., IL-2, IL-12, GM-CSF), cytokine-inducing molecules (e.g., LeIF), costimulatory molecules, or for HTL responses, pan-DR binding proteins (PADRE™, Epimmune, San Diego, Calif.). Helper (HTL) epitopes can be joined to intracellular targeting signals and expressed separately from expressed CTL epitopes; this allows direction of the HTL epitopes to a cell compartment different than that of the CTL epitopes. If required, this could facilitate more efficient entry of HTL epitopes into the HLA class II pathway, thereby improving HTL induction. In contrast to HTL or CTL induction, specifically decreasing the immune response by co-expression of immunosuppressive molecules (e.g. TGF-β) may be beneficial in certain diseases.
  • Therapeutic quantities of plasmid DNA can be produced for example, by fermentation in E. coli, followed by purification. Aliquots from the working cell bank are used to inoculate growth medium, and grown to saturation in shaker flasks or a bioreactor according to well-known techniques. Plasmid DNA can be purified using standard bioseparation technologies such as solid phase anion-exchange resins supplied by QIAGEN, Inc. (Valencia, Calif.). If required, supercoiled DNA can be isolated from the open circular and linear forms using gel electrophoresis or other methods.
  • Purified plasmid DNA can be prepared for injection using a variety of formulations. The simplest of these is reconstitution of lyophilized DNA in sterile phosphate-buffer saline (PBS). This approach, known as “naked DNA,” is currently being used for intramuscular (IM) administration in clinical trials. To maximize the immunotherapeutic effects of minigene DNA vaccines, an alternative method for formulating purified plasmid DNA may be desirable. A variety of methods have been described, and new techniques may become available. Cationic lipids, glycolipids, and fusogenic liposomes can also be used in the formulation (see, e.g., as described by WO 93/24640; Mannino & Gould-Fogerite, BioTechniques 6(7): 682 (1988); U.S. Pat. No. 5,279,833; WO 91/06309; and Feigner, et al., Proc. Nat'l Acad. Sci. USA 84:7413 (1987). In addition, peptides and compounds referred to collectively as protective, interactive, non-condensing compounds (PINC) could also be complexed to purified plasmid DNA to influence variables such as stability, intramuscular dispersion, or trafficking to specific organs or cell types.
  • Target cell sensitization can be used as a functional assay for expression and HLA class I presentation of minigene-encoded CTL epitopes. For example, the plasmid DNA is introduced into a mammalian cell line that is suitable as a target for standard CTL chromium release assays. The transfection method used will be dependent on the final formulation. Electroporation can be used for “naked” DNA, whereas cationic lipids allow direct in vitro transfection. A plasmid expressing green fluorescent protein (GFP) can be co-transfected to allow enrichment of transfected cells using fluorescence activated cell sorting (FACS). These cells are then chromium-51 (51Cr) labeled and used as target cells for epitope-specific CTL lines; cytolysis, detected by 51Cr release, indicates both production of, and HLA presentation of, minigene-encoded CTL epitopes. Expression of HTL epitopes may be evaluated in an analogous manner using assays to assess HTL activity.
  • In vivo immunogenicity is a second approach for functional testing of minigene DNA formulations. Transgenic mice expressing appropriate human HLA proteins are immunized with the DNA product. The dose and route of administration are formulation dependent (e.g., IM for DNA in PBS, intraperitoneal (i.p.) for lipid-complexed DNA). Twenty-one days after immunization, splenocytes are harvested and restimulated for one week in the presence of peptides encoding each epitope being tested. Thereafter, for CTL effector cells, assays are conducted for cytolysis of peptide-loaded, 51Cr-labeled target cells using standard techniques. Lysis of target cells that were sensitized by HLA loaded with peptide epitopes, corresponding to minigene-encoded epitopes, demonstrates DNA vaccine function for in vivo induction of CTLs. Immunogenicity of HTL epitopes is confirmed in transgenic mice in an analogous manner.
  • Alternatively, the nucleic acids can be administered using ballistic delivery as described, for instance, in U.S. Pat. No. 5,204,253. Using this technique, particles comprised solely of DNA are administered. In a further alternative embodiment, DNA can be adhered to particles, such as gold particles.
  • Minigenes can also be delivered using other bacterial or viral delivery systems well known in the art, e.g., an expression construct encoding epitopes of the invention can be incorporated into a viral vector such as vaccinia.
  • X.C.2. Combinations of CTL Peptides with Helper Peptides
  • Vaccine compositions comprising CTL peptides of the invention can be modified, e.g., analoged, to provide desired attributes, such as improved serum half life, broadened population coverage or enhanced immunogenicity.
  • For instance, the ability of a peptide to induce CTL activity can be enhanced by linking the peptide to a sequence which contains at least one epitope that is capable of inducing a T helper cell response. Although a CTL peptide can be directly linked to a T helper peptide, often CTL epitope/HTL epitope conjugates are linked by a spacer molecule. The spacer is typically comprised of relatively small, neutral molecules, such as amino acids or amino acid mimetics, which are substantially uncharged under physiological conditions. The spacers are typically selected from, e.g., Ala, Gly, or other neutral spacers of nonpolar amino acids or neutral polar amino acids. It will be understood that the optionally present spacer need not be comprised of the same residues and thus may be a hetero- or homo-oligomer. When present, the spacer will usually be at least one or two residues, more usually three to six residues and sometimes 10 or more residues. The CTL peptide epitope can be linked to the T helper peptide epitope either directly or via a spacer either at the amino or carboxy terminus of the CTL peptide. The amino terminus of either the immunogenic peptide or the T helper peptide may be acylated.
  • In certain embodiments, the T helper peptide is one that is recognized by T helper cells present in a majority of a genetically diverse population. This can be accomplished by selecting peptides that bind to many, most, or all of the HLA class II molecules. Examples of such amino acid bind many HLA Class II molecules include sequences from antigens such as tetanus toxoid at positions 830-843 QYIKANSKFIGITE; (SEQ ID NO:18), Plasmodium falciparum circumsporozoite (CS) protein at positions 378-398 DIEKKIAKMEKASSVFNVVNS; (SEQ ID NO:19), and Streptococcus 18 kD protein at positions 116-131 GAVDSILGGVATYGAA; (SEQ ID NO:20). Other examples include peptides bearing a DR 1-4-7 supermotif, or either of the DR3 motifs.
  • Alternatively, it is possible to prepare synthetic peptides capable of stimulating T helper lymphocytes, in a loosely HLA-restricted fashion, using amino acid sequences not found in nature (see, e.g., PCT publication WO 95/07707). These synthetic compounds called Pan-DR-binding epitopes (e.g., PADRE™, Epimmune, Inc., San Diego, Calif.) are designed, most preferably, to bind most HLA-DR (human HLA class II) molecules. For instance, a pan-DR-binding epitope peptide having the formula: xKXVAAWTLKAAx (SEQ ID NO:21), where “X” is either cyclohexylalanine, phenylalanine, or tyrosine, and a is either d-alanine or 1-alanine, has been found to bind to most HLA-DR alleles, and to stimulate the response of T helper lymphocytes from most individuals, regardless of their HLA type. An alternative of a pan-DR binding epitope comprises all “L” natural amino acids and can be provided in the form of nucleic acids that encode the epitope.
  • HTL peptide epitopes can also be modified to alter their biological properties. For example, they can be modified to include d-amino acids to increase their resistance to proteases and thus extend their serum half life, or they can be conjugated to other molecules such as lipids, proteins, carbohydrates, and the like to increase their biological activity. For example, a T helper peptide can be conjugated to one or more palmitic acid chains at either the amino or carboxyl termini.
  • X.C.3. Combinations of CTL Peptides with T Cell Priming Agents
  • In some embodiments it may be desirable to include in the pharmaceutical compositions of the invention at least one component which primes B lymphocytes or T lymphocytes. Lipids have been identified as agents capable of priming CTL in vivo. For example, palmitic acid residues can be attached to the ε- and α-amino groups of a lysine residue and then linked, e.g., via one or more linking residues such as Gly, Gly-Gly-, Ser, Ser-Ser, or the like, to an immunogenic peptide. The lipidated peptide can then be administered either directly in a micelle or particle, incorporated into a liposome, or emulsified in an adjuvant, e.g., incomplete Freund's adjuvant. In a preferred embodiment, a particularly effective immunogenic composition comprises palmitic acid attached to ε- and α-amino groups of Lys, which is attached via linkage, e.g., Ser-Ser, to the amino terminus of the immunogenic peptide.
  • As another example of lipid priming of CTL responses, E. coli lipoproteins, such as tripalmitoyl-5-glycerylcysteinlyseryl-serine (P3CSS) can be used to prime virus specific CTL when covalently attached to an appropriate peptide (see, e.g., Deres, et al., Nature 342:561, 1989). Peptides of the invention can be coupled to P3CSS, for example, and the lipopeptide administered to an individual to prime specifically an immune response to the target antigen. Moreover, because the induction of neutralizing antibodies can also be primed with P3CSS-conjugated epitopes, two such compositions can be combined to more effectively elicit both humoral and cell-mediated responses.
  • X.C.4. Vaccine Compositions Comprising DC Pulsed with CTL and/or HTL Peptides
  • An embodiment of a vaccine composition in accordance with the invention comprises ex vivo administration of a cocktail of epitope-bearing peptides to PBMC, or isolated DC therefrom, from the patient's blood. A pharmaceutical to facilitate harvesting of DC can be used, such as Progenipoietin™ (Pharmacia-Monsanto, St. Louis, Mo.) or GM-CSF/IL-4. After pulsing the DC with peptides and prior to reinfusion into patients, the DC are washed to remove unbound peptides. In this embodiment, a vaccine comprises peptide-pulsed DCs which present the pulsed peptide epitopes complexed with HLA molecules on their surfaces.
  • The DC can be pulsed ex vivo with a cocktail of peptides, some of which stimulate CTL responses to 58P1D12. Optionally, a helper T cell (HTL) peptide, such as a natural or artificial loosely restricted HLA Class II peptide, can be included to facilitate the CTL response. Thus, a vaccine in accordance with the invention is used to treat a cancer which expresses or overexpresses 58P1D12.
  • X.D. Adoptive Immunotherapy
  • Antigenic 58P1D12-related peptides are used to elicit a CTL and/or HTL response ex vivo, as well. The resulting CTL or HTL cells, can be used to treat tumors in patients that do not respond to other conventional forms of therapy, or will not respond to a therapeutic vaccine peptide or nucleic acid in accordance with the invention. Ex vivo CTL or HTL responses to a particular antigen are induced by incubating in tissue culture the patient's, or genetically compatible, CTL or HTL precursor cells together with a source of antigen-presenting cells (APC), such as dendritic cells, and the appropriate immunogenic peptide. After an appropriate incubation time (typically about 7-28 days), in which the precursor cells are activated and expanded into effector cells, the cells are infused back into the patient, where they will destroy (CTL) or facilitate destruction (HTL) of their specific target cell (e.g., a tumor cell). Transfected dendritic cells may also be used as antigen presenting cells.
  • X.E. Administration of Vaccines for Therapeutic or Prophylactic Purposes
  • Pharmaceutical and vaccine compositions of the invention are typically used to treat and/or prevent a cancer that expresses or overexpresses 58P1D12. In therapeutic applications, peptide and/or nucleic acid compositions are administered to a patient in an amount sufficient to elicit an effective B cell, CTL and/or HTL response to the antigen and to cure or at least partially arrest or slow symptoms and/or complications. An amount adequate to accomplish this is defined as “therapeutically effective dose.” Amounts effective for this use will depend on, e.g., the particular composition administered, the manner of administration, the stage and severity of the disease being treated, the weight and general state of health of the patient, and the judgment of the prescribing physician.
  • For pharmaceutical compositions, the immunogenic peptides of the invention, or DNA encoding them, are generally administered to an individual already bearing a tumor that expresses 58P1D12. The peptides or DNA encoding them can be administered individually or as fusions of one or more peptide sequences. Patients can be treated with the immunogenic peptides separately or in conjunction with other treatments, such as surgery, as appropriate.
  • For therapeutic use, administration should generally begin at the first diagnosis of 58P1D12-associated cancer. This is followed by boosting doses until at least symptoms are substantially abated and for a period thereafter. The embodiment of the vaccine composition (i.e., including, but not limited to embodiments such as peptide cocktails, polyepitopic polypeptides, minigenes, or TAA-specific CTLs or pulsed dendritic cells) delivered to the patient may vary according to the stage of the disease or the patient's health status. For example, in a patient with a tumor that expresses 58P1D12, a vaccine comprising 58P1D12-specific CTL may be more efficacious in killing tumor cells in patient with advanced disease than alternative embodiments.
  • It is generally important to provide an amount of the peptide epitope delivered by a mode of administration sufficient to stimulate effectively a cytotoxic T cell response; compositions which stimulate helper T cell responses can also be given in accordance with this embodiment of the invention.
  • The dosage for an initial therapeutic immunization generally occurs in a unit dosage range where the lower value is about 1, 5, 50, 500, or 1,000 μg and the higher value is about 10,000; 20,000; 30,000; or 50,000 μg. Dosage values for a human typically range from about 500 μg to about 50,000 μg per 70 kilogram patient. Boosting dosages of between about 1.0 p.g to about 50,000 μg of peptide pursuant to a boosting regimen over weeks to months may be administered depending upon the patient's response and condition as determined by measuring the specific activity of CTL and HTL obtained from the patient's blood. Administration should continue until at least clinical symptoms or laboratory tests indicate that the neoplasia, has been eliminated or reduced and for a period thereafter. The dosages, routes of administration, and dose schedules are adjusted in accordance with methodologies known in the art.
  • In certain embodiments, the peptides and compositions of the present invention are employed in serious disease states, that is, life-threatening or potentially life threatening situations. In such cases, as a result of the minimal amounts of extraneous substances and the relative nontoxic nature of the peptides in preferred compositions of the invention, it is possible and may be felt desirable by the treating physician to administer substantial excesses of these peptide compositions relative to these stated dosage amounts.
  • The vaccine compositions of the invention can also be used purely as prophylactic agents. Generally the dosage for an initial prophylactic immunization generally occurs in a unit dosage range where the lower value is about 1, 5, 50, 500, or 1000 μg and the higher value is about 10,000; 20,000; 30,000; or 50,000 μg. Dosage values for a human typically range from about 500 μg to about 50,000 μg per 70 kilogram patient. This is followed by boosting dosages of between about 1.0 μg to about 50,000 μg of peptide administered at defined intervals from about four weeks to six months after the initial administration of vaccine. The immunogenicity of the vaccine can be assessed by measuring the specific activity of CTL and HTL obtained from a sample of the patient's blood.
  • The pharmaceutical compositions for therapeutic treatment are intended for parenteral, topical, oral, nasal, intrathecal, or local (e.g. as a cream or topical ointment) administration. Preferably, the pharmaceutical compositions are administered parentally, e.g., intravenously, subcutaneously, intradermally, or intramuscularly. Thus, the invention provides compositions for parenteral administration which comprise a solution of the immunogenic peptides dissolved or suspended in an acceptable carrier, preferably an aqueous carrier.
  • A variety of aqueous carriers may be used, e.g., water, buffered water, 0.8% saline, 0.3% glycine, hyaluronic acid and the like. These compositions may be sterilized by conventional, well-known sterilization techniques, or may be sterile filtered. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile solution prior to administration.
  • The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH-adjusting and buffering agents, tonicity adjusting agents, wetting agents, preservatives, and the like, for example, sodium acetate, sodium lactate, sodium chloride, potassium chloride, calcium chloride, sorbitan monolaurate, triethanolamine oleate, etc.
  • The concentration of peptides of the invention in the pharmaceutical formulations can vary widely, i.e., from less than about 0.1%, usually at or at least about 2% to as much as 20% to 50% or more by weight, and will be selected primarily by fluid volumes, viscosities, etc., in accordance with the particular mode of administration selected.
  • A human unit dose form of a composition is typically included in a pharmaceutical composition that comprises a human unit dose of an acceptable carrier, in one embodiment an aqueous carrier, and is administered in a volume/quantity that is known by those of skill in the art to be used for administration of such compositions to humans (see, e.g., Remington's Pharmaceutical Sciences, 17th Edition, A. Gennaro, Editor, Mack Publishing Co., Easton, Pa., 1985). For example a peptide dose for initial immunization can be from about 1 to about 50,000 μg, generally 100-5,000 μg, for a 70 kg patient. For example, for nucleic acids an initial immunization may be performed using an expression vector in the form of naked nucleic acid administered IM (or SC or ID) in the amounts of 0.5-5 mg at multiple sites. The nucleic acid (0.1 to 1000 μg) can also be administered using a gene gun. Following an incubation period of 3-4 weeks, a booster dose is then administered. The booster can be recombinant fowlpox virus administered at a dose of 5-107 to 5×109 pfu.
  • For antibodies, a treatment generally involves repeated administration of the anti-58P1D12 antibody preparation, via an acceptable route of administration such as intravenous injection (IV), typically at a dose in the range of about 0.1 to about 10 mg/kg body weight. In general, doses in the range of 10-500 mg mAb per week are effective and well tolerated. Moreover, an initial loading dose of approximately 4 mg/kg patient body weight IV, followed by weekly doses of about 2 mg/kg IV of the anti-58P1D12 mAb preparation represents an acceptable dosing regimen. As appreciated by those of skill in the art, various factors can influence the ideal dose in a particular case. Such factors include, for example, half life of a composition, the binding affinity of an Ab, the immunogenicity of a substance, the degree of 58P1D12 expression in the patient, the extent of circulating shed 58P1D12 antigen, the desired steady-state concentration level, frequency of treatment, and the influence of chemotherapeutic or other agents used in combination with the treatment method of the invention, as well as the health status of a particular patient. Non-limiting preferred human unit doses are, for example, 500 μg-1 mg, 1 mg-50 mg, 50 mg-100 mg, 100 mg-200 mg, 200 mg-300 mg, 400 mg-500 mg, 500 mg-600 mg, 600 mg-700 mg, 700 mg-800 mg, 800 mg-900 mg, 900 mg-1 g, or 1 mg-700 mg. In certain embodiments, the dose is in a range of 2-5 mg/kg body weight, e.g., with follow on weekly doses of 1-3 mg/kg; 0.5 mg, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 mg/kg body weight followed, e.g., in two, three or four weeks by weekly doses; 0.5-10 mg/kg body weight, e.g., followed in two, three or four weeks by weekly doses; 225, 250, 275, 300, 325, 350, 375, 400 mg m2 of body area weekly; 1-600 mg m2 of body area weekly; 225-400 mg m2 of body area weekly; these does can be followed by weekly doses for 2, 3, 4, 5, 6, 7, 8, 9, 19, 11, 12 or more weeks.
  • In one embodiment, human unit dose forms of polynucleotides comprise a suitable dosage range or effective amount that provides any therapeutic effect. As appreciated by one of ordinary skill in the art a therapeutic effect depends on a number of factors, including the sequence of the polynucleotide, molecular weight of the polynucleotide and route of administration. Dosages are generally selected by the physician or other health care professional in accordance with a variety of parameters known in the art, such as severity of symptoms, history of the patient and the like. Generally, for a polynucleotide of about 20 bases, a dosage range may be selected from, for example, an independently selected lower limit such as about 0.1, 0.25, 0.5, 1, 2, 5, 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400 or 500 mg/kg up to an independently selected upper limit, greater than the lower limit, of about 60, 80, 100, 200, 300, 400, 500, 750, 1000, 1500, 2000, 3000, 4000, 5000, 6000, 7000, 8000, 9000 or 10,000 mg/kg. For example, a dose may be about any of the following: 0.1 to 100 mg/kg, 0.1 to 50 mg/kg, 0.1 to 25 mg/kg, 0.1 to 10 mg/kg, 1 to 500 mg/kg, 100 to 400 mg/kg, 200 to 300 mg/kg, 1 to 100 mg/kg, 100 to 200 mg/kg, 300 to 400 mg/kg, 400 to 500 mg/kg, 500 to 1000 mg/kg, 500 to 5000 mg/kg, or 500 to 10,000 mg/kg. Generally, parenteral routes of administration may require higher doses of polynucleotide compared to more direct application to the nucleotide to diseased tissue, as do polynucleotides of increasing length.
  • In one embodiment, human unit dose forms of T-cells comprise a suitable dosage range or effective amount that provides any therapeutic effect. As appreciated by one of ordinary skill in the art, a therapeutic effect depends on a number of factors. Dosages are generally selected by the physician or other health care professional in accordance with a variety of parameters known in the art, such as severity of symptoms, history of the patient and the like. A dose may be about 104 cells to about 106 cells, about 106 cells to about 108 cells, about 108 to about 1011 cells, or about 108 to about 5×1010 cells. A dose may also about 106 cells/m2 to about 1010 cells/m2, or about 106 cells/m2 to about 108 cells/m2.
  • Proteins(s) of the invention, and/or nucleic acids encoding the protein(s), can also be administered via liposomes, which may also serve to: 1) target the proteins(s) to a particular tissue, such as lymphoid tissue; 2) to target selectively to diseases cells; or, 3) to increase the half-life of the peptide composition. Liposomes include emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like. In these preparations, the peptide to be delivered is incorporated as part of a liposome, alone or in conjunction with a molecule which binds to a receptor prevalent among lymphoid cells, such as monoclonal antibodies which bind to the CD45 antigen, or with other therapeutic or immunogenic compositions. Thus, liposomes either filled or decorated with a desired peptide of the invention can be directed to the site of lymphoid cells, where the liposomes then deliver the peptide compositions. Liposomes for use in accordance with the invention are formed from standard vesicle-forming lipids, which generally include neutral and negatively charged phospholipids and a sterol, such as cholesterol. The selection of lipids is generally guided by consideration of, e.g., liposome size, acid lability and stability of the liposomes in the blood stream. A variety of methods are available for preparing liposomes, as described in, e.g., Szoka, et al., Ann. Rev. Biophys. Bioeng. 9:467 (1980), and U.S. Pat. Nos. 4,235,871, 4,501,728, 4,837,028, and 5,019,369.
  • For targeting cells of the immune system, a ligand to be incorporated into the liposome can include, e.g., antibodies or fragments thereof specific for cell surface determinants of the desired immune system cells. A liposome suspension containing a peptide may be administered intravenously, locally, topically, etc. in a dose which varies according to, inter alia, the manner of administration, the peptide being delivered, and the stage of the disease being treated.
  • For solid compositions, conventional nontoxic solid carriers may be used which include, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, talcum, cellulose, glucose, sucrose, magnesium carbonate, and the like. For oral administration, a pharmaceutically acceptable nontoxic composition is formed by incorporating any of the normally employed excipients, such as those carriers previously listed, and generally 10-95% of active ingredient, that is, one or more peptides of the invention, and more preferably at a concentration of 25%-75%.
  • For aerosol administration, immunogenic peptides are preferably supplied in finely divided form along with a surfactant and propellant. Typical percentages of peptides are about 0.01%-20% by weight, preferably about 1%-10%. The surfactant must, of course, be nontoxic, and preferably soluble in the propellant. Representative of such agents are the esters or partial esters of fatty acids containing from about 6 to 22 carbon atoms, such as caproic, octanoic, lauric, palmitic, stearic, linoleic, linolenic, olesteric and oleic acids with an aliphatic polyhydric alcohol or its cyclic anhydride. Mixed esters, such as mixed or natural glycerides may be employed. The surfactant may constitute about 0.1%-20% by weight of the composition, preferably about 0.25-5%. The balance of the composition is ordinarily propellant. A carrier can also be included, as desired, as with, e.g., lecithin for intranasal delivery.
  • XI.) DIAGNOSTIC AND PROGNOSTIC EMBODIMENTS OF 58P1D12
  • As disclosed herein, 58P1D12 polynucleotides, polypeptides, reactive cytotoxic T cells (CTL), reactive helper T cells (HTL) and anti-polypeptide antibodies are used in well known diagnostic, prognostic and therapeutic assays that examine conditions associated with dysregulated cell growth such as cancer, in particular the cancers listed in Table I (see, e.g., both its specific pattern of tissue expression as well as its overexpression in certain cancers as described for example in the Example entitled “Expression analysis of 58P1D12 in normal tissues, and patient specimens”).
  • 58P1D12 can be analogized to a prostate associated antigen PSA, the archetypal marker that has been used by medical practitioners for years to identify and monitor the presence of prostate cancer (see, e.g., Merrill et al., J. Urol. 163(2): 503-5120 (2000); Polascik et al., J. Urol. August; 162(2):293-306 (1999) and Fortier et al., J. Nat. Cancer Inst. 91(19): 1635-1640 (1999)). A variety of other diagnostic markers are also used in similar contexts including p53 and K-ras (see, e.g., Tulchinsky et al., Int J Mol Med 1999 July 4(1):99-102 and Minimoto et al., Cancer Detect Prev 2000; 24(1):1-12). Therefore, this disclosure of 58P1D12 polynucleotides and polypeptides (as well as 58P1D12 polynucleotide probes and anti-58P1D12 antibodies used to identify the presence of these molecules) and their properties allows skilled artisans to utilize these molecules in methods that are analogous to those used, for example, in a variety of diagnostic assays directed to examining conditions associated with cancer.
  • Typical embodiments of diagnostic methods which utilize the 58P1D12 polynucleotides, polypeptides, reactive T cells and antibodies are analogous to those methods from well-established diagnostic assays, which employ, e.g., PSA polynucleotides, polypeptides, reactive T cells and antibodies. For example, just as PSA polynucleotides are used as probes (for example in Northern analysis, see, e.g., Sharief et al., Biochem. Mol. Biol. Int. 33(3):567-74 (1994)) and primers (for example in PCR analysis, see, e.g., Okegawa et al., J. Urol. 163(4): 1189-1190 (2000)) to observe the presence and/or the level of PSA mRNAs in methods of monitoring PSA overexpression or the metastasis of prostate cancers, the 58P1D12 polynucleotides described herein can be utilized in the same way to detect 58P1D12 overexpression or the metastasis of prostate and other cancers expressing this gene. Alternatively, just as PSA polypeptides are used to generate antibodies specific for PSA which can then be used to observe the presence and/or the level of PSA proteins in methods to monitor PSA protein overexpression (see, e.g., Stephan et al., Urology 55(4):560-3 (2000)) or the metastasis of prostate cells (see, e.g., Alanen et al., Pathol. Res. Pract. 192(3):233-7 (1996)), the 58P1D12 polypeptides described herein can be utilized to generate antibodies for use in detecting 58P1D12 overexpression or the metastasis of prostate cells and cells of other cancers expressing this gene.
  • Specifically, because metastases involves the movement of cancer cells from an organ of origin (such as the lung or prostate gland etc.) to a different area of the body (such as a lymph node), assays which examine a biological sample for the presence of cells expressing 58P1D12 polynucleotides and/or polypeptides can be used to provide evidence of metastasis. For example, when a biological sample from tissue that does not normally contain 58P1D12-expressing cells (lymph node) is found to contain 58P1D12-expressing cells such as the 58P1D12 expression seen in LAPC4 and LAPC9, xenografts isolated from lymph node and bone metastasis, respectively, this finding is indicative of metastasis.
  • Alternatively 58P1D12 polynucleotides and/or polypeptides can be used to provide evidence of cancer, for example, when cells in a biological sample that do not normally express 58P1D12 or express 58P1D12 at a different level are found to express 58P1D12 or have an increased expression of 58P1D12 (see, e.g., the 58P1D12 expression in the cancers listed in Table I and in patient samples etc. shown in the accompanying Figures). In such assays, artisans may further wish to generate supplementary evidence of metastasis by testing the biological sample for the presence of a second tissue restricted marker (in addition to 58P1D12) such as PSA, PSCA etc. (see, e.g., Alanen et al., Pathol. Res. Pract. 192(3): 233-237 (1996)).
  • The use of immunohistochemistry to identify the presence of a 58P1D12 polypeptide within a tissue section can indicate an altered state of certain cells within that tissue. It is well understood in the art that the ability of an antibody to localize to a polypeptide that is expressed in cancer cells is a way of diagnosing presence of disease, disease stage, progression and/or tumor aggressiveness. Such an antibody can also detect an altered distribution of the polypeptide within the cancer cells, as compared to corresponding non-malignant tissue.
  • The 58P1D12 polypeptide and immunogenic compositions are also useful in view of the phenomena of altered subcellular protein localization in disease states. Alteration of cells from normal to diseased state causes changes in cellular morphology and is often associated with changes in subcellular protein localization/distribution. For example, cell membrane proteins that are expressed in a polarized manner in normal cells can be altered in disease, resulting in distribution of the protein in a non-polar manner over the whole cell surface.
  • The phenomenon of altered subcellular protein localization in a disease state has been demonstrated with MUC1 and Her2 protein expression by use of immunohistochemical means. Normal epithelial cells have a typical apical distribution of MUC1, in addition to some supranuclear localization of the glycoprotein, whereas malignant lesions often demonstrate an apolar staining pattern (Diaz et al, The Breast Journal, 7; 40-45 (2001); Zhang et al, Clinical Cancer Research, 4; 2669-2676 (1998): Cao, et al, The Journal of Histochemistry and Cytochemistry, 45: 1547-1557 (1997)). In addition, normal breast epithelium is either negative for Her2 protein or exhibits only a basolateral distribution whereas malignant cells can express the protein over the whole cell surface (De Potter, et al, International Journal of Cancer, 44; 969-974 (1989): McCormick, et al, 117; 935-943 (2002)). Alternatively, distribution of the protein may be altered from a surface only localization to include diffuse cytoplasmic expression in the diseased state. Such an example can be seen with MUC1 (Diaz, et al, The Breast Journal, 7: 40-45 (2001)).
  • Alteration in the localization/distribution of a protein in the cell, as detected by immunohistochemical methods, can also provide valuable information concerning the favorability of certain treatment modalities. This last point is illustrated by a situation where a protein may be intracellular in normal tissue, but cell surface in malignant cells; the cell surface location makes the cells favorably amenable to antibody-based diagnostic and treatment regimens. When such an alteration of protein localization occurs for 58P1D12, the 58P1D12 protein and immune responses related thereto are very useful. Accordingly, the ability to determine whether alteration of subcellular protein localization occurred for 24P4C12 make the 58P1D12 protein and immune responses related thereto very useful. Use of the 58P1D12 compositions allows those skilled in the art to make important diagnostic and therapeutic decisions.
  • Immunohistochemical reagents specific to 58P1D12 are also useful to detect metastases of tumors expressing 58P1D12 when the polypeptide appears in tissues where 58P1D12 is not normally produced.
  • Thus, 58P1D12 polypeptides and antibodies resulting from immune responses thereto are useful in a variety of important contexts such as diagnostic, prognostic, preventative and/or therapeutic purposes known to those skilled in the art.
  • Just as PSA polynucleotide fragments and polynucleotide variants are employed by skilled artisans for use in methods of monitoring PSA, 58P1D12 polynucleotide fragments and polynucleotide variants are used in an analogous manner. In particular, typical PSA polynucleotides used in methods of monitoring PSA are probes or primers which consist of fragments of the PSA cDNA sequence. Illustrating this, primers used to PCR amplify a PSA polynucleotide must include less than the whole PSA sequence to function in the polymerase chain reaction. In the context of such PCR reactions, skilled artisans generally create a variety of different polynucleotide fragments that can be used as primers in order to amplify different portions of a polynucleotide of interest or to optimize amplification reactions (see, e.g., Caetano-Anolles, G. Biotechniques 25(3): 472-476, 478-480 (1998); Robertson et al., Methods Mol. Biol. 98:121-154 (1998)). An additional illustration of the use of such fragments is provided in the Example entitled “Expression analysis of 58P1D12 in normal tissues, and patient specimens,” where a 58P1D12 polynucleotide fragment is used as a probe to show the expression of 58P1D12 RNAs in cancer cells. In addition, variant polynucleotide sequences are typically used as primers and probes for the corresponding mRNAs in PCR and Northern analyses (see, e.g., Sawai et al., Fetal Diagn. Ther. 1996 November-December 11(6):407-13 and Current Protocols In Molecular Biology, Volume 2, Unit 2, Frederick M. Ausubel et al. eds., 1995)). Polynucleotide fragments and variants are useful in this context where they are capable of binding to a target polynucleotide sequence (e.g., a 58P1D12 polynucleotide shown in FIG. 2 or variant thereof) under conditions of high stringency.
  • Furthermore, PSA polypeptides which contain an epitope that can be recognized by an antibody or T cell that specifically binds to that epitope are used in methods of monitoring PSA. 58P1D12 polypeptide fragments and polypeptide analogs or variants can also be used in an analogous manner. This practice of using polypeptide fragments or polypeptide variants to generate antibodies (such as anti-PSA antibodies or T cells) is typical in the art with a wide variety of systems such as fusion proteins being used by practitioners (see, e.g., Current Protocols In Molecular Biology, Volume 2, Unit 16, Frederick M. Ausubel et al. eds., 1995). In this context, each epitope(s) functions to provide the architecture with which an antibody or T cell is reactive. Typically, skilled artisans create a variety of different polypeptide fragments that can be used in order to generate immune responses specific for different portions of a polypeptide of interest (see, e.g., U.S. Pat. No. 5,840,501 and U.S. Pat. No. 5,939,533). For example it may be preferable to utilize a polypeptide comprising one of the 58P1D12 biological motifs discussed herein or a motif-bearing subsequence which is readily identified by one of skill in the art based on motifs available in the art. Polypeptide fragments, variants or analogs are typically useful in this context as long as they comprise an epitope capable of generating an antibody or T cell specific for a target polypeptide sequence (e.g. a 58P1D12 polypeptide shown in FIG. 3).
  • As shown herein, the 58P1D12 polynucleotides and polypeptides (as well as the 58P1D12 polynucleotide probes and anti-58P1D12 antibodies or T cells used to identify the presence of these molecules) exhibit specific properties that make them useful in diagnosing cancers such as those listed in Table I. Diagnostic assays that measure the presence of 58P1D12 gene products, in order to evaluate the presence or onset of a disease condition described herein, such as prostate cancer, are used to identify patients for preventive measures or further monitoring, as has been done so successfully with PSA. Moreover, these materials satisfy a need in the art for molecules having similar or complementary characteristics to PSA in situations where, for example, a definite diagnosis of metastasis of prostatic origin cannot be made on the basis of a test for PSA alone (see, e.g., Alanen et al., Pathol. Res. Pract. 192(3): 233-237 (1996)), and consequently, materials such as 58P1D12 polynucleotides and polypeptides (as well as the 58P1D12 polynucleotide probes and anti-58P1D12 antibodies used to identify the presence of these molecules) need to be employed to confirm a metastases of prostatic origin.
  • Finally, in addition to their use in diagnostic assays, the 58P1D12 polynucleotides disclosed herein have a number of other utilities such as their use in the identification of oncogenetic associated chromosomal abnormalities in the chromosomal region to which the 58P1D12 gene maps (see the Example entitled “Chromosomal Mapping of 58P1D12” below). Moreover, in addition to their use in diagnostic assays, the 58P1D12-related proteins and polynucleotides disclosed herein have other utilities such as their use in the forensic analysis of tissues of unknown origin (see, e.g., Takahama K Forensic Sci Int 1996 Jun. 28; 80(1-2): 63-9).
  • Additionally, 58P1D12-related proteins or polynucleotides of the invention can be used to treat a pathologic condition characterized by the over-expression of 58P1D12. For example, the amino acid or nucleic acid sequence of FIG. 2 or FIG. 3, or fragments of either, can be used to generate an immune response to a 58P1D12 antigen. Antibodies or other molecules that react with 58P1D12 can be used to modulate the function of this molecule, and thereby provide a therapeutic benefit.
  • XII.) INHIBITION OF 58P1D12 PROTEIN FUNCTION
  • The invention includes various methods and compositions for inhibiting the binding of 58P1D12 to its binding partner or its association with other protein(s) as well as methods for inhibiting 58P1D12 function.
  • XII.A.) Inhibition of 58P1D12 with Intracellular Antibodies
  • In one approach, a recombinant vector that encodes single chain antibodies that specifically bind to 58P1D12 are introduced into 58P1D12 expressing cells via gene transfer technologies. Accordingly, the encoded single chain anti-58P1D12 antibody is expressed intracellularly, binds to 58P1D12 protein, and thereby inhibits its function. Methods for engineering such intracellular single chain antibodies are well known. Such intracellular antibodies, also known as “intrabodies”, are specifically targeted to a particular compartment within the cell, providing control over where the inhibitory activity of the treatment is focused. This technology has been successfully applied in the art (for review, see Richardson and Marasco, 1995, TIBTECH vol. 13). Intrabodies have been shown to virtually eliminate the expression of otherwise abundant cell surface receptors (see, e.g., Richardson et al., 1995, Proc. Natl. Acad. Sci. USA 92: 3137-3141; Beerli et al., 1994, J. Biol. Chem. 289: 23931-23936; Deshane et al., 1994, Gene Ther. 1: 332-337).
  • Single chain antibodies comprise the variable domains of the heavy and light chain joined by a flexible linker polypeptide, and are expressed as a single polypeptide. Optionally, single chain antibodies are expressed as a single chain variable region fragment joined to the light chain constant region. Well-known intracellular trafficking signals are engineered into recombinant polynucleotide vectors encoding such single chain antibodies in order to target precisely the intrabody to the desired intracellular compartment. For example, intrabodies targeted to the endoplasmic reticulum (ER) are engineered to incorporate a leader peptide and, optionally, a C-terminal ER retention signal, such as the KDEL amino acid motif. Intrabodies intended to exert activity in the nucleus are engineered to include a nuclear localization signal. Lipid moieties are joined to intrabodies in order to tether the intrabody to the cytosolic side of the plasma membrane. Intrabodies can also be targeted to exert function in the cytosol. For example, cytosolic intrabodies are used to sequester factors within the cytosol, thereby preventing them from being transported to their natural cellular destination.
  • In one embodiment, intrabodies are used to capture 58P1D12 in the nucleus, thereby preventing its activity within the nucleus. Nuclear targeting signals are engineered into such 58P1D12 intrabodies in order to achieve the desired targeting. Such 58P1D12 intrabodies are designed to bind specifically to a particular 58P1D12 domain. In another embodiment, cytosolic intrabodies that specifically bind to a 58P1D12 protein are used to prevent 58P1D12 from gaining access to the nucleus, thereby preventing it from exerting any biological activity within the nucleus (e.g., preventing 58P1D12 from forming transcription complexes with other factors).
  • In order to specifically direct the expression of such intrabodies to particular cells, the transcription of the intrabody is placed under the regulatory control of an appropriate tumor-specific promoter and/or enhancer. In order to target intrabody expression specifically to prostate, for example, the PSA promoter and/or promoter/enhancer can be utilized (See, for example, U.S. Pat. No. 5,919,652 issued 6 Jul. 1999).
  • XII.B.) Inhibition of 58P1D12 with Recombinant Proteins
  • In another approach, recombinant molecules bind to 58P1D12 and thereby inhibit 58P1D12 function. For example, these recombinant molecules prevent or inhibit 58P1D12 from accessing/binding to its binding partner(s) or associating with other protein(s). Such recombinant molecules can, for example, contain the reactive part(s) of a 58P1D12 specific antibody molecule. In a particular embodiment, the 58P1D12 binding domain of a 58P1D12 binding partner is engineered into a dimeric fusion protein, whereby the fusion protein comprises two 58P1D12 ligand binding domains linked to the Fc portion of a human IgG, such as human IgG1. Such IgG portion can contain, for example, the CH2 and CH3 domains and the hinge region, but not the CH1 domain. Such dimeric fusion proteins are administered in soluble form to patients suffering from a cancer associated with the expression of 58P1D12, whereby the dimeric fusion protein specifically binds to 58P1D12 and blocks 58P1D12 interaction with a binding partner. Such dimeric fusion proteins are further combined into multimeric proteins using known antibody linking technologies.
  • XII.C.) Inhibition of 58P1D12 Transcription or Translation
  • The present invention also comprises various methods and compositions for inhibiting the transcription of the 58P1D12 gene. Similarly, the invention also provides methods and compositions for inhibiting the translation of 58P1D12 mRNA into protein.
  • In one approach, a method of inhibiting the transcription of the 58P1D12 gene comprises contacting the 58P1D12 gene with a 58P1D12 antisense polynucleotide. In another approach, a method of inhibiting 58P1D12 mRNA translation comprises contacting a 58P1D12 mRNA with an antisense polynucleotide. In another approach, a 58P1D12 specific ribozyme is used to cleave a 58P1D12 message, thereby inhibiting translation. Such antisense and ribozyme based methods can also be directed to the regulatory regions of the 58P1D12 gene, such as 58P1D12 promoter and/or enhancer elements. Similarly, proteins capable of inhibiting a 58P1D12 gene transcription factor are used to inhibit 58P1D12 mRNA transcription. The various polynucleotides and compositions useful in the aforementioned methods have been described above. The use of antisense and ribozyme molecules to inhibit transcription and translation is well known in the art.
  • Other factors that inhibit the transcription of 58P1D12 by interfering with 58P1D12 transcriptional activation are also useful to treat cancers expressing 58P1D12. Similarly, factors that interfere with 58P1D12 processing are useful to treat cancers that express 58P1D12. Cancer treatment methods utilizing such factors are also within the scope of the invention.
  • XII.D.) General Considerations for Therapeutic Strategies
  • Gene transfer and gene therapy technologies can be used to deliver therapeutic polynucleotide molecules to tumor cells synthesizing 58P1D12 (i.e., antisense, ribozyme, polynucleotides encoding intrabodies and other 58P1D12 inhibitory molecules). A number of gene therapy approaches are known in the art. Recombinant vectors encoding 58P1D12 antisense polynucleotides, ribozymes, factors capable of interfering with 58P1D12 transcription, and so forth, can be delivered to target tumor cells using such gene therapy approaches.
  • The above therapeutic approaches can be combined with any one of a wide variety of surgical, chemotherapy or radiation therapy regimens. The therapeutic approaches of the invention can enable the use of reduced dosages of chemotherapy (or other therapies) and/or less frequent administration, an advantage for all patients and particularly for those that do not tolerate the toxicity of the chemotherapeutic agent well.
  • The anti-tumor activity of a particular composition (e.g., antisense, ribozyme, intrabody), or a combination of such compositions, can be evaluated using various in vitro and in vivo assay systems. In vitro assays that evaluate therapeutic activity include cell growth assays, soft agar assays and other assays indicative of tumor promoting activity, binding assays capable of determining the extent to which a therapeutic composition will inhibit the binding of 58P1D12 to a binding partner, etc.
  • In vivo, the effect of a 58P1D12 therapeutic composition can be evaluated in a suitable animal model. For example, xenogenic prostate cancer models can be used, wherein human prostate cancer explants or passaged xenograft tissues are introduced into immune compromised animals, such as nude or SCID mice (Klein et al., 1997, Nature Medicine 3: 402-408). For example, PCT Patent Application WO98/16628 and U.S. Pat. No. 6,107,540 describe various xenograft models of human prostate cancer capable of recapitulating the development of primary tumors, micrometastasis, and the formation of osteoblastic metastases characteristic of late stage disease. Efficacy can be predicted using assays that measure inhibition of tumor formation, tumor regression or metastasis, and the like.
  • In vivo assays that evaluate the promotion of apoptosis are useful in evaluating therapeutic compositions. In one embodiment, xenografts from tumor bearing mice treated with the therapeutic composition can be examined for the presence of apoptotic foci and compared to untreated control xenograft-bearing mice. The extent to which apoptotic foci are found in the tumors of the treated mice provides an indication of the therapeutic efficacy of the composition.
  • The therapeutic compositions used in the practice of the foregoing methods can be formulated into pharmaceutical compositions comprising a carrier suitable for the desired delivery method. Suitable carriers include any material that when combined with the therapeutic composition retains the anti-tumor function of the therapeutic composition and is generally non-reactive with the patient's immune system. Examples include, but are not limited to, any of a number of standard pharmaceutical carriers such as sterile phosphate buffered saline solutions, bacteriostatic water, and the like (see, generally, Remington's Pharmaceutical Sciences 16th Edition, A. Osal., Ed., 1980).
  • Therapeutic formulations can be solubilized and administered via any route capable of delivering the therapeutic composition to the tumor site. Potentially effective routes of administration include, but are not limited to, intravenous, parenteral, intraperitoneal, intramuscular, intratumor, intradermal, intraorgan, orthotopic, and the like. A preferred formulation for intravenous injection comprises the therapeutic composition in a solution of preserved bacteriostatic water, sterile unpreserved water, and/or diluted in polyvinylchloride or polyethylene bags containing 0.9% sterile Sodium Chloride for Injection, USP. Therapeutic protein preparations can be lyophilized and stored as sterile powders, preferably under vacuum, and then reconstituted in bacteriostatic water (containing for example, benzyl alcohol preservative) or in sterile water prior to injection.
  • Dosages and administration protocols for the treatment of cancers using the foregoing methods will vary with the method and the target cancer, and will generally depend on a number of other factors appreciated in the art.
  • XIII.) IDENTIFICATION, CHARACTERIZATION AND USE OF MODULATORS OF 58P1D12 Methods to Identify and Use Modulators
  • In one embodiment, screening is performed to identify modulators that induce or suppress a particular expression profile, suppress or induce specific pathways, preferably generating the associated phenotype thereby. In another embodiment, having identified differentially expressed genes important in a particular state; screens are performed to identify modulators that alter expression of individual genes, either increase or decrease. In another embodiment, screening is performed to identify modulators that alter a biological function of the expression product of a differentially expressed gene. Again, having identified the importance of a gene in a particular state, screens are performed to identify agents that bind and/or modulate the biological activity of the gene product.
  • In addition, screens are done for genes that are induced in response to a candidate agent. After identifying a modulator (one that suppresses a cancer expression pattern leading to a normal expression pattern, or a modulator of a cancer gene that leads to expression of the gene as in normal tissue) a screen is performed to identify genes that are specifically modulated in response to the agent. Comparing expression profiles between normal tissue and agent-treated cancer tissue reveals genes that are not expressed in normal tissue or cancer tissue, but are expressed in agent treated tissue, and vice versa. These agent-specific sequences are identified and used by methods described herein for cancer genes or proteins. In particular these sequences and the proteins they encode are used in marking or identifying agent-treated cells. In addition, antibodies are raised against the agent-induced proteins and used to target novel therapeutics to the treated cancer tissue sample.
  • Modulator-Related Identification and Screening Assays:
  • Gene Expression-Related Assays
  • Proteins, nucleic acids, and antibodies of the invention are used in screening assays. The cancer-associated proteins, antibodies, nucleic acids, modified proteins and cells containing these sequences are used in screening assays, such as evaluating the effect of drug candidates on a “gene expression profile,” expression profile of polypeptides or alteration of biological function. In one embodiment, the expression profiles are used, preferably in conjunction with high throughput screening techniques to allow monitoring for expression profile genes after treatment with a candidate agent (e.g., Davis, G F, et al, J Biol Screen 7:69 (2002); Zlokamik, et al., Science 279:84-8 (1998); Heid, Genome Res 6:986-94, 1996).
  • The cancer proteins, antibodies, nucleic acids, modified proteins and cells containing the native or modified cancer proteins or genes are used in screening assays. That is, the present invention comprises methods for screening for compositions which modulate the cancer phenotype or a physiological function of a cancer protein of the invention. This is done on a gene itself or by evaluating the effect of drug candidates on a “gene expression profile” or biological function. In one embodiment, expression profiles are used, preferably in conjunction with high throughput screening techniques to allow monitoring after treatment with a candidate agent, see Zlokarnik, supra.
  • A variety of assays are executed directed to the genes and proteins of the invention. Assays are run on an individual nucleic acid or protein level. That is, having identified a particular gene as up regulated in cancer, test compounds are screened for the ability to modulate gene expression or for binding to the cancer protein of the invention. “Modulation” in this context includes an increase or a decrease in gene expression. The preferred amount of modulation will depend on the original change of the gene expression in normal versus tissue undergoing cancer, with changes of at least 10%, preferably 50%, more preferably 100-300%, and in some embodiments 300-1000% or greater. Thus, if a gene exhibits a 4-fold increase in cancer tissue compared to normal tissue, a decrease of about four-fold is often desired; similarly, a 10-fold decrease in cancer tissue compared to normal tissue a target value of a 10-fold increase in expression by the test compound is often desired. Modulators that exacerbate the type of gene expression seen in cancer are also useful, e.g., as an upregulated target in further analyses.
  • The amount of gene expression is monitored using nucleic acid probes and the quantification of gene expression levels, or, alternatively, a gene product itself is monitored, e.g., through the use of antibodies to the cancer protein and standard immunoassays. Proteomics and separation techniques also allow for quantification of expression.
  • Expression Monitoring to Identify Compounds that Modify Gene Expression
  • In one embodiment, gene expression monitoring, i.e., an expression profile, is monitored simultaneously for a number of entities. Such profiles will typically involve one or more of the genes of FIG. 2. In this embodiment, e.g., cancer nucleic acid probes are attached to biochips to detect and quantify cancer sequences in a particular cell. Alternatively, PCR can be used. Thus, a series, e.g., wells of a microtiter plate, can be used with dispensed primers in desired wells. A PCR reaction can then be performed and analyzed for each well.
  • Expression monitoring is performed to identify compounds that modify the expression of one or more cancer-associated sequences, e.g., a polynucleotide sequence set out in FIG. 2. Generally, a test modulator is added to the cells prior to analysis. Moreover, screens are also provided to identify agents that modulate cancer, modulate cancer proteins of the invention, bind to a cancer protein of the invention, or interfere with the binding of a cancer protein of the invention and an antibody or other binding partner.
  • In one embodiment, high throughput screening methods involve providing a library containing a large number of potential therapeutic compounds (candidate compounds). Such “combinatorial chemical libraries” are then screened in one or more assays to identify those library members (particular chemical species or subclasses) that display a desired characteristic activity. The compounds thus identified can serve as conventional “lead compounds,” as compounds for screening, or as therapeutics.
  • In certain embodiments, combinatorial libraries of potential modulators are screened for an ability to bind to a cancer polypeptide or to modulate activity. Conventionally, new chemical entities with useful properties are generated by identifying a chemical compound (called a “lead compound”) with some desirable property or activity, e.g., inhibiting activity, creating variants of the lead compound, and evaluating the property and activity of those variant compounds. Often, high throughput screening (HTS) methods are employed for such an analysis.
  • As noted above, gene expression monitoring is conveniently used to test candidate modulators (e.g., protein, nucleic acid or small molecule). After the candidate agent has been added and the cells allowed to incubate for a period, the sample containing a target sequence to be analyzed is, e.g., added to a biochip.
  • If required, the target sequence is prepared using known techniques. For example, a sample is treated to lyse the cells, using known lysis buffers, electroporation, etc., with purification and/or amplification such as PCR performed as appropriate. For example, an in vitro transcription with labels covalently attached to the nucleotides is performed. Generally, the nucleic acids are labeled with biotin-FITC or PE, or with cy3 or cy5.
  • The target sequence can be labeled with, e.g., a fluorescent, a chemiluminescent, a chemical, or a radioactive signal, to provide a means of detecting the target sequence's specific binding to a probe. The label also can be an enzyme, such as alkaline phosphatase or horseradish peroxidase, which when provided with an appropriate substrate produces a product that is detected. Alternatively, the label is a labeled compound or small molecule, such as an enzyme inhibitor, that binds but is not catalyzed or altered by the enzyme. The label also can be a moiety or compound, such as, an epitope tag or biotin which specifically binds to streptavidin. For the example of biotin, the streptavidin is labeled as described above, thereby, providing a detectable signal for the bound target sequence. Unbound labeled streptavidin is typically removed prior to analysis.
  • As will be appreciated by those in the art, these assays can be direct hybridization assays or can comprise “sandwich assays”, which include the use of multiple probes, as is generally outlined in U.S. Pat. Nos. 5,681,702; 5,597,909; 5,545,730; 5,594,117; 5,591,584; 5,571,670; 5,580,731; 5,571,670; 5,591,584; 5,624,802; 5,635,352; 5,594,118; 5,359,100; 5,124, 246; and 5,681,697. In this embodiment, in general, the target nucleic acid is prepared as outlined above, and then added to the biochip comprising a plurality of nucleic acid probes, under conditions that allow the formation of a hybridization complex.
  • A variety of hybridization conditions are used in the present invention, including high, moderate and low stringency conditions as outlined above. The assays are generally run under stringency conditions which allow formation of the label probe hybridization complex only in the presence of target. Stringency can be controlled by altering a step parameter that is a thermodynamic variable, including, but not limited to, temperature, formamide concentration, salt concentration, chaotropic salt concentration pH, organic solvent concentration, etc. These parameters may also be used to control non-specific binding, as is generally outlined in U.S. Pat. No. 5,681,697. Thus, it can be desirable to perform certain steps at higher stringency conditions to reduce non-specific binding.
  • The reactions outlined herein can be accomplished in a variety of ways. Components of the reaction can be added simultaneously, or sequentially, in different orders, with preferred embodiments outlined below. In addition, the reaction may include a variety of other reagents. These include salts, buffers, neutral proteins, e.g. albumin, detergents, etc. which can be used to facilitate optimal hybridization and detection, and/or reduce nonspecific or background interactions. Reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., may also be used as appropriate, depending on the sample preparation methods and purity of the target. The assay data are analyzed to determine the expression levels of individual genes, and changes in expression levels as between states, forming a gene expression profile.
  • Biological Activity-Related Assays
  • The invention provides methods identify or screen for a compound that modulates the activity of a cancer-related gene or protein of the invention. The methods comprise adding a test compound, as defined above, to a cell comprising a cancer protein of the invention. The cells contain a recombinant nucleic acid that encodes a cancer protein of the invention. In another embodiment, a library of candidate agents is tested on a plurality of cells.
  • In one aspect, the assays are evaluated in the presence or absence or previous or subsequent exposure of physiological signals, e.g. hormones, antibodies, peptides, antigens, cytokines, growth factors, action potentials, pharmacological agents including chemotherapeutics, radiation, carcinogenics, or other cells (i.e., cell-cell contacts). In another example, the determinations are made at different stages of the cell cycle process. In this way, compounds that modulate genes or proteins of the invention are identified. Compounds with pharmacological activity are able to enhance or interfere with the activity of the cancer protein of the invention. Once identified, similar structures are evaluated to identify critical structural features of the compound.
  • In one embodiment, a method of modulating (e.g., inhibiting) cancer cell division is provided; the method comprises administration of a cancer modulator. In another embodiment, a method of modulating (e.g., inhibiting) cancer is provided; the method comprises administration of a cancer modulator. In a further embodiment, methods of treating cells or individuals with cancer are provided; the method comprises administration of a cancer modulator.
  • In one embodiment, a method for modulating the status of a cell that expresses a gene of the invention is provided. As used herein status comprises such art-accepted parameters such as growth, proliferation, survival, function, apoptosis, senescence, location, enzymatic activity, signal transduction, etc. of a cell. In one embodiment, a cancer inhibitor is an antibody as discussed above. In another embodiment, the cancer inhibitor is an antisense molecule. A variety of cell growth, proliferation, and metastasis assays are known to those of skill in the art, as described herein.
  • High Throughput Screening to Identify Modulators
  • The assays to identify suitable modulators are amenable to high throughput screening. Preferred assays thus detect enhancement or inhibition of cancer gene transcription, inhibition or enhancement of polypeptide expression, and inhibition or enhancement of polypeptide activity.
  • In one embodiment, modulators evaluated in high throughput screening methods are proteins, often naturally occurring proteins or fragments of naturally occurring proteins. Thus, e.g., cellular extracts containing proteins, or random or directed digests of proteinaceous cellular extracts, are used. In this way, libraries of proteins are made for screening in the methods of the invention. Particularly preferred in this embodiment are libraries of bacterial, fungal, viral, and mammalian proteins, with the latter being preferred, and human proteins being especially preferred. Particularly useful test compound will be directed to the class of proteins to which the target belongs, e.g., substrates for enzymes, or ligands and receptors.
  • Use of Soft Agar Growth and Colony Formation to Identify and Characterize Modulators
  • Normal cells require a solid substrate to attach and grow. When cells are transformed, they lose this phenotype and grow detached from the substrate. For example, transformed cells can grow in stirred suspension culture or suspended in semi-solid media, such as semi-solid or soft agar. The transformed cells, when transfected with tumor suppressor genes, can regenerate normal phenotype and once again require a solid substrate to attach to and grow. Soft agar growth or colony formation in assays are used to identify modulators of cancer sequences, which when expressed in host cells, inhibit abnormal cellular proliferation and transformation. A modulator reduces or eliminates the host cells' ability to grow suspended in solid or semisolid media, such as agar.
  • Techniques for soft agar growth or colony formation in suspension assays are described in Freshney, Culture of Animal Cells a Manual of Basic Technique (3rd ed., 1994). See also, the methods section of Garkavtsev et al. (1996), supra.
  • Evaluation of Contact Inhibition and Growth Density Limitation to Identify and Characterize Modulators
  • Normal cells typically grow in a flat and organized pattern in cell culture until they touch other cells. When the cells touch one another, they are contact inhibited and stop growing. Transformed cells, however, are not contact inhibited and continue to grow to high densities in disorganized foci. Thus, transformed cells grow to a higher saturation density than corresponding normal cells. This is detected morphologically by the formation of a disoriented monolayer of cells or cells in foci. Alternatively, labeling index with (3H)-thymidine at saturation density is used to measure density limitation of growth, similarly an MTT or Alamar blue assay will reveal proliferation capacity of cells and the ability of modulators to affect same. See Freshney (1994), supra. Transformed cells, when transfected with tumor suppressor genes, can regenerate a normal phenotype and become contact inhibited and would grow to a lower density.
  • In this assay, labeling index with 3H)-thymidine at saturation density is a preferred method of measuring density limitation of growth. Transformed host cells are transfected with a cancer-associated sequence and are grown for 24 hours at saturation density in non-limiting medium conditions. The percentage of cells labeling with (3H)-thymidine is determined by incorporated cpm.
  • Contact independent growth is used to identify modulators of cancer sequences, which had led to abnormal cellular proliferation and transformation. A modulator reduces or eliminates contact independent growth, and returns the cells to a normal phenotype.
  • Evaluation of Growth Factor or Serum Dependence to Identify and Characterize Modulators
  • Transformed cells have lower serum dependence than their normal counterparts (see, e.g., Temin, J. Natl. Cancer Inst. 37:167-175 (1966); Eagle et al., J. Exp. Med. 131:836-879 (1970)); Freshney, supra. This is in part due to release of various growth factors by the transformed cells. The degree of growth factor or serum dependence of transformed host cells can be compared with that of control. For example, growth factor or serum dependence of a cell is monitored in methods to identify and characterize compounds that modulate cancer-associated sequences of the invention.
  • Use of Tumor-specific Marker Levels to Identify and Characterize Modulators
  • Tumor cells release an increased amount of certain factors (hereinafter “tumor specific markers”) than their normal counterparts. For example, plasminogen activator (PA) is released from human glioma at a higher level than from normal brain cells (see, e.g., Gullino, Angiogenesis, Tumor Vascularization, and Potential Interference with Tumor Growth, in Biological Responses in Cancer, pp. 178-184 (Mihich (ed.) 1985)). Similarly, Tumor Angiogenesis Factor (TAF) is released at a higher level in tumor cells than their normal counterparts. See, e.g., Folkman, Angiogenesis and Cancer, Sem Cancer Biol. (1992)), while bFGF is released from endothelial tumors (Ensoli, B et al).
  • Various techniques which measure the release of these factors are described in Freshney (1994), supra. Also, see, Unkless et al., J. Biol. Chem. 249:4295-4305 (1974); Strickland & Beers, J. Biol. Chem. 251:5694-5702 (1976); Whur et al., Br. J. Cancer 42:305 312 (1980); Gullino, Angiogenesis, Tumor Vascularization, and Potential Interference with Tumor Growth, in Biological Responses in Cancer, pp. 178-184 (Mihich (ed.) 1985); Freshney, Anticancer Res. 5:111-130 (1985). For example, tumor specific marker levels are monitored in methods to identify and characterize compounds that modulate cancer-associated sequences of the invention.
  • Invasiveness into Matrigel to Identify and Characterize Modulators
  • The degree of invasiveness into Matrigel or an extracellular matrix constituent can be used as an assay to identify and characterize compounds that modulate cancer associated sequences. Tumor cells exhibit a positive correlation between malignancy and invasiveness of cells into Matrigel or some other extracellular matrix constituent. In this assay, tumorigenic cells are typically used as host cells. Expression of a tumor suppressor gene in these host cells would decrease invasiveness of the host cells. Techniques described in Cancer Res. 1999; 59:6010; Freshney (1994), supra, can be used. Briefly, the level of invasion of host cells is measured by using filters coated with Matrigel or some other extracellular matrix constituent. Penetration into the gel, or through to the distal side of the filter, is rated as invasiveness, and rated histologically by number of cells and distance moved, or by prelabeling the cells with 1251 and counting the radioactivity on the distal side of the filter or bottom of the dish. See, e.g., Freshney (1984), supra.
  • Evaluation of Tumor Growth In vivo to Identify and Characterize Modulators
  • Effects of cancer-associated sequences on cell growth are tested in transgenic or immune-suppressed organisms. Transgenic organisms are prepared in a variety of art-accepted ways. For example, knock-out transgenic organisms, e.g., mammals such as mice, are made, in which a cancer gene is disrupted or in which a cancer gene is inserted. Knock-out transgenic mice are made by insertion of a marker gene or other heterologous gene into the endogenous cancer gene site in the mouse genome via homologous recombination. Such mice can also be made by substituting the endogenous cancer gene with a mutated version of the cancer gene, or by mutating the endogenous cancer gene, e.g., by exposure to carcinogens.
  • To prepare transgenic chimeric animals, e.g., mice, a DNA construct is introduced into the nuclei of embryonic stem cells. Cells containing the newly engineered genetic lesion are injected into a host mouse embryo, which is re-implanted into a recipient female. Some of these embryos develop into chimeric mice that possess germ cells some of which are derived from the mutant cell line. Therefore, by breeding the chimeric mice it is possible to obtain a new line of mice containing the introduced genetic lesion (see, e.g., Capecchi et al., Science 244:1288 (1989)). Chimeric mice can be derived according to U.S. Pat. No. 6,365,797, issued 2 Apr. 2002; U.S. Pat. No. 6,107,540 issued 22 Aug. 2000; Hogan et al., Manipulating the Mouse Embryo: A laboratory Manual, Cold Spring Harbor Laboratory (1988) and Teratocarcinomas and Embryonic Stem Cells: A Practical Approach, Robertson, ed., IRL Press, Washington, D.C., (1987).
  • Alternatively, various immune-suppressed or immune-deficient host animals can be used. For example, a genetically athymic “nude” mouse (see, e.g., Giovanella et al., J. Natl. Cancer Inst. 52:921 (1974)), a SCID mouse, a thymectornized mouse, or an irradiated mouse (see, e.g., Bradley et al., Br. J. Cancer 38:263 (1978); Selby et al., Br. J. Cancer 41:52 (1980)) can be used as a host. Transplantable tumor cells (typically about 106 cells) injected into isogenic hosts produce invasive tumors in a high proportion of cases, while normal cells of similar origin will not. In hosts which developed invasive tumors, cells expressing cancer-associated sequences are injected subcutaneously or orthotopically. Mice are then separated into groups, including control groups and treated experimental groups) e.g. treated with a modulator). After a suitable length of time, preferably 4-8 weeks, tumor growth is measured (e.g., by volume or by its two largest dimensions, or weight) and compared to the control. Tumors that have statistically significant reduction (using, e.g., Student's T test) are said to have inhibited growth.
  • In Vitro Assays to Identify and Characterize Modulators
  • Assays to identify compounds with modulating activity can be performed in vitro. For example, a cancer polypeptide is first contacted with a potential modulator and incubated for a suitable amount of time, e.g., from 0.5 to 48 hours. In one embodiment, the cancer polypeptide levels are determined in vitro by measuring the level of protein or mRNA. The level of protein is measured using immunoassays such as Western blotting, ELISA and the like with an antibody that selectively binds to the cancer polypeptide or a fragment thereof. For measurement of mRNA, amplification, e.g., using PCR, LCR, or hybridization assays, e.g., Northern hybridization, RNAse protection, dot blotting, are preferred. The level of protein or mRNA is detected using directly or indirectly labeled detection agents, e.g., fluorescently or radioactively labeled nucleic acids, radioactively or enzymatically labeled antibodies, and the like, as described herein.
  • Alternatively, a reporter gene system can be devised using a cancer protein promoter operably linked to a reporter gene such as luciferase, green fluorescent protein, CAT, or P-gal. The reporter construct is typically transfected into a cell. After treatment with a potential modulator, the amount of reporter gene transcription, translation, or activity is measured according to standard techniques known to those of skill in the art (Davis GF, supra; Gonzalez, J. & Negulescu, P. Curr. Opin. Biotechnol. 1998: 9:624).
  • As outlined above, in vitro screens are done on individual genes and gene products. That is, having identified a particular differentially expressed gene as important in a particular state, screening of modulators of the expression of the gene or the gene product itself is performed.
  • In one embodiment, screening for modulators of expression of specific gene(s) is performed. Typically, the expression of only one or a few genes is evaluated. In another embodiment, screens are designed to first find compounds that bind to differentially expressed proteins. These compounds are then evaluated for the ability to modulate differentially expressed activity. Moreover, once initial candidate compounds are identified, variants can be further screened to better evaluate structure activity relationships.
  • Binding Assays to Identify and Characterize Modulators
  • In binding assays in accordance with the invention, a purified or isolated gene product of the invention is generally used. For example, antibodies are generated to a protein of the invention, and immunoassays are run to determine the amount and/or location of protein. Alternatively, cells comprising the cancer proteins are used in the assays.
  • Thus, the methods comprise combining a cancer protein of the invention and a candidate compound such as a ligand, and determining the binding of the compound to the cancer protein of the invention. Preferred embodiments utilize the human cancer protein; animal models of human disease of can also be developed and used. Also, other analogous mammalian proteins also can be used as appreciated by those of skill in the art. Moreover, in some embodiments variant or derivative cancer proteins are used.
  • Generally, the cancer protein of the invention, or the ligand, is non-diffusibly bound to an insoluble support. The support can, e.g., be one having isolated sample receiving areas (a microtiter plate, an array, etc.). The insoluble supports can be made of any composition to which the compositions can be bound, is readily separated from soluble material, and is otherwise compatible with the overall method of screening. The surface of such supports can be solid or porous and of any convenient shape.
  • Examples of suitable insoluble supports include microtiter plates, arrays, membranes and beads. These are typically made of glass, plastic (e.g., polystyrene), polysaccharide, nylon, nitrocellulose, or Teflon™, etc. Microtiter plates and arrays are especially convenient because a large number of assays can be carried out simultaneously, using small amounts of reagents and samples. The particular manner of binding of the composition to the support is not crucial so long as it is compatible with the reagents and overall methods of the invention, maintains the activity of the composition and is nondiffusable. Preferred methods of binding include the use of antibodies which do not sterically block either the ligand binding site or activation sequence when attaching the protein to the support, direct binding to “sticky” or ionic supports, chemical crosslinking, the synthesis of the protein or agent on the surface, etc. Following binding of the protein or ligand/binding agent to the support, excess unbound material is removed by washing. The sample receiving areas may then be blocked through incubation with bovine serum albumin (BSA), casein or other innocuous protein or other moiety.
  • Once a cancer protein of the invention is bound to the support, and a test compound is added to the assay. Alternatively, the candidate binding agent is bound to the support and the cancer protein of the invention is then added. Binding agents include specific antibodies, non-natural binding agents identified in screens of chemical libraries, peptide analogs, etc.
  • Of particular interest are assays to identify agents that have a low toxicity for human cells. A wide variety of assays can be used for this purpose, including proliferation assays, cAMP assays, labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays for protein binding, functional assays (phosphorylation assays, etc.) and the like.
  • A determination of binding of the test compound (ligand, binding agent, modulator, etc.) to a cancer protein of the invention can be done in a number of ways. The test compound can be labeled, and binding determined directly, e.g., by attaching all or a portion of the cancer protein of the invention to a solid support, adding a labeled candidate compound (e.g., a fluorescent label), washing off excess reagent, and determining whether the label is present on the solid support. Various blocking and washing steps can be utilized as appropriate.
  • In certain embodiments, only one of the components is labeled, e.g., a protein of the invention or ligands labeled. Alternatively, more than one component is labeled with different labels, e.g., I125, for the proteins and a fluorophor for the compound. Proximity reagents, e.g., quenching or energy transfer reagents are also useful.
  • Competitive Binding to Identify and Characterize Modulators
  • In one embodiment, the binding of the “test compound” is determined by competitive binding assay with a “competitor.” The competitor is a binding moiety that binds to the target molecule (e.g., a cancer protein of the invention). Competitors include compounds such as antibodies, peptides, binding partners, ligands, etc. Under certain circumstances, the competitive binding between the test compound and the competitor displaces the test compound. In one embodiment, the test compound is labeled. Either the test compound, the competitor, or both, is added to the protein for a time sufficient to allow binding. Incubations are performed at a temperature that facilitates optimal activity, typically between four and 40° C. Incubation periods are typically optimized, e.g., to facilitate rapid high throughput screening; typically between zero and one hour will be sufficient. Excess reagent is generally removed or washed away. The second component is then added, and the presence or absence of the labeled component is followed, to indicate binding.
  • In one embodiment, the competitor is added first, followed by the test compound. Displacement of the competitor is an indication that the test compound is binding to the cancer protein and thus is capable of binding to, and potentially modulating, the activity of the cancer protein. In this embodiment, either component can be labeled. Thus, e.g., if the competitor is labeled, the presence of label in the post-test compound wash solution indicates displacement by the test compound. Alternatively, if the test compound is labeled, the presence of the label on the support indicates displacement.
  • In an alternative embodiment, the test compound is added first, with incubation and washing, followed by the competitor. The absence of binding by the competitor indicates that the test compound binds to the cancer protein with higher affinity than the competitor. Thus, if the test compound is labeled, the presence of the label on the support, coupled with a lack of competitor binding, indicates that the test compound binds to and thus potentially modulates the cancer protein of the invention.
  • Accordingly, the competitive binding methods comprise differential screening to identity agents that are capable of modulating the activity of the cancer proteins of the invention. In this embodiment, the methods comprise combining a cancer protein and a competitor in a first sample. A second sample comprises a test compound, the cancer protein, and a competitor. The binding of the competitor is determined for both samples, and a change, or difference in binding between the two samples indicates the presence of an agent capable of binding to the cancer protein and potentially modulating its activity. That is, if the binding of the competitor is different in the second sample relative to the first sample, the agent is capable of binding to the cancer protein.
  • Alternatively, differential screening is used to identify drug candidates that bind to the native cancer protein, but cannot bind to modified cancer proteins. For example the structure of the cancer protein is modeled and used in rational drug design to synthesize agents that interact with that site, agents which generally do not bind to site-modified proteins. Moreover, such drug candidates that affect the activity of a native cancer protein are also identified by screening drugs for the ability to either enhance or reduce the activity of such proteins.
  • Positive controls and negative controls can be used in the assays. Preferably control and test samples are performed in at least triplicate to obtain statistically significant results. Incubation of all samples occurs for a time sufficient to allow for the binding of the agent to the protein. Following incubation, samples are washed free of non-specifically bound material and the amount of bound, generally labeled agent determined For example, where a radiolabel is employed, the samples can be counted in a scintillation counter to determine the amount of bound compound.
  • A variety of other reagents can be included in the screening assays. These include reagents like salts, neutral proteins, e.g. albumin, detergents, etc. which are used to facilitate optimal protein-protein binding and/or reduce non-specific or background interactions. Also reagents that otherwise improve the efficiency of the assay, such as protease inhibitors, nuclease inhibitors, anti-microbial agents, etc., can be used. The mixture of components is added in an order that provides for the requisite binding.
  • Use of Polynucleotides to Down-regulate or Inhibit a Protein of the Invention.
  • Polynucleotide modulators of cancer can be introduced into a cell containing the target nucleotide sequence by formation of a conjugate with a ligand-binding molecule, as described in WO 91/04753. Suitable ligand-binding molecules include, but are not limited to, cell surface receptors, growth factors, other cytokines, or other ligands that bind to cell surface receptors. Preferably, conjugation of the ligand binding molecule does not substantially interfere with the ability of the ligand binding molecule to bind to its corresponding molecule or receptor, or block entry of the sense or antisense oligonucleotide or its conjugated version into the cell. Alternatively, a polynucleotide modulator of cancer can be introduced into a cell containing the target nucleic acid sequence, e.g., by formation of a polynucleotide-lipid complex, as described in WO 90/10448. It is understood that the use of antisense molecules or knock out and knock in models may also be used in screening assays as discussed above, in addition to methods of treatment.
  • Inhibitory and Antisense Nucleotides
  • In certain embodiments, the activity of a cancer-associated protein is down-regulated, or entirely inhibited, by the use of antisense polynucleotide or inhibitory small nuclear RNA (snRNA), i.e., a nucleic acid complementary to, and which can preferably hybridize specifically to, a coding mRNA nucleic acid sequence, e.g., a cancer protein of the invention, mRNA, or a subsequence thereof. Binding of the antisense polynucleotide to the mRNA reduces the translation and/or stability of the mRNA.
  • In the context of this invention, antisense polynucleotides can comprise naturally occurring nucleotides, or synthetic species formed from naturally occurring subunits or their close homologs. Antisense polynucleotides may also have altered sugar moieties or inter-sugar linkages. Exemplary among these are the phosphorothioate and other sulfur containing species which are known for use in the art. Analogs are comprised by this invention so long as they function effectively to hybridize with nucleotides of the invention. See, e.g., Isis Pharmaceuticals, Carlsbad, Calif.; Sequitor, Inc., Natick, Mass.
  • Such antisense polynucleotides can readily be synthesized using recombinant means, or can be synthesized in vitro. Equipment for such synthesis is sold by several vendors, including Applied Biosystems. The preparation of other oligonucleotides such as phosphorothioates and alkylated derivatives is also well known to those of skill in the art.
  • Antisense molecules as used herein include antisense or sense oligonucleotides. Sense oligonucleotides can, e.g., be employed to block transcription by binding to the anti-sense strand. The antisense and sense oligonucleotide comprise a single stranded nucleic acid sequence (either RNA or DNA) capable of binding to target mRNA (sense) or DNA (antisense) sequences for cancer molecules. Antisense or sense oligonucleotides, according to the present invention, comprise a fragment generally at least about 12 nucleotides, preferably from about 12 to 30 nucleotides. The ability to derive an antisense or a sense oligonucleotide, based upon a cDNA sequence encoding a given protein is described in, e.g., Stein &Cohen (Cancer Res. 48:2659 (1988 and van der Krol et al. (BioTechniques 6:958 (1988)).
  • Ribozymes
  • In addition to antisense polynucleotides, ribozymes can be used to target and inhibit transcription of cancer-associated nucleotide sequences. A ribozyme is an RNA molecule that catalytically cleaves other RNA molecules. Different kinds of ribozymes have been described, including group I ribozymes, hammerhead ribozymes, hairpin ribozymes, RNase P, and axhead ribozymes (see, e.g., Castanotto et al., Adv. in Pharmacology 25: 289-317 (1994) for a general review of the properties of different ribozymes).
  • The general features of hairpin ribozymes are described, e.g., in Hampel et al., Nucl. Acids Res. 18:299-304 (1990); European Patent Publication No. 0360257; U.S. Pat. No. 5,254,678. Methods of preparing are well known to those of skill in the art (see, e.g., WO 94/26877; Ojwang et al., Proc. Natl. Acad. Sci. USA 90:6340-6344 (1993); Yamada et al., Human Gene Therapy 1:39-45 (1994); Leavitt et al., Proc. Natl. Acad. Sci. USA 92:699-703 (1995); Leavitt et al., Human Gene Therapy 5: 1151-120 (1994); and Yamada et al., Virology 205: 121-126 (1994)).
  • Use of Modulators in Phenotypic Screening
  • In one embodiment, a test compound is administered to a population of cancer cells, which have an associated cancer expression profile. By “administration” or “contacting” herein is meant that the modulator is added to the cells in such a manner as to allow the modulator to act upon the cell, whether by uptake and intracellular action, or by action at the cell surface. In some embodiments, a nucleic acid encoding a proteinaceous agent (i.e., a peptide) is put into a viral construct such as an adenoviral or retroviral construct, and added to the cell, such that expression of the peptide agent is accomplished, e.g., PCT US97/01019. Regulatable gene therapy systems can also be used. Once the modulator has been administered to the cells, the cells are washed if desired and are allowed to incubate under preferably physiological conditions for some period. The cells are then harvested and a new gene expression profile is generated. Thus, e.g., cancer tissue is screened for agents that modulate, e.g., induce or suppress, the cancer phenotype. A change in at least one gene, preferably many, of the expression profile indicates that the agent has an effect on cancer activity. Similarly, altering a biological function or a signaling pathway is indicative of modulator activity. By defining such a signature for the cancer phenotype, screens for new drugs that alter the phenotype are devised. With this approach, the drug target need not be known and need not be represented in the original gene/protein expression screening platform, nor does the level of transcript for the target protein need to change. The modulator inhibiting function will serve as a surrogate marker.
  • As outlined above, screens are done to assess genes or gene products. That is, having identified a particular differentially expressed gene as important in a particular state, screening of modulators of either the expression of the gene or the gene product itself is performed.
  • Use of Modulators to Affect Peptides of the Invention
  • Measurements of cancer polypeptide activity, or of the cancer phenotype are performed using a variety of assays. For example, the effects of modulators upon the function of a cancer polypeptide(s) are measured by examining parameters described above. A physiological change that affects activity is used to assess the influence of a test compound on the polypeptides of this invention. When the functional outcomes are determined using intact cells or animals, a variety of effects can be assesses such as, in the case of a cancer associated with solid tumors, tumor growth, tumor metastasis, neovascularization, hormone release, transcriptional changes to both known and uncharacterized genetic markers (e.g., by Northern blots), changes in cell metabolism such as cell growth or pH changes, and changes in intracellular second messengers such as cGNIP.
  • Methods of Identifying Characterizing Cancer-Associated Sequences
  • Expression of various gene sequences is correlated with cancer. Accordingly, disorders based on mutant or variant cancer genes are determined. In one embodiment, the invention provides methods for identifying cells containing variant cancer genes, e.g., determining the presence of, all or part, the sequence of at least one endogenous cancer gene in a cell. This is accomplished using any number of sequencing techniques. The invention comprises methods of identifying the cancer genotype of an individual, e.g., determining all or part of the sequence of at least one gene of the invention in the individual. This is generally done in at least one tissue of the individual, e.g., a tissue set forth in Table I, and may include the evaluation of a number of tissues or different samples of the same tissue. The method may include comparing the sequence of the sequenced gene to a known cancer gene, i.e., a wild-type gene to determine the presence of family members, homologies, mutations or variants. The sequence of all or part of the gene can then be compared to the sequence of a known cancer gene to determine if any differences exist. This is done using any number of known homology programs, such as BLAST, Bestfit, etc. The presence of a difference in the sequence between the cancer gene of the patient and the known cancer gene correlates with a disease state or a propensity for a disease state, as outlined herein.
  • In a preferred embodiment, the cancer genes are used as probes to determine the number of copies of the cancer gene in the genome. The cancer genes are used as probes to determine the chromosomal localization of the cancer genes. Information such as chromosomal localization finds use in providing a diagnosis or prognosis in particular when chromosomal abnormalities such as translocations, and the like are identified in the cancer gene locus.
  • XIV.) KITS/ARTICLES OF MANUFACTURE
  • For use in the laboratory, prognostic, prophylactic, diagnostic and therapeutic applications described herein, kits are within the scope of the invention. Such kits can comprise a carrier, package, or container that is compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) comprising one of the separate elements to be used in the method, along with a label or insert comprising instructions for use, such as a use described herein. For example, the container(s) can comprise a probe that is or can be detectably labeled. Such probe can be an antibody or polynucleotide specific for a protein or a gene or message of the invention, respectively. Where the method utilizes nucleic acid hybridization to detect the target nucleic acid, the kit can also have containers containing nucleotide(s) for amplification of the target nucleic acid sequence. Kits can comprise a container comprising a reporter, such as a biotin-binding protein, such as avidin or streptavidin, bound to a reporter molecule, such as an enzymatic, fluorescent, or radioisotope label; such a reporter can be used with, e.g., a nucleic acid or antibody. The kit can include all or part of the amino acid sequences in FIG. 2 or FIG. 3 or analogs thereof, or a nucleic acid molecule that encodes such amino acid sequences.
  • The kit of the invention will typically comprise the container described above and one or more other containers associated therewith that comprise materials desirable from a commercial and user standpoint, including buffers, diluents, filters, needles, syringes; carrier, package, container, vial and/or tube labels listing contents and/or instructions for use, and package inserts with instructions for use.
  • A label can be present on or with the container to indicate that the composition is used for a specific therapy or non-therapeutic application, such as a prognostic, prophylactic, diagnostic or laboratory application, and can also indicate directions for either in vivo or in vitro use, such as those described herein. Directions and or other information can also be included on an insert(s) or label(s) which is included with or on the kit. The label can be on or associated with the container. A label a can be on a container when letters, numbers or other characters forming the label are molded or etched into the container itself; a label can be associated with a container when it is present within a receptacle or carrier that also holds the container, e.g., as a package insert. The label can indicate that the composition is used for diagnosing, treating, prophylaxing or prognosing a condition, such as a neoplasia of a tissue set forth in Table I.
  • The terms “kit” and “article of manufacture” can be used as synonyms.
  • In another embodiment of the invention, an article(s) of manufacture containing compositions, such as amino acid sequence(s), small molecule(s), nucleic acid sequence(s), and/or antibody(s), e.g., materials useful for the diagnosis, prognosis, prophylaxis and/or treatment of neoplasias of tissues such as those set forth in Table I is provided. The article of manufacture typically comprises at least one container and at least one label. Suitable containers include, for example, bottles, vials, syringes, and test tubes. The containers can be formed from a variety of materials such as glass, metal or plastic. The container can hold amino acid sequence(s), small molecule(s), nucleic acid sequence(s), cell population(s) and/or antibody(s). In one embodiment, the container holds a polynucleotide for use in examining the mRNA expression profile of a cell, together with reagents used for this purpose. In another embodiment a container comprises an antibody, binding fragment thereof or specific binding protein for use in evaluating protein expression of 58P1D12 in cells and tissues, or for relevant laboratory, prognostic, diagnostic, prophylactic and therapeutic purposes; indications and/or directions for such uses can be included on or with such container, as can reagents and other compositions or tools used for these purposes. In another embodiment, a container comprises materials for eliciting a cellular or humoral immune response, together with associated indications and/or directions. In another embodiment, a container comprises materials for adoptive immunotherapy, such as cytotoxic T cells (CTL) or helper T cells (HTL), together with associated indications and/or directions; reagents and other compositions or tools used for such purpose can also be included.
  • The container can alternatively hold a composition that is effective for treating, diagnosis, prognosing or prophylaxing a condition and can have a sterile access port (for example the container can be an intravenous solution bag or a vial having a stopper pierceable by a hypodermic injection needle). The active agents in the composition can be an antibody capable of specifically binding 58P1D12 and modulating the function of 58P1D12.
  • The article of manufacture can further comprise a second container comprising a pharmaceutically-acceptable buffer, such as phosphate-buffered saline, Ringer's solution and/or dextrose solution. It can further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, stirrers, needles, syringes, and/or package inserts with indications and/or instructions for use.
  • EXAMPLES
  • Various aspects of the invention are further described and illustrated by way of the several examples that follow, none of which is intended to limit the scope of the invention.
  • Example 1 SSH-Generated Isolation of a cDNA Fragment of the 58P1D12 Gene
  • To isolate genes that are over-expressed in prostate cancer we used the Suppression Subtractive Hybridization (SSH) procedure using cDNA derived from prostate cancer tissues. The 58P1D12 SSH cDNA sequence was identified from an SSH experiment consisting of LAPC-9AD cDNA minus LAPC-9AI cDNA.
  • Materials and Methods:
  • Human Tissues:
  • The patient cancer and normal tissues were purchased from different sources such as the NDR1 (Philadelphia, Pa.). mRNA for some normal tissues were purchased from different companies such as Clontech, Palo Alto, Calif.
  • RNA Isolation:
  • Tissues were homogenized in Trizol reagent (Life Technologies, Gibco BRL) using 10 ml/g tissue to isolate total RNA. Poly A RNA was purified from total RNA using Qiagen's Oligotex mRNA Mini and Midi kits. Total and mRNA were quantified by spectrophotometric analysis (O.D. 260/280 nm) and analyzed by gel electrophoresis.
  • Oligonucleotides:
  • The following HPLC purified oligonucleotides were used:
  • DPNCDN (cDNA synthesis primer):
    (SEQ ID NO: 22)
    5′TTTTGATCAAGCTT 303′
    Adaptor 1:
    (SEQ ID NO: 23)
    5′CTAATACGACTCACTATAGGGCTCGAGCGGCCGCCCGGGCAG3′
    (SEQ ID NO: 24)
    3′GGCCCGTCCTAG5′
    Adaptor 2:
    (SEQ ID NO: 25)
    5′GTAATACGACTCACTATAGGGCAGCGTGGTCGCGGCCGAG3′
    (SEQ ID NO: 26)
    3′CGGCTCCTAG5′
    PCR primer 1:
    (SEQ ID NO: 27)
    5′CTAATACGACTCACTATAGGGC3′
    Nested primer (NP)1:
    (SEQ ID NO: 28)
    5′TCGAGCGGCCGCCCGGGCAGGA3′
    Nested primer (NP)2:
    (SEQ ID NO: 29)
    5′AGCGTGGTCGCGGCCGAGGA3′
  • Suppression Subtractive Hybridization:
  • Suppression Subtractive Hybridization (SSH) was used to identify cDNAs corresponding to genes that may be differentially expressed in prostate cancer. The SSH reaction utilized cDNA from LAPC-9AD and LAPC-9AI prostate cancer xenograft tissues.
  • The gene 58P1D12 sequence was derived from the androgen dependent xenograft LAPC-9AD minus the androgen independent xenograft LAPC-9AI cDNA subtraction. The SSH DNA sequence 58P1D12 (FIG. 1) was identified.
  • The cDNA derived from LAPC-9AI was used as the source of the “driver” cDNA, while the cDNA from LAPC-9AD was used as the source of the “tester” cDNA. Double stranded cDNAs corresponding to tester and driver cDNAs were synthesized from 2 μg of poly(A)+ RNA isolated from the relevant xenograft tissue, as described above, using CLONTECH's PCR-Select cDNA Subtraction Kit and 1 ng of oligonucleotide DPNCDN as primer. First- and second-strand synthesis were carried out as described in the Kit's user manual protocol (CLONTECH Protocol No. PT1117-1, Catalog No. K1804-1). The resulting cDNA was digested with Dpn II for 3 hrs at 37° C. Digested cDNA was extracted with phenol/chloroform (1:1) and ethanol precipitated.
  • Tester cDNA was generated by diluting 1 μl of Dpn II digested cDNA from the relevant tissue source (see above) (400 ng) in 5 μl of water. The diluted cDNA (2 μl, 160 ng) was then ligated to 2 μl of Adaptor 1 and Adaptor 2 (10 μM), in separate ligation reactions, in a total volume of 10 μl at 16° C. overnight, using 400 μl of T4 DNA ligase (CLONTECH). Ligation was terminated with 1 μl of 0.2 M EDTA and heating at 72° C. for 5 min
  • The first hybridization was performed by adding 1.5 μl (600 ng) of driver cDNA to each of two tubes containing 1.5 μl (20 ng) Adaptor 1- and Adaptor 2-ligated tester cDNA. In a final volume of 4 μl, the samples were overlaid with mineral oil, denatured in an MJ Research thermal cycler at 98° C. for 1.5 minutes, and then were allowed to hybridize for 8 hrs at 68° C. The two hybridizations were then mixed together with an additional 1 μl of fresh denatured driver cDNA and were allowed to hybridize overnight at 68° C. The second hybridization was then diluted in 200 μl of 20 mM Hepes, pH 8.3, 50 mM NaCl, 0.2 mM EDTA, heated at 70° C. for 7 min. and stored at −20° C.
  • PCR Amplification, Cloning and Sequencing of Gene Fragments Generated from SSH: To amplify gene fragments resulting from SSH reactions, two PCR amplifications were performed. In the primary PCR reaction 1 μl of the diluted final hybridization mix was added to 1 μl of PCR primer 1 (10 μM), 0.5 μl dNTP mix (10 μM), 2.5 μl 10× reaction buffer (CLONTECH) and 0.5 μl 50× Advantage cDNA polymerase Mix (CLONTECH) in a final volume of 25 μl. PCR 1 was conducted using the following conditions: 75° C. for 5 min., 94° C. for 25 sec., then 27 cycles of 94° C. for 10 sec, 66° C. for 30 sec, 72° C. for 1.5 min. Five separate primary PCR reactions were performed for each experiment. The products were pooled and diluted 1:10 with water. For the secondary PCR reaction, 1 μl from the pooled and diluted primary PCR reaction was added to the same reaction mix as used for PCR 1, except that primers NP1 and NP2 (10 μM) were used instead of PCR primer 1. PCR 2 was performed using 10-12 cycles of 94° C. for 10 sec, 68° C. for 30 sec, and 72° C. for 1.5 minutes. The PCR products were analyzed using 2% agarose gel electrophoresis.
  • The PCR products were inserted into pCR2.1 using the T/A vector cloning kit (Invitrogen). Transformed E. coli were subjected to blue/white and ampicillin selection. White colonies were picked and arrayed into 96 well plates and were grown in liquid culture overnight. To identify inserts, PCR amplification was performed on 1 μl of bacterial culture using the conditions of PCR1 and NP1 and NP2 as primers. PCR products were analyzed using 2% agarose gel electrophoresis.
  • Bacterial clones were stored in 20% glycerol in a 96 well format. Plasmid DNA was prepared, sequenced, and subjected to nucleic acid homology searches of the GenBank, dBest, and NCI-CGAP databases.
  • RT-PCR Expression Analysis: First strand cDNAs can be generated from 1 μg of mRNA with oligo (dT)12-18 priming using the Gibco-BRL Superscript Preamplification system. The manufacturer's protocol was used which included an incubation for 50 min at 42° C. with reverse transcriptase followed by RNAse H treatment at 37° C. for 20 min. After completing the reaction, the volume can be increased to 200 μl with water prior to normalization. First strand cDNAs from 16 different normal human tissues can be obtained from Clontech.
  • Normalization of the first strand cDNAs from multiple tissues was performed by using the primers 5′ atatcgccgcgctcgtcgtcgacaa3′ (SEQ ID NO:30) and 5′ agccacacgcagetcattgtagaagg 3′ (SEQ ID NO:31) to amplify β-actin. First strand cDNA (5 μl) were amplified in a total volume of 50 μl containing 0.4 μM primers, 0.2 μM each dNTPs, 1×PCR buffer (Clontech, 10 mM Tris-HCL, 1.5 mM MgCl2, 50 mM KCl, pH8.3) and 1× Klentaq DNA polymerase (Clontech). Five μl of the PCR reaction can be removed at 18, 20, and 22 cycles and used for agarose gel electrophoresis. PCR was performed using an MJ Research thermal cycler under the following conditions: Initial denaturation can be at 94° C. for 15 sec, followed by a 18, 20, and 22 cycles of 94° C. for 15, 65° C. for 2 min., 72° C. for 5 sec. A final extension at 72° C. was carried out for 2 min. After agarose gel electrophoresis, the band intensities of the 283 b.p. β-actin bands from multiple tissues were compared by visual inspection. Dilution factors for the first strand cDNAs were calculated to result in equal β-actin band intensities in all tissues after 22 cycles of PCR. Three rounds of normalization can be required to achieve equal band intensities in all tissues after 22 cycles of PCR.
  • To determine expression levels of the 58P1D12 gene, 5 μl of normalized first strand cDNA were analyzed by PCR using 26, and 30 cycles of amplification. Semi-quantitative expression analysis can be achieved by comparing the PCR products at cycle numbers that give light band intensities. The primers used for RT-PCR were designed using the 58P1D12 SSH sequence and are listed below:
  • 58P1D12.1
    5′-CCTGCTTCAGTAACAACCACATTCT-3′ (SEQ ID NO: 32)
    58P1D12.2
    5′-CTTTACCAGTGGAATGATGACAGG-3′ (SEQ ID NO: 33)
  • A typical RT-PCR expression analysis is shown in FIG. 14.
  • Example 2 Full Length Cloning of 58P1D12
  • The 58P1D12 SSH sequence of 427 by (FIG. 1) was identified from LAPC xenograft SSH experiment. A full length cDNA clone for 58P1D12 was isolated from an LAPC-9 AD library. The cDNA (clone 2) is 2550 by in length and encodes a 273 amino acid ORF (FIG. 2 and FIG. 3). 58P1D12 v.1 exhibited 100% homology to human chondrolectin. Other variants of 58P1D12 were also identified and these are listed in FIG. 2 and FIG. 3.
  • Example 3 Chromosomal Mapping of 58P1D12
  • Chromosomal localization can implicate genes in disease pathogenesis. Several chromosome mapping approaches are available including fluorescent in situ hybridization (FISH), human/hamster radiation hybrid (RH) panels (Walter et al., 1994; Nature Genetics 7:22; Research Genetics, Huntsville Ala.), human-rodent somatic cell hybrid panels such as is available from the Coriell Institute (Camden, N.J.), and genomic viewers utilizing BLAST homologies to sequenced and mapped genomic clones (NCBI, Bethesda, Md.).
  • 58P1D12 maps to chromosome 21q11.2 using 58P1D12 sequence and the NCBI BLAST tool located on the World Wide Web at: (.ncbi.nlm.nih gov/genome/seq/page.cgi?F=HsBlast.html&&ORG=Hs).
  • Example 4 Expression Analysis of 58P1D12 in Normal Tissues and Patient Specimens
  • 58P1D12 expression in normal and patient cancer tissues was tested by PCR (FIG. 14). First strand cDNA was prepared from normal tissues (bladder, brain, heart, kidney, liver, lung, prostate, spleen, skeletal muscle, testis, pancreas, colon and stomach), and from pools of patient cancer specimens (prostate cancer pool, bladder cancer pool, lung cancer pool, ovary cancer pool, prostate cancer xenograft pool, bone and melanoma cancer pool, and cervical cancer pool). Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using primers to 58P1D12, was performed at 30 cycles of amplification. Results expression of 58P1D12 predominantly in testis amongst the normal tissues tested. 58P1D12 is also expressed in prostate cancer, bladder cancer, lung cancer, ovary cancer, prostate cancer xenograft, bone/melanoma cancer pool, and cervical cancer.
  • FIG. 15 shows expression of 58P1D12 in normal tissues by Northern blot analysis. Two multiple tissue northern blots (Clontech) both with 2 μg of mRNA/lane were probed with the 58P1D12 sequence. Size standards in kilobases (kb) are indicated on the side. Results show predominant expression of 58P1D12 transcript in normal testis. A significantly weaker signal is detected in spleen, lung, kidney and pancreas.
  • In order to show expression of 58P1D12 in prostate cancer tissues, RNA was extracted from LAPC prostate cancer xenografts, LAPC-4AD, LAPC-4AI, LAPC-9AD and LAPC-9AI, as well as from normal prostate. Northern blots with 10 μg of total RNA were probed with the 58P1D12 sequence. Size standards in kilobases are on the side. Results show strong expression of 58P1D12 in LAPC-9AD, and a weaker signal in LAPC-9AI, but not in the other tissues tested. (FIG. 16).
  • FIG. 17 shows 58P1D12 expression in normal and patient cancer specimens. RNA was extracted from a pool of 3 ovary tumor patient specimens, normal prostate (NP), normal bladder (NB), normal kidney (NK), and normal colon (NC). Northern blots with 10 μg of total RNA were probed with the 58P1D12 sequence. Size standards in kilobases are on the side. Results show expression of 58P1D12 in ovary tumor patient specimens but not in the normal tissues tested.
  • Analysis of 58P1D12 expression in individual ovarian cancer patient specimens is shown in FIG. 18. RNA was extracted from normal ovary, LAPC-9AD prostate cancer xenograft, and a panel of ovary tumor patient specimens (Ovary T). Northern blots with 10 μg of total RNA were probed with the 58P1D12 sequence. Size standards in kilobases are on the side. Results show expression of 58P1D12 in most of the patient specimens tested as well as in the prostate xenograft.
  • 58P1D12 expression was tested in a large panel of ovarian cancer patient specimens (FIG. 19). First strand cDNA was prepared from a panel of ovary patient cancer specimens as well as two different normal ovary tissues. Normalization was performed by PCR using primers to actin. Semi-quantitative PCR, using primers to 58P1D12, was performed at 26 and 30 cycles of amplification. Samples were run on an agarose gel, and PCR products were quantitated using the AlphaImager software. Expression was recorded as negative, weak, medium or strong. Results show expression of 58P1D12 in the majority of patient cancer specimens tested (75%), but not in normal ovary.
  • Example 5 Transcript Variants of 58P1D12
  • Transcript variants are variants of mature mRNA from the same gene which arise by alternative transcription or alternative splicing. Alternative transcripts are transcripts from the same gene but start transcription at different points. Splice variants are mRNA variants spliced differently from the same transcript. In eukaryotes, when a multi-exon gene is transcribed from genomic DNA, the initial RNA is spliced to produce functional mRNA, which has only exons and is used for translation into an amino acid sequence. Accordingly, a given gene can have zero to many alternative transcripts and each transcript can have zero to many splice variants. Each transcript variant has a unique exon makeup, and can have different coding and/or non-coding (5′ or 3′ end) portions, from the original transcript. Transcript variants can code for similar or different proteins with the same or a similar function or can encode proteins with different functions, and can be expressed in the same tissue at the same time, or in different tissues at the same time, or in the same tissue at different times, or in different tissues at different times. Proteins encoded by transcript variants can have similar or different cellular or extracellular localizations, e.g., secreted versus intracellular.
  • Transcript variants are identified by a variety of art-accepted methods. For example, alternative transcripts and splice variants are identified by full-length cloning experiment, or by use of full-length transcript and EST sequences. First, all human ESTs were grouped into clusters which show direct or indirect identity with each other. Second, ESTs in the same cluster were further grouped into sub-clusters and assembled into a consensus sequence. The original gene sequence is compared to the consensus sequence(s) or other full-length sequences. Each consensus sequence is a potential splice variant for that gene (see, e.g., the World Wide Web address doubletwist.com/products/c11_agentsOverview.jhtml). Even when a variant is identified that is not a full-length clone, that portion of the variant is very useful for antigen generation and for further cloning of the full-length splice variant, using techniques known in the art.
  • Moreover, computer programs are available in the art that identify transcript variants based on genomic sequences. Genomic-based transcript variant identification programs include FgenesH (A. Salamov and V. Solovyev, “Ab initio gene finding in Drosophila genomic DNA,” Genome Research. 2000 April; 10(4):516-22); Grail (URL compbio.ornl.gov/Grail-bin/EmptyGrailForm) and GenScan (URL genes.mit.edu/GENSCAN.html). For a general discussion of splice variant identification protocols see., e.g., Southan, C., A genomic perspective on human proteases, FEBS Lett. 2001 Jun. 8; 498(2-3):214-8; de Souza, S. J., et al., Identification of human chromosome 22 transcribed sequences with ORF expressed sequence tags, Proc. Natl. Acad Sci USA. 2000 Nov. 7; 97(23):12690-3.
  • To further confirm the parameters of a transcript variant, a variety of techniques are available in the art, such as full-length cloning, proteomic validation, PCR-based validation, and 5′ RACE validation, etc. (see e.g., Proteomic Validation: Brennan, S. O., et al., Albumin banks peninsula: a new termination variant characterized by electrospray mass spectrometry, Biochem Biophys Acta. 1999 Aug. 17; 1433(1-2):321-6; Ferranti P, et al., Differential splicing of pre-messenger RNA produces multiple forms of mature caprine alpha(s1)-casein, Eur J. Biochem. 1997 October 1; 249(1):1-7. For PCR-based Validation: Wellmann S, et al., Specific reverse transcription-PCR quantification of vascular endothelial growth factor (VEGF) splice variants by LightCycler technology, Clin Chem. 2001 April; 47(4):654-60; Jia, H. P., et al., Discovery of new human beta-defensins using a genomics-based approach, Gene. 2001 Jan. 24; 263(1-2):211-8. For PCR-based and 5′ RACE Validation: Brigle, K. E., et al., Organization of the murine reduced folate carrier gene and identification of variant splice forms, Biochem Biophys Acta. 1997 Aug. 7; 1353(2): 191-8).
  • It is known in the art that genomic regions are modulated in cancers. When the genomic region to which a gene maps is modulated in a particular cancer, the alternative transcripts or splice variants of the gene are modulated as well. Disclosed herein is that 58P1D12 has a particular expression profile related to cancer. Alternative transcripts and splice variants of 58P1D12 may also be involved in cancers in the same or different tissues, thus serving as tumor-associated markers/antigens.
  • Using the full-length gene and EST sequences, four transcript variants were identified, designated as 58P1D12 v.2, v.3, v.4 and v.5. The boundaries of the exon in the original transcript, 58P1D12 v.1 were shown in Table LI. Exon compositions of the variants were shown in FIG. 10. Theoretically, each different combination of exons in spatial order, e.g. exon 1 of v.2 and exons 3, 4, 5 and 6 of v.1, is a potential splice variant.
  • Tables LII(a)-(d) through LV(a)-(d) are set forth on a variant-by-variant bases. LII(a)-(d) shows nucleotide sequence of the transcript variant. Table LIII(a)-(d) shows the alignment of the transcript variant with nucleic acid sequence of 58P1D12 v.1. LIV(a)-(d) lays out amino acid translation of the transcript variant for the identified reading frame orientation. Table LV(a)-(d) displays alignments of the amino acid sequence encoded by the splice variant with that of 58P1D12 v.1.
  • Example 6 Single Nucleotide Polymorphisms of 58P1D12
  • A Single Nucleotide Polymorphism (SNP) is a single base pair variation in a nucleotide sequence at a specific location. At any given point of the genome, there are four possible nucleotide base pairs: A/T, C/G, G/C and T/A. Genotype refers to the specific base pair sequence of one or more locations in the genome of an individual. Haplotype refers to the base pair sequence of more than one location on the same DNA molecule (or the same chromosome in higher organisms), often in the context of one gene or in the context of several tightly linked genes. SNP that occurs on a cDNA is called cSNP. This cSNP may change amino acids of the protein encoded by the gene and thus change the functions of the protein. Some SNP cause inherited diseases; others contribute to quantitative variations in phenotype and reactions to environmental factors including diet and drugs among individuals. Therefore, SNP and/or combinations of alleles (called haplotypes) have many applications, including diagnosis of inherited diseases, determination of drug reactions and dosage, identification of genes responsible for diseases, and analysis of the genetic relationship between individuals (P. Nowotny, J. M. Kwon and A. M. Goate, “SNP analysis to dissect human traits,” Curr. Opin. Neurobiol. 2001 October; 11(5):637-641; M. Pirmohamed and B. K. Park, “Genetic susceptibility to adverse drug reactions,” Trends Pharmacol. Sci. 2001 June; 22(6):298-305; J. H. Riley, C. J. Allan, E. Lai and A. Roses, “The use of single nucleotide polymorphisms in the isolation of common disease genes,” Pharmacogenomics. 2000 February; 1(1):39-47; R. Judson, J. C. Stephens and A. Windemuth, “The predictive power of haplotypes in clinical response,” Pharmacogenomics. 2000 feb; 1(1):15-26).
  • SNP are identified by a variety of art-accepted methods (P. Bean, “The promising voyage of SNP target discovery,” Am. Clin. Lab. 2001 October-November; 20(9):18-20; K. M. Weiss, “In search of human variation,” Genome Res. 1998 July; 8(7):691-697; M. M. She, “Enabling large-scale pharmacogenetic studies by high-throughput mutation detection and genotyping technologies,” Clin. Chem. 2001 February; 47(2):164-172). For example, SNP can be identified by sequencing DNA fragments that show polymorphism by gel-based methods such as restriction fragment length polymorphism (RFLP) and denaturing gradient gel electrophoresis (DGGE). They can also be discovered by direct sequencing of DNA samples pooled from different individuals or by comparing sequences from different DNA samples. With the rapid accumulation of sequence data in public and private databases, one can discover SNP by comparing sequences using computer programs (Z. Gu, L. Hillier and P. Y. Kwok, “Single nucleotide polymorphism hunting in cyberspace,” Hum. Mutat. 1998; 12(4):221-225). SNP can be verified and genotype or haplotype of an individual can be determined by a variety of methods including direct sequencing and high throughput microarrays (P. Y. Kwok, “Methods for genotyping single nucleotide polymorphisms,” Annu. Rev. Genomics Hum. Genet. 2001; 2:235-258; M. Kokoris, K. Dix, K. Moynihan, J. Mathis, B. Erwin, P. Grass, B. Hines and A. Duesterhoeft, “High-throughput SNP genotyping with the Masscode system,” Mol. Diagn. 2000 December; 5(4):329-340).
  • Using the methods described above, ten SNP were identified in the original transcript, 58P1D12 v.1, at positions 1764 (A/C), 1987 (G/A), 2045 (A/G), 2066 (T/C), 2134 (T/A), 2350 (G/T), 2435 (G/T), 302 (G/T), 304 (G/T) and 1533 (C/T). The transcripts or proteins with alternative allele were designated as variant 58P1D12 v.6 through v.15, respectively. FIG. 12 shows the schematic alignment of the SNP variants. FIG. 11 shows the schematic alignment of protein variants, corresponding to nucleotide variants. Nucleotide variants that code for the same amino acid sequence as v.1 are not shown in FIG. 11. These alleles of the SNP, though shown separately here, can occur in different combinations (haplotypes) and in any one of the transcript variants (such as 58P1D12 v.2) that contains the site of the SNP.
  • Example 7 Production of Recombinant 58P1D12 in Prokaryotic Systems
  • To express recombinant 58P1D12 and 58P1D12 variants in prokaryotic cells, the full or partial length 58P1D12 and 58P1D12 variant cDNA sequences are cloned into any one of a variety of expression vectors known in the art. One or more of the following regions of 58P1D12 variants are expressed: the full length sequence presented in FIGS. 2 and 3, or any 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more contiguous amino acids from 58P1D12, variants, or analogs thereof.
  • A. In Vitro Transcription and Translation Constructs:
  • pCRII: To generate 58P1D12 sense and anti-sense RNA probes for RNA in situ investigations, pCRII constructs (Invitrogen, Carlsbad Calif.) are generated encoding either all or fragments of the 58P1D12 cDNA. The pCRII vector has Sp6 and T7 promoters flanking the insert to drive the transcription of 58P1D12 RNA for use as probes in RNA in situ hybridization experiments. These probes are used to analyze the cell and tissue expression of 58P1D12 at the RNA level. Transcribed 58P1D12 RNA representing the cDNA amino acid coding region of the 58P1D12 gene is used in in vitro translation systems such as the TnT™ Coupled Reticulolysate System (Promega, Corp., Madison, Wis.) to synthesize 58P1D12 protein.
  • B. Bacterial Constructs:
  • pGEX Constructs: To generate recombinant 58P1D12 proteins in bacteria that are fused to the Glutathione S-transferase (GST) protein, all or parts of the 58P1D12 cDNA protein coding sequence are cloned into the pGEX family of GST-fusion vectors (Amersham Pharmacia Biotech, Piscataway, N.J.). These constructs allow controlled expression of recombinant 58P1D12 protein sequences with GST fused at the amino-terminus and a six histidine epitope (6× His) at the carboxyl-terminus The GST and 6× His tags permit purification of the recombinant fusion protein from induced bacteria with the appropriate affinity matrix and allow recognition of the fusion protein with anti-GST and anti-His antibodies. The 6× His tag is generated by adding 6 histidine codons to the cloning primer at the 3′ end, e.g., of the open reading frame (ORF). A proteolytic cleavage site, such as the PreScission™ recognition site in pGEX-6P-1, may be employed such that it permits cleavage of the GST tag from 58P1D12-related protein. The ampicillin resistance gene and pBR322 origin permits selection and maintenance of the pGEX plasmids in E. coli.
  • pMAL Constructs: To generate, in bacteria, recombinant 58P1D12 proteins that are fused to maltose-binding protein (MBP), all or parts of the 58P1D12 cDNA protein coding sequence are fused to the MBP gene by cloning into the pMAL-c2X and pMAL-p2X vectors (New England Biolabs, Beverly, Mass.). These constructs allow controlled expression of recombinant 58P1D12 protein sequences with MBP fused at the amino-terminus and a 6× His epitope tag at the carboxyl-terminus The MBP and 6× His tags permit purification of the recombinant protein from induced bacteria with the appropriate affinity matrix and allow recognition of the fusion protein with anti-MBP and anti-His antibodies. The 6× His epitope tag is generated by adding 6 histidine codons to the 3′ cloning primer. A Factor Xa recognition site permits cleavage of the pMAL tag from 58P1D12. The pMAL-c2X and pMAL-p2X vectors are optimized to express the recombinant protein in the cytoplasm or periplasm respectively. Periplasm expression enhances folding of proteins with disulfide bonds.
  • pET Constructs: To express 58P1D12 in bacterial cells, all or parts of the 58P1D12 cDNA protein coding sequence are cloned into the pET family of vectors (Novagen, Madison, Wis.). These vectors allow tightly controlled expression of recombinant 58P1D12 protein in bacteria with and without fusion to proteins that enhance solubility, such as NusA and thioredoxin (Trx), and epitope tags, such as 6× His and S-Tag™ that aid purification and detection of the recombinant protein. For example, constructs are made utilizing pET NusA fusion system 43.1 such that regions of the 58P1D12 protein are expressed as amino-terminal fusions to NusA. The cDNA encoding amino acids 22-213 of 58P1D12 was cloned into the pET-21b vector, expressed, and purified from bacteria. The recombinant protein was used to generate rabbit polyclonal antibodies.
  • C. Yeast Constructs:
  • pESC Constructs: To express 58P1D12 in the yeast species Saccharomyces cerevisiae for generation of recombinant protein and functional studies, all or parts of the 58P1D12 cDNA protein coding sequence are cloned into the pESC family of vectors each of which contain 1 of 4 selectable markers, HIS3, TRP1, LEU2, and URA3 (Stratagene, La Jolla, Calif.). These vectors allow controlled expression from the same plasmid of up to 2 different genes or cloned sequences containing either Flag™ or Myc epitope tags in the same yeast cell. This system is useful to confirm protein-protein interactions of 58P1D12. In addition, expression in yeast yields similar post-translational modifications, such as glycosylations and phosphorylations that are found when expressed in eukaryotic cells.
  • pESP Constructs: To express 58P1D12 in the yeast species Saccharomyces pombe, all or parts of the 58P1D12 cDNA protein coding sequence are cloned into the pESP family of vectors. These vectors allow controlled high level of expression of a 58P1D12 protein sequence that is fused at either the amino terminus or at the carboxyl terminus to GST which aids purification of the recombinant protein. A Flag™ epitope tag allows detection of the recombinant protein with anti-Flag™ antibody.
  • Example 8 Production of Recombinant 58P1D12 in Higher Eukaryotic Systems
  • A. Mammalian Constructs:
  • To express recombinant 58P1D12 in eukaryotic cells, the full or partial length 58P1D12 cDNA sequences were cloned into any one of a variety of expression vectors known in the art. One or more of the following regions of 58P1D12 were expressed in these constructs, amino acids 1 to 273 or any 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 or more contiguous amino acids from 58P1D12 v.1; amino acids 1 to 232 of v.2; amino acids 1 to 236 of v.4; or any 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or more contiguous amino acids from 58P1D12 variants, or analogs thereof.
  • The constructs can be transfected into any one of a wide variety of mammalian cells such as 293T cells. Transfected 293T cell lysates can be probed with the anti-58P1D12 polyclonal serum, described herein.
  • pcDNA4/H isMax Constructs: To express 58P1D12 in mammalian cells, a 58P1D12 ORF, or portions thereof, of 58P1D12 are cloned into pcDNA4/H isMax Version A (Invitrogen, Carlsbad, Calif.). Protein expression is driven from the cytomegalovirus (CMV) promoter and the SP16 translational enhancer. The recombinant protein has Xpress™ and six histidine (6× His) epitopes fused to the amino-terminus. The pcDNA4/H isMax vector also contains the bovine growth hormone (BGH) polyadenylation signal and transcription termination sequence to enhance mRNA stability along with the SV40 origin for episomal replication and simple vector rescue in cell lines expressing the large T antigen. The Zeocin resistance gene allows for selection of mammalian cells expressing the protein and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli.
  • pcDNA3.1/MycHis Constructs: To express 58P1D12 in mammalian cells, a 58P1D12 ORF, or portions thereof, of 58P1D12 with a consensus Kozak translation initiation site was cloned into pcDNA3.1/MycHis Version A (Invitrogen, Carlsbad, Calif.). Protein expression is driven from the cytomegalovirus (CMV) promoter. The recombinant proteins have the myc epitope and 6×His epitope fused to the carboxyl-terminus The pcDNA3.1/MycHis vector also contains the bovine growth hormone (BGH) polyadenylation signal and transcription termination sequence to enhance mRNA stability, along with the SV40 origin for episomal replication and simple vector rescue in cell lines expressing the large T antigen. The Neomycin resistance gene can be used, as it allows for selection of mammalian cells expressing the protein and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli.
  • The complete ORF of 58P1D12 v.1 was cloned into the pcDNA3.1/MycHis construct to generate 58P1D12.pcDNA3.1/MycHis.
  • pcDNA3.1/CT-GFP-TOPO Construct: To express 58P1D12 in mammalian cells and to allow detection of the recombinant proteins using fluorescence, a 58P1D12 ORF, or portions thereof, with a consensus Kozak translation initiation site are cloned into pcDNA3.1/CT-GFP-TOPO (Invitrogen, CA). Protein expression is driven from the cytomegalovirus (CMV) promoter. The recombinant proteins have the Green Fluorescent Protein (GFP) fused to the carboxyl-terminus facilitating non-invasive, in vivo detection and cell biology studies. The pcDNA3.1CT-GFP-TOPO vector also contains the bovine growth hormone (BGH) polyadenylation signal and transcription termination sequence to enhance mRNA stability along with the SV40 origin for episomal replication and simple vector rescue in cell lines expressing the large T antigen. The Neomycin resistance gene allows for selection of mammalian cells that express the protein and the ampicillin resistance gene and ColE1 origin permits selection and maintenance of the plasmid in E. coli. Additional constructs with an amino-terminal GFP fusion are made in pcDNA3.1/NT-GFP-TOPO spanning the entire length of a 58P1D12 protein.
  • PAPtag: A 58P1D12 ORF, or portions thereof, were cloned into pAPtag-5 (GenHunter Corp. Nashville, Tenn.). This construct generates an alkaline phosphatase fusion at the carboxyl-terminus of a 58P1D12 protein while fusing the IgGκ signal sequence to the amino-terminus. Constructs are also generated in which alkaline phosphatase with an amino-terminal IgGκ signal sequence is fused to the amino-terminus of a 58P1D12 protein. The resulting recombinant 58P1D12 proteins are optimized for secretion into the media of transfected mammalian cells and can be used to identify proteins such as ligands or receptors that interact with 58P1D12 proteins. Protein expression is driven from the CMV promoter and the recombinant proteins also contain myc and 6× His epitopes fused at the carboxyl-terminus that facilitates detection and purification. The Zeocin resistance gene present in the vector allows for selection of mammalian cells expressing the recombinant protein and the ampicillin resistance gene permits selection of the plasmid in E. coli.
  • pTag5: A 58P1D12 ORF, or portions thereof, were cloned into pTag-5. This vector is similar to pAPtag but without the alkaline phosphatase fusion. This construct generated 58P1D12 protein with an amino-terminal IgGκ signal sequence and myc and 6× His epitope tags at the carboxyl-terminus that facilitate detection and affinity purification. The resulting recombinant 58P1D12 protein was optimized for secretion into the media of transfected mammalian cells, and was used as immunogen or ligand to identify proteins such as ligands or receptors that interact with the 58P1D12 proteins. Protein expression is driven from the CMV promoter. The Zeocin resistance gene present in the vector allows for selection of mammalian cells expressing the protein, and the ampicillin resistance gene permits selection of the plasmid in E. coli.
  • PsecFc: A 58P1D12 ORF, or portions thereof, were cloned into psecFc. The psecFc vector was assembled by cloning the human immunoglobulin G1 (IgG) Fc (hinge, CH2, CH3 regions) into pSecTag2 (Invitrogen, California). This construct generated an IgG1 Fc fusion at the carboxyl-terminus of the 58P1D12 proteins, while fusing the IgGK signal sequence to N-terminus 58P1D12 fusions utilizing the murine IgG1 Fc region are also used. The resulting recombinant 58P1D12 proteins are optimized for secretion into the media of transfected mammalian cells, and can be used as immunogens or to identify proteins such as ligands or receptors that interact with 58P1D12 protein. Protein expression is driven from the CMV promoter. The hygromycin resistance gene present in the vector allows for selection of mammalian cells that express the recombinant protein, and the ampicillin resistance gene permits selection of the plasmid in E. coli.
  • pSRα Constructs: To generate mammalian cell lines that express 58P1D12 constitutively, 58P1D12 ORF, or portions thereof, were cloned into pSRα constructs. Amphotropic and ecotropic retroviruses were generated by transfection of pSRα constructs into the 293T-10A1 packaging line or co-transfection of pSRα and a helper plasmid (containing deleted packaging sequences) into the 293 cells, respectively. The retrovirus is used to infect a variety of mammalian cell lines, resulting in the integration of the cloned gene, 58P1D12, into the host cell-lines. Protein expression is driven from a long terminal repeat (LTR). The Neomycin resistance gene present in the vector allows for selection of mammalian cells that express the protein, and the ampicillin resistance gene and ColE1 origin permit selection and maintenance of the plasmid in E. coli. The retroviral vectors can thereafter be used for infection and generation of various cell lines using, for example, PC3, NIH 3T3, TsuPrl, 293 or rat-1 cells.
  • Additional pSRα constructs are made that fuse an epitope tag such as the FLAG tag to the carboxyl-terminus of 58P1D12 sequences to allow detection using anti-Flag antibodies. For example, the FLAG sequence 5′ gat tac aag gat gac gac gat aag 3′ (SEQ ID NO:34) is added to cloning primer at the 3′ end of the ORF. Additional pSRα constructs are made to produce both amino-terminal and carboxyl-terminal GFP and myc/6× His fusion proteins of the full-length 58P1D12 proteins.
  • Additional Viral Vectors: Additional constructs are made for viral-mediated delivery and expression of 58P1D12. High virus titer leading to high level expression of 58P1D12 is achieved in viral delivery systems such as adenoviral vectors and herpes amplicon vectors. A 58P1D12 coding sequence or fragments thereof are amplified by PCR and subcloned into the AdEasy shuttle vector (Stratagene). Recombination and virus packaging are performed according to the manufacturer's instructions to generate adenoviral vectors. Alternatively, 58P1D12 coding sequences or fragments thereof are cloned into the HSV-1 vector (Imgenex) to generate herpes viral vectors. The viral vectors are thereafter used for infection of various cell lines such as PC3, NIH 3T3, 293 or rat-1 cells.
  • Regulated Expression Systems: To control expression of 58P1D12 in mammalian cells, coding sequences of 58P1D12, or portions thereof, are cloned into regulated mammalian expression systems such as the T-Rex System (Invitrogen), the GeneSwitch System (Invitrogen) and the tightly-regulated Ecdysone System (Sratagene). These systems allow the study of the temporal and concentration dependent effects of recombinant 58P1D12. These vectors are thereafter used to control expression of 58P1D12 in various cell lines such as PC3, NIH 3T3, 293 or rat-1 cells.
  • B. Baculovirus Expression Systems:
  • To generate recombinant 58P1D12 proteins in a baculovirus expression system, 58P1D12 ORF, or portions thereof, are cloned into the baculovirus transfer vector pBlueBac 4.5 (Invitrogen), which provides a His-tag at the N-terminus. Specifically, pBlueBac-58P1D12 is co-transfected with helper plasmid pBac-N-Blue (Invitrogen) into SF9 (Spodoptera frugiperda) insect cells to generate recombinant baculovirus (see Invitrogen instruction manual for details). Baculovirus is then collected from cell supernatant and purified by plaque assay.
  • Recombinant 58P1D12 protein is then generated by infection of HighFive insect cells (Invitrogen) with purified baculovirus. Recombinant 58P1D12 protein can be detected using anti-58P1D12 or anti-His-tag antibody. 58P1D12 protein can be purified and used in various cell-based assays or as immunogen to generate polyclonal and monoclonal antibodies specific for 58P1D12.
  • Example 9 Antigenicity Profiles and Secondary Structure
  • FIG. 5A-C, FIG. 6A-C, FIG. 7A-C, FIG. 8A-C, and FIG. 9A-C depict graphically five amino acid profiles of 58P1D12 variant 1, variant 2, and variant 3, A through C respectively, each assessment available by accessing the ProtScale website (at the World Wide Web address expasy.ch/cgi-bin/protscale.pl) on the ExPasy molecular biology server.
  • These profiles: FIG. 5, Hydrophilicity, (Hopp T. P., Woods K. R., 1981. Proc. Natl. Acad. Sci. U.S.A. 78:3824-3828); FIG. 6, Hydropathicity, (Kyte J., Doolittle R.F., 1982. J. Mol. Biol. 157:105-132); FIG. 7, Percentage Accessible Residues (Janin J., 1979 Nature 277:491-492); FIG. 8, Average Flexibility, (Bhaskaran R., and Ponnuswamy P.K., 1988. Int. J. Pept. Protein Res. 32:242-255); FIG. 9, Beta-turn (Deleage, G., Roux B. 1987 Protein Engineering 1:289-294); and optionally others available in the art, such as on the ProtScale website, were used to identify antigenic regions of each of the 58P1D12 variant proteins. Each of the above amino acid profiles of 58P1D12 variants were generated using the following ProtScale parameters for analysis: 1) A window size of 9; 2) 100% weight of the window edges compared to the window center; and, 3) amino acid profile values normalized to lie between 0 and 1.
  • Hydrophilicity (FIG. 5), Hydropathicity (FIG. 6) and Percentage Accessible Residues (FIG. 7) profiles were used to determine stretches of hydrophilic amino acids (i.e., values greater than 0.5 on the Hydrophilicity and Percentage Accessible Residues profile, and values less than 0.5 on the Hydropathicity profile). Such regions are likely to be exposed to the aqueous environment, be present on the surface of the protein, and thus available for immune recognition, such as by antibodies.
  • Average Flexibility (FIG. 8) and Beta-turn (FIG. 9) profiles determine stretches of amino acids (i.e., values greater than 0.5 on the Beta-turn profile and the Average Flexibility profile) that are not constrained in secondary structures such as beta sheets and alpha helices. Such regions are also more likely to be exposed on the protein and thus accessible to immune recognition, such as by antibodies.
  • Antigenic sequences of the 58P1D12 variant proteins indicated, e.g., by the profiles set forth in FIG. 5, FIG. 6, FIG. 7, FIG. 8, and/or FIG. 9 are used to prepare immunogens, either peptides or nucleic acids that encode them, to generate therapeutic and diagnostic anti-58P1D12 antibodies. The immunogen can be any 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 30, 35, 40, 45, 50 or more than 50 contiguous amino acids, or the corresponding nucleic acids that encode them, from the 58P1D12 protein variants listed in FIGS. 2 and 3. In particular, peptide immunogens of the invention can comprise, a peptide region of at least 5 amino acids of FIGS. 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Hydrophilicity profiles of FIG. 5; a peptide region of at least 5 amino acids of FIGS. 2 and 3 in any whole number increment that includes an amino acid position having a value less than 0.5 in the Hydropathicity profile of FIG. 6; a peptide region of at least 5 amino acids of FIGS. 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Percent Accessible Residues profiles of FIG. 7; a peptide region of at least 5 amino acids of FIGS. 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Average Flexibility profiles on FIG. 8; and, a peptide region of at least 5 amino acids of FIGS. 2 and 3 in any whole number increment that includes an amino acid position having a value greater than 0.5 in the Beta-turn profile of FIG. 9. Peptide immunogens of the invention can also comprise nucleic acids that encode any of the forgoing.
  • All immunogens of the invention, peptide or nucleic acid, can be embodied in human unit dose form, or comprised by a composition that includes a pharmaceutical excipient compatible with human physiology.
  • The secondary structures of 58P1D12 protein variants 1, 2, and 3, namely the predicted presence and location of alpha helices, extended strands, and random coils, are predicted from their primary amino acid sequences using the HNN—Hierarchical Neural Network method (NPS@: Network Protein Sequence Analysis TIBS 2000 March Vol. 25, No 3 [291]:147-150 Combet C., Blanchet C., Geourjon C. and Deléage G., at the World Wide Web address pbil.ibcp.fr/cgi-bin/npsa_automat.pl?page=npsa_nn.html), accessed from the ExPasy molecular biology server (at the World Wide Web address expasy.ch/tools/). The analysis indicates that 58P1D12 variant 1 is composed of 24.18% alpha helix, 18.68% extended strand, and 57.14% random coil (FIG. 13A). 58P1D12 variant 2 is composed of 19.83% alpha helix, 18.97% extended strand, and 61.21% random coil (FIG. 13B). 58P1D12 variant 3 is composed of 32.20% alpha helix, 15.25% extended strand, and 52.54% random coil (FIG. 13C).
  • Analysis for the potential presence of transmembrane domains in the 58P1D12 variant proteins 1, 2 and 3, was carried out using a variety of transmembrane prediction algorithms accessed from the ExPasy molecular biology server (at the World Wide Web address expasy.ch/tools/). Shown graphically in FIG. 13D is the result of analysis of variant 1 using the TMpred program and in FIG. 13E results using the TMHMM program, both of which predict the presence of 1 transmembrane domain Shown graphically in FIG. 13F is the result of analysis of variant 2 using the TMpred program and in FIG. 13G results using the TMHMM program, both of which predict the presence of 1 transmembrane domain. Variant 3 is not predicted to contain transmembrane domains (FIGS. 13H and 13I). Analyses of the variants using other structural prediction programs are summarized in Table VI and Table L.
  • Example 10 Generation of 58P1D12 Polyclonal Antibodies
  • Polyclonal antibodies can be raised in a mammal, for example, by one or more injections of an immunizing agent and, if desired, an adjuvant. Typically, the immunizing agent and/or adjuvant will be injected in the mammal by multiple subcutaneous or intraperitoneal injections. In addition to immunizing with a full length 58P1D12 protein variant, computer algorithms are employed in design of immunogens that, based on amino acid sequence analysis contain characteristics of being antigenic and available for recognition by the immune system of the immunized host (see the Example entitled “Antigenicity Profiles and Secondary Structure”). Such regions would be predicted to be hydrophilic, flexible, in beta-turn conformations, and be exposed on the surface of the protein (see, e.g., FIG. 5, FIG. 6, FIG. 7, FIG. 8, or FIG. 9 for amino acid profiles that indicate such regions of 58P1D12 protein variant 1).
  • For example, recombinant bacterial fusion proteins or peptides containing hydrophilic, flexible, beta-turn regions of 58P1D12 protein variants are used as antigens to generate polyclonal antibodies in New Zealand White rabbits or monoclonal antibodies as described in Example 11. For example, in 58P1D12 variant 1, such regions include, but are not limited to, amino acids 19-30, amino acids 49-66, amino acids 70-82, amino acids 88-115, amino acids 131-145, amino acids 165-203, amino acids 243-255, and amino acids 262-273. It is useful to conjugate the immunizing agent to a protein known to be immunogenic in the mammal being immunized. Examples of such immunogenic proteins include, but are not limited to, keyhole limpet hemocyanin (KLH), serum albumin, bovine thyroglobulin, and soybean trypsin inhibitor. In one embodiment, a peptide encoding amino acids 102-115 of 58P1D12 variant 1 was conjugated to KLH and used to immunize a rabbit. Alternatively the immunizing agent may include all or portions of the 58P1D12 variant proteins, analogs or fusion proteins thereof. For example, the 58P1D12 variant 1 amino acid sequence can be fused using recombinant DNA techniques to any one of a variety of fusion protein partners that are well known in the art, such as glutathione-S-transferase (GST) and HIS tagged fusion proteins. In another embodiment, amino acids 22-213 of 58P1D12 variant 1 is fused to H is using recombinant techniques and the pET21b expression vector. The protein was then expressed, purified, and used to immunize 2 rabbits. Such fusion proteins are purified from induced bacteria using the appropriate affinity matrix.
  • Other recombinant bacterial fusion proteins that may be employed include maltose binding protein, LacZ, thioredoxin, NusA, or an immunoglobulin constant region (see the section entitled “Production of 58P1D12 in Prokaryotic Systems” and Current Protocols In Molecular Biology, Volume 2, Unit 16, Frederick M. Ausubul et al. eds., 1995; Linsley, P.S., Brady, W., Urnes, M., Grosmaire, L., Damle, N., and Ledbetter, J. (1991) J. Exp. Med. 174, 561-566).
  • In addition to bacterial derived fusion proteins, mammalian expressed protein antigens are also used. These antigens are expressed from mammalian expression vectors such as the Tag5 and Fc-fusion vectors (see the section entitled “Production of Recombinant 58P1D12 in Eukaryotic Systems”), and retain post-translational modifications such as glycosylations found in native protein. In one embodiment, amino acids 22-213 of 58P1D12 variant 1 was cloned into the Tag5 mammalian secretion vector, and expressed in 293T cells. The recombinant protein was purified by metal chelate chromatography from tissue culture supernatants of 293T cells stably expressing the recombinant vector. The purified Tag5 58P1D12 protein was then used as immunogen.
  • During the immunization protocol, it is useful to mix or emulsify the antigen in adjuvants that enhance the immune response of the host animal. Examples of adjuvants include, but are not limited to, complete Freund's adjuvant (CFA) and MPL-TDM adjuvant (monophosphoryl Lipid A, synthetic trehalose dicorynomycolate).
  • In a typical protocol, rabbits are initially immunized subcutaneously with up to 200 μg, typically 100-200 μg, of fusion protein or peptide conjugated to KLH mixed in complete Freund's adjuvant (CFA). Rabbits are then injected subcutaneously every two weeks with up to 200 μg, typically 100-200 μg, of the immunogen in incomplete Freund's adjuvant (IFA). Test bleeds are taken approximately 7-10 days following each immunization and used to monitor the titer of the antiserum by ELISA.
  • To test reactivity and specificity of immune serum, such as the rabbit serum derived from immunization with the His-fusion of 58P1D12 variant 1 protein, the full-length 58P1D12 variant 1 cDNA was cloned into pcDNA 3.1 myc-his expression vector (Invitrogen, see the Example entitled “Production of Recombinant 58P1D12 in Eukaryotic Systems”). After transfection of the constructs into 293T cells, cell lysates are probed with the anti-58P1D12 serum and with anti-His antibody (Santa Cruz Biotechnologies, Santa Cruz, Calif.) to determine specific reactivity to denatured 58P1D12 protein using the Western blot technique. In addition, the immune serum is tested by fluorescence microscopy, flow cytometry and immunoprecipitation against 293T and other recombinant 58P1D12-expressing cells to determine specific recognition of native protein. Western blot, immunoprecipitation, fluorescent microscopy, and flow cytometric techniques using cells that endogenously express 58P1D12 are also carried out to test reactivity and specificity.
  • Anti-serum from rabbits immunized with 58P1D12 variant fusion proteins, such as GST and MBP fusion proteins, are purified by depletion of antibodies reactive to the fusion partner sequence by passage over an affinity column containing the fusion partner either alone or in the context of an irrelevant fusion protein. For example, antiserum derived from a GST-58P1D12 variant 1 fusion protein is first purified by passage over a column of GST protein covalently coupled to AffiGel matrix (BioRad, Hercules, Calif.). The antiserum is then affinity purified by passage over a column composed of a MBP-58P1D12 fusion protein covalently coupled to Affigel matrix. The serum is then further purified by protein G affinity chromatography to isolate the IgG fraction. Sera from other His-tagged antigens and peptide immunized rabbits as well as fusion partner depleted sera are affinity purified by passage over a column matrix composed of the original protein immunogen or free peptide.
  • Example 11 Generation of 58P1D12 Monoclonal Antibodies (mAbs)
  • In one embodiment, therapeutic mAbs to 58P1D12 variants comprise those that react with epitopes specific for each variant protein or specific to sequences in common between the variants that would disrupt or modulate the biological function of the 58P1D12 variants, for example those that would disrupt the interaction with ligands and binding partners Immunogens for generation of such mAbs include those designed to encode or contain the entire 58P1D12 protein variant sequence, regions predicted to contain functional motifs, and regions of the 58P1D12 protein variants predicted to be antigenic from computer analysis of the amino acid sequence (see, e.g., FIG. 5, FIG. 6, FIG. 7, FIG. 8, or FIG. 9, and the Example entitled “Antigenicity Profiles and Secondary Structure”) Immunogens include peptides, recombinant bacterial proteins, and mammalian expressed Tag 5 proteins and human and murine IgG FC fusion proteins. In addition, cells engineered to express high levels of a respective 58P1D12 variant, such as Rat1-58P1D12 variant 1 or 300.19-58P1D12 variant lmurine Pre-B cells, were used to immunize mice.
  • To generate mAbs to a 58P1D12 variant, mice are first immunized intraperitoneally (IP) with, typically, 10-50 μg of protein immunogen or 107 58P1D12-expressing cells mixed in complete Freund's adjuvant. Mice are then subsequently immunized IP every 2-4 weeks with, typically, 10-50 μg of protein immunogen or 107 cells mixed in incomplete Freund's adjuvant. Alternatively, MPL-TDM adjuvant is used in immunizations. In addition to the above protein and cell-based immunization strategies, a DNA-based immunization protocol is employed in which a mammalian expression vector encoding a 58P1D12 variant sequence is used to immunize mice by direct injection of the plasmid DNA. For example, amino acids 22-213 of 58P1D12 of variant 1 is cloned into the Tag5 mammalian secretion vector and the recombinant vector will then be used as immunogen. In another example the same amino acids are cloned into an Fc-fusion secretion vector in which the 58P1D12 variant 1 sequence is fused at the amino-terminus to an IgK leader sequence and at the carboxyl-terminus to the coding sequence of the human or murine IgG Fc region. This recombinant vector is then used as immunogen. The plasmid immunization protocols are used in combination with purified proteins expressed from the same vector and with cells expressing the respective 58P1D12 variant.
  • During the immunization protocol, test bleeds are taken 7-10 days following an injection to monitor titer and specificity of the immune response. Once appropriate reactivity and specificity is obtained as determined by ELISA, Western blotting, immunoprecipitation, fluorescence microscopy, and flow cytometric analyses, fusion and hybridoma generation is then carried out with established procedures well known in the art (see, e.g., Harlow and Lane, 1988).
  • In one embodiment for generating 58P1D12 monoclonal antibodies, amino acids 22-213 of 58P1D12 variant 1 was cloned into the pTag5 vector, the protein was then expressed in 293T cells and purified by metal chelate chromatography. Balb C mice were immunized with 10 μg of recombinant variant 1 protein mixed in adjuvant. Mice were subsequently immunized over several weeks with the protein. ELISA using the antigen determined the titer of serum from the immunized mice. Reactivity and specificity of the serum to full length 58P1D12 variant 1 protein was monitored by flow cytometry using Rat-58P1D12 cells compared to Rat1-neo control cells (FIG. 20). Other recombinant 58P1D12 variant 1-expressing cells or cells endogenously expressing 58P1D12 variant 1 are also used. Mice showing the strongest reactivity were rested and given a final injection of antigen and sacrificed for fusion. The lymph nodes of the sacrificed mice were harvested and fused to SP2/0 myeloma cells using standard procedures (Harlow and Lane, 1988). Supernatants from HAT selected growth wells were analyzed by flow cytometry to identify specific-58P1D12 surface binding MAbs (FIG. 21). Supernatants are also be screened by ELISA, Western blot, immunoprecipitation, and fluorescent microscopy to identify 58P1D12 specific antibody-producing clones.
  • In another embodiment, 58P1D12 variant 3 specific MAbs are generated by employing immunogens that encode amino acid sequences unique to variant 3. For example, a GST-fusion protein is constructed to encode amino acids 171-236, expressed, purified, and used to immunize mice. Hybridomas resulting from fusion of the B-cells from the mice are screened on cells expressing 58P1D12 variant 3 protein and cross-screened on cells expressing variant 1 protein to identify variant 3-specific MAbs.
  • The binding affinity of 58P1D12 variant specific monoclonal antibodies are determined using standard technologies. Affinity measurements quantify the strength of antibody to epitope binding and are used to help define which 58P1D12 variant monoclonal antibodies preferred for diagnostic or therapeutic use, as appreciated by one of skill in the art. The BIAcore system (Uppsala, Sweden) is a preferred method for determining binding affinity. The BIAcore system uses surface plasmon resonance (SPR, Welford K. 1991, Opt. Quant. Elect. 23:1; Morton and Myszka, 1998, Methods in Enzymology 295: 268) to monitor biomolecular interactions in real time. BIAcore analysis conveniently generates association rate constants, dissociation rate constants, equilibrium dissociation constants, and affinity constants. In addition, equilibrium binding analysis of a dilution series of the MAb is also used to determine affinity defined by the dissociation constant (KD). The KD is determined by non-linear regression of the equilibrium binding data of the concentration series. The KD is defined as the concentration at which half-maximal binding of the MAb to the antigen is attained. Such analysis for MAb M8-143(5).10.1 is shown in FIG. 22.
  • Example 12 HLA Class I and Class II Binding Assays
  • HLA class I and class II binding assays using purified HLA molecules are performed in accordance with disclosed protocols (e.g., PCT publications WO 94/20127 and WO 94/03205; Sidney et al., Current Protocols in Immunology 18.3.1 (1998); Sidney, et al., J. Immunol. 154:247 (1995); Sette, et al., Mol. Immunol. 31:813 (1994)). Briefly, purified MHC molecules (5 to 500 nM) are incubated with various unlabeled peptide inhibitors and 1-10 nM 125I-radiolabeled probe peptides as described. Following incubation, MHC-peptide complexes are separated from free peptide by gel filtration and the fraction of peptide bound is determined. Typically, in preliminary experiments, each MHC preparation is titered in the presence of fixed amounts of radiolabeled peptides to determine the concentration of HLA molecules necessary to bind 10-20% of the total radioactivity. All subsequent inhibition and direct binding assays are performed using these HLA concentrations.
  • Since under these conditions [label]<[HLA] and IC50≧[HLA], the measured IC50 values are reasonable approximations of the true KD values. Peptide inhibitors are typically tested at concentrations ranging from 120 μg/ml to 1.2 ng/ml, and are tested in two to four completely independent experiments. To allow comparison of the data obtained in different experiments, a relative binding figure is calculated for each peptide by dividing the IC50 of a positive control for inhibition by the IC50 for each tested peptide (typically unlabeled versions of the radiolabeled probe peptide). For database purposes, and inter-experiment comparisons, relative binding values are compiled. These values can subsequently be converted back into IC50 nM values by dividing the IC50 nM of the positive controls for inhibition by the relative binding of the peptide of interest. This method of data compilation is accurate and consistent for comparing peptides that have been tested on different days, or with different lots of purified MHC.
  • Binding assays as outlined above may be used to analyze HLA supermotif and/or HLA motif-bearing peptides (see Table IV).
  • Example 13 Identification of HLA Supermotif- and Motif-Bearing CTL Candidate Epitopes
  • HLA vaccine compositions of the invention can include multiple epitopes. The multiple epitopes can comprise multiple HLA supermotifs or motifs to achieve broad population coverage. This example illustrates the identification and confirmation of supermotif- and motif-bearing epitopes for the inclusion in such a vaccine composition. Calculation of population coverage is performed using the strategy described below.
  • Computer searches and algorithms for identification of supermotif and/or motif-bearing epitopes
  • The searches performed to identify the motif-bearing peptide sequences in the Example entitled “Antigenicity Profiles” and Tables VIII-XXI and XXII-XLIX employ the protein sequence data from the gene product of 58P1D12 set forth in FIGS. 2 and 3, the specific search peptides used to generate the tables are listed in Table VII.
  • Computer searches for epitopes bearing HLA Class I or Class II supermotifs or motifs are performed as follows. All translated 58P1D12 protein sequences are analyzed using a text string search software program to identify potential peptide sequences containing appropriate HLA binding motifs; such programs are readily produced in accordance with information in the art in view of known motif/supermotif disclosures. Furthermore, such calculations can be made mentally.
  • Identified A2-, A3-, and DR-supermotif sequences are scored using polynomial algorithms to predict their capacity to bind to specific HLA-Class I or Class II molecules. These polynomial algorithms account for the impact of different amino acids at different positions, and are essentially based on the premise that the overall affinity (or AG) of peptide-HLA molecule interactions can be approximated as a linear polynomial function of the type:

  • “ΔG”=a1i×a2i×a3 . . . i×ani
  • where aji is a coefficient which represents the effect of the presence of a given amino acid (j) at a given position (i) along the sequence of a peptide of n amino acids. The crucial assumption of this method is that the effects at each position are essentially independent of each other (i.e., independent binding of individual side-chains). When residue j occurs at position i in the peptide, it is assumed to contribute a constant amount ji to the free energy of binding of the peptide irrespective of the sequence of the rest of the peptide.
  • The method of derivation of specific algorithm coefficients has been described in Gulukota et al., J. Mol. Biol. 267:1258-126, 1997; (see also Sidney et al., Human Immunol. 45:79-93, 1996; and Southwood et al., J. Immunol. 160:3363-3373, 1998). Briefly, for all i positions, anchor and non-anchor alike, the geometric mean of the average relative binding (ARB) of all peptides carrying j is calculated relative to the remainder of the group, and used as the estimate of ji. For Class II peptides, if multiple alignments are possible, only the highest scoring alignment is utilized, following an iterative procedure. To calculate an algorithm score of a given peptide in a test set, the ARB values corresponding to the sequence of the peptide are multiplied. If this product exceeds a chosen threshold, the peptide is predicted to bind. Appropriate thresholds are chosen as a function of the degree of stringency of prediction desired.
  • Selection of HLA-A2 Supertype Cross-Reactive Peptides
  • Protein sequences from 58P1D12 are scanned utilizing motif identification software, to identify 8-, 9-10- and 11-mer sequences containing the HLA-A2-supermotif main anchor specificity. Typically, these sequences are then scored using the protocol described above and the peptides corresponding to the positive-scoring sequences are synthesized and tested for their capacity to bind purified HLA-A*0201 molecules in vitro (HLA-A*0201 is considered a prototype A2 supertype molecule).
  • These peptides are then tested for the capacity to bind to additional A2-supertype molecules (A*0202, A*0203, A*0206, and A*6802). Peptides that bind to at least three of the five A2-supertype alleles tested are typically deemed A2-supertype cross-reactive binders. Preferred peptides bind at an affinity equal to or less than 500 nM to three or more HLA-A2 supertype molecules.
  • Selection of HLA-A3 Supermotif-Bearing Epitopes
  • The 58P1D12 protein sequence(s) scanned above is also examined for the presence of peptides with the HLA-A3-supermotif primary anchors. Peptides corresponding to the HLA A3 supermotif-bearing sequences are then synthesized and tested for binding to HLA-A*0301 and HLA-A*1101 molecules, the molecules encoded by the two most prevalent A3-supertype alleles. The peptides that bind at least one of the two alleles with binding affinities of 500 nM, often 200 nM, are then tested for binding cross-reactivity to the other common A3-supertype alleles (e.g., A*3101, A*3301, and A*6801) to identify those that can bind at least three of the five HLA-A3-supertype molecules tested.
  • Selection of HLA-B7 Supermotif Bearing Epitopes
  • The 58P1D12 protein(s) scanned above is also analyzed for the presence of 8-, 9-10-, or 11-mer peptides with the HLA-B7-supermotif. Corresponding peptides are synthesized and tested for binding to HLA-B*0702, the molecule encoded by the most common B7-supertype allele (i.e., the prototype B7 supertype allele). Peptides binding B*0702 with IC50 of ≦500 nM are identified using standard methods. These peptides are then tested for binding to other common B7-supertype molecules (e.g., B*3501, B*5101, B*5301, and B*5401). Peptides capable of binding to three or more of the five B7-supertype alleles tested are thereby identified.
  • Selection of A1 and A24 Motif-Bearing Epitopes
  • To further increase population coverage, HLA-A1 and -A24 epitopes can also be incorporated into vaccine compositions. An analysis of the 58P1D12 protein can also be performed to identify HLA-A1- and A24-motif-containing sequences.
  • High affinity and/or cross-reactive binding epitopes that bear other motif and/or supermotifs are identified using analogous methodology.
  • Example 14 Confirmation of Immunogenicity
  • Cross-reactive candidate CTL A2-supermotif-bearing peptides that are identified as described herein are selected to confirm in vitro immunogenicity. Confirmation is performed using the following methodology:
  • Target Cell Lines for Cellular Screening:
  • The 0.221A2.1 cell line, produced by transferring the HLA-A2.1 gene into the HLA-A, -B, -C null mutant human B-lymphoblastoid cell line 721.221, is used as the peptide-loaded target to measure activity of HLA-A2.1-restricted CTL. This cell line is grown in RPMI-1640 medium supplemented with antibiotics, sodium pyruvate, nonessential amino acids and 10% (v/v) heat inactivated FCS. Cells that express an antigen of interest, or transfectants comprising the gene encoding the antigen of interest, can be used as target cells to confirm the ability of peptide-specific CTLs to recognize endogenous antigen.
  • Primary CTL Induction Cultures:
  • Generation of Dendritic Cells (DC): PBMCs are thawed in RPMI with 30 μg/ml DNAse, washed twice and resuspended in complete medium (RPMI-1640 plus 5% AB human serum, non-essential amino acids, sodium pyruvate, L-glutamine and penicillin/streptomycin). The monocytes are purified by plating 10×106 PBMC/well in a 6-well plate. After 2 hours at 37° C., the non-adherent cells are removed by gently shaking the plates and aspirating the supernatants. The wells are washed a total of three times with 3 ml RPMI to remove most of the non-adherent and loosely adherent cells. Three ml of complete medium containing 50 ng/ml of GM-CSF and 1,000 U/ml of IL-4 are then added to each well. TNFα is added to the DCs on day 6 at 75 ng/ml and the cells are used for CTL induction cultures on day 7.
  • Induction of CTL with DC and Peptide: CD8+ T-cells are isolated by positive selection with Dynal immunomagnetic beads (Dynabeads® M-450) and the Detacha-bead® reagent. Typically about 200−250×106 PBMC are processed to obtain 24×106 CD8+ T-cells (enough for a 48-well plate culture). Briefly, the PBMCs are thawed in RPMI with 30 μg/ml DNAse, washed once with PBS containing 1% human AB serum and resuspended in PBS/1% AB serum at a concentration of 20×106 cells/ml. The magnetic beads are washed 3 times with PBS/AB serum, added to the cells (140 μl beads/20×106 cells) and incubated for 1 hour at 4° C. with continuous mixing. The beads and cells are washed 4× with PBS/AB serum to remove the nonadherent cells and resuspended at 100×106 cells/ml (based on the original cell number) in PBS/AB serum containing 100 μl/ml Detacha-bead® reagent and 30 μg/ml DNAse. The mixture is incubated for 1 hour at room temperature with continuous mixing. The beads are washed again with PBS/AB/DNAse to collect the CD8+ T-cells. The DC are collected and centrifuged at 1300 rpm for 5-7 minutes, washed once with PBS with 1% BSA, counted and pulsed with 40 μg/ml of peptide at a cell concentration of 1-2×106/ml in the presence of 3 μg/ml β2-microglobulin for 4 hours at 20° C. The DC are then irradiated (4,200 rads), washed 1 time with medium and counted again.
  • Setting up induction cultures: 0.25 ml cytokine-generated DC (at 1×105 cells/ml) are co-cultured with 0.25 ml of CD8+ T-cells (at 2×106 cell/ml) in each well of a 48-well plate in the presence of 10 ng/ml of IL-7. Recombinant human IL-10 is added the next day at a final concentration of 10 ng/ml and rhuman IL-2 is added 48 hours later at 10 IU/ml.
  • Restimulation of the induction cultures with peptide-pulsed adherent cells: Seven and fourteen days after the primary induction, the cells are restimulated with peptide-pulsed adherent cells. The PBMCs are thawed and washed twice with RPMI and DNAse. The cells are resuspended at 5×106 cells/ml and irradiated at ˜4200 rads. The PBMCs are plated at 2×106 in 0.5 ml complete medium per well and incubated for 2 hours at 37° C. The plates are washed twice with RPMI by tapping the plate gently to remove the nonadherent cells and the adherent cells pulsed with 10 μg/ml of peptide in the presence of 3 μg/ml 132 microglobulin in 0.25 ml RPMI/5% AB per well for 2 hours at 37° C. Peptide solution from each well is aspirated and the wells are washed once with RPMI. Most of the media is aspirated from the induction cultures (CD8+ cells) and brought to 0.5 ml with fresh media. The cells are then transferred to the wells containing the peptide-pulsed adherent cells. Twenty four hours later recombinant human IL-10 is added at a final concentration of 10 ng/ml and recombinant human IL2 is added the next day and again 2-3 days later at 50 IU/ml (Tsai et al., Critical Reviews in Immunology 18(1-2):65-75, 1998). Seven days later, the cultures are assayed for CTL activity in a 51Cr release assay. In some experiments the cultures are assayed for peptide-specific recognition in the in situ IFNγ ELISA at the time of the second restimulation followed by assay of endogenous recognition 7 days later. After expansion, activity is measured in both assays for a side-by-side comparison.
  • Measurement of CTL Lytic Activity by 51Cr Release.
  • Seven days after the second restimulation, cytotoxicity is determined in a standard (5 hr) 51Cr release assay by assaying individual wells at a single E:T. Peptide-pulsed targets are prepared by incubating the cells with 10 μg/ml peptide overnight at 37° C.
  • Adherent target cells are removed from culture flasks with trypsin-EDTA. Target cells are labeled with 200 μCi of 51Cr sodium chromate (Dupont, Wilmington, Del.) for 1 hour at 37° C. Labeled target cells are resuspended at 106 per ml and diluted 1:10 with K562 cells at a concentration of 3.3×106/ml (an NK-sensitive erythroblastoma cell line used to reduce non-specific lysis). Target cells (100 μl) and effectors (100 μl) are plated in 96 well round-bottom plates and incubated for 5 hours at 37° C. At that time, 100 μl of supernatant are collected from each well and percent lysis is determined according to the formula:

  • [(cpm of the test sample−cpm of the spontaneous 51Cr release sample)/(cpm of the maximal 51Cr release sample−cpm of the spontaneous 51Cr release sample)]×100.
  • Maximum and spontaneous release are determined by incubating the labeled targets with 1% Triton X-100 and media alone, respectively. A positive culture is defined as one in which the specific lysis (sample-background) is 10% or higher in the case of individual wells and is 15% or more at the two highest E:T ratios when expanded cultures are assayed.
  • In situ Measurement of Human IFNγ Production as an Indicator of Peptide-Specific and Endogenous Recognition
  • Immulon 2 plates are coated with mouse anti-human IFNγ monoclonal antibody (4 μg/ml 0.1M NaHCO3, pH8.2) overnight at 4° C. The plates are washed with Ca2+, Mg2+-free PBS/0.05% Tween 20 and blocked with PBS/10% FCS for two hours, after which the CTLs (100 μl/well) and targets (100 μl/well) are added to each well, leaving empty wells for the standards and blanks (which received media only). The target cells, either peptide-pulsed or endogenous targets, are used at a concentration of 1×106 cells/ml. The plates are incubated for 48 hours at 37° C. with 5% CO2.
  • Recombinant human IFN-gamma is added to the standard wells starting at 400 pg or 1200 pg/100 microliter/well and the plate incubated for two hours at 37° C. The plates are washed and 100 μl of biotinylated mouse anti-human IFN-gamma monoclonal antibody (2 microgram/ml in PBS/3% FCS/0.05% Tween 20) are added and incubated for 2 hours at room temperature. After washing again, 100 microliter HRP-streptavidin (1:4000) are added and the plates incubated for one hour at room temperature. The plates are then washed 6× with wash buffer, 100 microliter/well developing solution (TMB 1:1) are added, and the plates allowed to develop for 5-15 minutes. The reaction is stopped with 50 microliter/well 1M H3PO4 and read at OD450. A culture is considered positive if it measured at least 50 pg of IFN-gamma/well above background and is twice the background level of expression.
  • CTL Expansion.
  • Those cultures that demonstrate specific lytic activity against peptide-pulsed targets and/or tumor targets are expanded over a two week period with anti-CD3. Briefly, 5×104 CD8+ cells are added to a T25 flask containing the following: 1×106 irradiated (4,200 rad) PBMC (autologous or allogeneic) per ml, 2×105 irradiated (8,000 rad) EBV-transformed cells per ml, and OKT3 (anti-CD3) at 30 ng per ml in RPMI-1640 containing 10% (v/v) human AB serum, non-essential amino acids, sodium pyruvate, 25 μM 2-mercaptoethanol, L-glutamine and penicillin/streptomycin. Recombinant human IL2 is added 24 hours later at a final concentration of 200 IU/ml and every three days thereafter with fresh media at 50 IU/ml. The cells are split if the cell concentration exceeds 1×106/ml and the cultures are assayed between days 13 and 15 at E:T ratios of 30, 10, 3 and 1:1 in the 51Cr release assay or at 1×106/ml in the in situ IFNγ assay using the same targets as before the expansion.
  • Cultures are expanded in the absence of anti-CD3+ as follows. Those cultures that demonstrate specific lytic activity against peptide and endogenous targets are selected and 5×104 CD8+ cells are added to a T25 flask containing the following: 1×106 autologous PBMC per ml which have been peptide-pulsed with 10 μg/ml peptide for two hours at 37° C. and irradiated (4,200 rad); 2×105 irradiated (8,000 rad) EBV-transformed cells per ml RPMI-1640 containing 10% (v/v) human AB serum, non-essential AA, sodium pyruvate, 25 mM 2-ME, L-glutamine and gentamicin.
  • Immunogenicity of A2 Supermotif-Bearing Peptides
  • A2-supermotif cross-reactive binding peptides are tested in the cellular assay for the ability to induce peptide-specific CTL in normal individuals. In this analysis, a peptide is typically considered to be an epitope if it induces peptide-specific CTLs in at least individuals, and preferably, also recognizes the endogenously expressed peptide.
  • Immunogenicity can also be confirmed using PBMCs isolated from patients bearing a tumor that expresses 58P1D12. Briefly, PBMCs are isolated from patients, re-stimulated with peptide-pulsed monocytes and assayed for the ability to recognize peptide-pulsed target cells as well as transfected cells endogenously expressing the antigen.
  • Evaluation of A*03/A11 Immunogenicity
  • HLA-A3 supermotif-bearing cross-reactive binding peptides are also evaluated for immunogenicity using methodology analogous for that used to evaluate the immunogenicity of the HLA-A2 supermotif peptides.
  • Evaluation of B7 Immunogenicity
  • Immunogenicity screening of the B7-supertype cross-reactive binding peptides identified as set forth herein are confirmed in a manner analogous to the confirmation of A2- and A3-supermotif-bearing peptides.
  • Peptides bearing other supermotifs/motifs, e.g., HLA-A1, HLA-A24 etc. are also confirmed using similar methodology
  • Example 15 Implementation of the Extended Supermotif to Improve the Binding Capacity of Native Epitopes by Creating Analogs
  • HLA motifs and supermotifs (comprising primary and/or secondary residues) are useful in the identification and preparation of highly cross-reactive native peptides, as demonstrated herein. Moreover, the definition of HLA motifs and supermotifs also allows one to engineer highly cross-reactive epitopes by identifying residues within a native peptide sequence which can be analoged to confer upon the peptide certain characteristics, e.g. greater cross-reactivity within the group of HLA molecules that comprise a supertype, and/or greater binding affinity for some or all of those HLA molecules. Examples of analoging peptides to exhibit modulated binding affinity are set forth in this example.
  • Analoging at Primary Anchor Residues
  • Peptide engineering strategies are implemented to further increase the cross-reactivity of the epitopes. For example, the main anchors of A2-supermotif-bearing peptides are altered, for example, to introduce a preferred L, I, V, or M at position 2, and I or V at the C-terminus.
  • To analyze the cross-reactivity of the analog peptides, each engineered analog is initially tested for binding to the prototype A2 supertype allele A*0201, then, if A*0201 binding capacity is maintained, for A2-supertype cross-reactivity.
  • Alternatively, a peptide is confirmed as binding one or all supertype members and then analoged to modulate binding affinity to any one (or more) of the supertype members to add population coverage.
  • The selection of analogs for immunogenicity in a cellular screening analysis is typically further restricted by the capacity of the parent wild type (WT) peptide to bind at least weakly, i.e., bind at an IC50 of 5000 nM or less, to three of more A2 supertype alleles. The rationale for this requirement is that the WT peptides must be present endogenously in sufficient quantity to be biologically relevant. Analoged peptides have been shown to have increased immunogenicity and cross-reactivity by T cells specific for the parent epitope (see, e.g., Parkhurst et al., J. Immunol. 157:2539, 1996; and Pogue et al., Proc. Natl. Acad. Sci. USA 92:8166, 1995).
  • In the cellular screening of these peptide analogs, it is important to confirm that analog-specific CTLs are also able to recognize the wild-type peptide and, when possible, target cells that endogenously express the epitope.
  • Analoging of HLA-A3 and B7-Supermotif-Bearing Peptides
  • Analogs of HLA-A3 supermotif-bearing epitopes are generated using strategies similar to those employed in analoging HLA-A2 supermotif-bearing peptides. For example, peptides binding to 3/5 of the A3-supertype molecules are engineered at primary anchor residues to possess a preferred residue (V, S, M, or A) at position 2.
  • The analog peptides are then tested for the ability to bind A*03 and A*11 (prototype A3 supertype alleles). Those peptides that demonstrate 500 nM binding capacity are then confirmed as having A3-supertype cross-reactivity.
  • Similarly to the A2- and A3-motif bearing peptides, peptides binding 3 or more B7-supertype alleles can be improved, where possible, to achieve increased cross-reactive binding or greater binding affinity or binding half life. B7 supermotif-bearing peptides are, for example, engineered to possess a preferred residue (V, I, L, or F) at the C-terminal primary anchor position, as demonstrated by Sidney et al. (J. Immunol. 157:3480-3490, 1996).
  • Analoging at primary anchor residues of other motif and/or supermotif-bearing epitopes is performed in a like manner
  • The analog peptides are then be confirmed for immunogenicity, typically in a cellular screening assay. Again, it is generally important to demonstrate that analog-specific CTLs are also able to recognize the wild-type peptide and, when possible, targets that endogenously express the epitope.
  • Analoging at Secondary Anchor Residues
  • Moreover, HLA supermotifs are of value in engineering highly cross-reactive peptides and/or peptides that bind HLA molecules with increased affinity by identifying particular residues at secondary anchor positions that are associated with such properties. For example, the binding capacity of a B7 supermotif-bearing peptide with an F residue at position 1 is analyzed. The peptide is then analoged to, for example, substitute L for F at position 1. The analoged peptide is evaluated for increased binding affinity, binding half life and/or increased cross-reactivity. Such a procedure identifies analoged peptides with enhanced properties.
  • Engineered analogs with sufficiently improved binding capacity or cross-reactivity can also be tested for immunogenicity in HLA-B7-transgenic mice, following for example, IFA immunization or lipopeptide immunization. Analoged peptides are additionally tested for the ability to stimulate a recall response using PBMC from patients with 58P1D12-expressing tumors.
  • Other Analoging Strategies
  • Another form of peptide analoging, unrelated to anchor positions, involves the substitution of a cysteine with α-amino butyric acid. Due to its chemical nature, cysteine has the propensity to form disulfide bridges and sufficiently alter the peptide structurally so as to reduce binding capacity. Substitution of α-amino butyric acid for cysteine not only alleviates this problem, but has been shown to improve binding and crossbinding capabilities in some instances (see, e.g., the review by Sette et al., In: Persistent Viral Infections, Eds. R. Ahmed and I. Chen, John Wiley & Sons, England, 1999).
  • Thus, by the use of single amino acid substitutions, the binding properties and/or cross-reactivity of peptide ligands for HLA supertype molecules can be modulated.
  • Example 16 Identification and Confirmation of 58P1D12-Derived Sequences with HLA-DR Binding Motifs
  • Peptide epitopes bearing an HLA class II supermotif or motif are identified and confirmed as outlined below using methodology similar to that described for HLA Class I peptides.
  • Selection of HLA-DR-Supermotif-Bearing Epitopes.
  • To identify 58P1D12-derived, HLA class II HTL epitopes, a 58P1D12 antigen is analyzed for the presence of sequences bearing an HLA-DR-motif or supermotif. Specifically, 15-mer sequences are selected comprising a DR-supermotif, comprising a 9-mer core, and three-residue N- and C-terminal flanking regions (15 amino acids total).
  • Protocols for predicting peptide binding to DR molecules have been developed (Southwood et al., J. Immunol. 160:3363-3373, 1998). These protocols, specific for individual DR molecules, allow the scoring, and ranking, of 9-mer core regions. Each protocol not only scores peptide sequences for the presence of DR-supermotif primary anchors (i.e., at position 1 and position 6) within a 9-mer core, but additionally evaluates sequences for the presence of secondary anchors. Using allele-specific selection tables (see, e.g., Southwood et al., ibid.), it has been found that these protocols efficiently select peptide sequences with a high probability of binding a particular DR molecule. Additionally, it has been found that performing these protocols in tandem, specifically those for DR1, DR4w4, and DR7, can efficiently select DR cross-reactive peptides.
  • The 58P1D12-derived peptides identified above are tested for their binding capacity for various common HLA-DR molecules. All peptides are initially tested for binding to the DR molecules in the primary panel: DR1, DR4w4, and DR7. Peptides binding at least two of these three DR molecules are then tested for binding to DR2w2β1, DR2w2 β2, DR6w19, and DR9 molecules in secondary assays. Finally, peptides binding at least two of the four secondary panel DR molecules, and thus cumulatively at least four of seven different DR molecules, are screened for binding to DR4w15, DR5w11, and DR8w2 molecules in tertiary assays. Peptides binding at least seven of the ten DR molecules comprising the primary, secondary, and tertiary screening assays are considered cross-reactive DR binders. 58P1D12-derived peptides found to bind common HLA-DR alleles are of particular interest.
  • Selection of DR3 motif Peptides
  • Because HLA-DR3 is an allele that is prevalent in Caucasian, Black, and Hispanic populations, DR3 binding capacity is a relevant criterion in the selection of HTL epitopes. Thus, peptides shown to be candidates may also be assayed for their DR3 binding capacity. However, in view of the binding specificity of the DR3 motif, peptides binding only to DR3 can also be considered as candidates for inclusion in a vaccine formulation.
  • To efficiently identify peptides that bind DR3, target 58P1D12 antigens are analyzed for sequences carrying one of the two DR3-specific binding motifs reported by Geluk et al. (J. Immunol. 152:5742-5748, 1994). The corresponding peptides are then synthesized and confirmed as having the ability to bind DR3 with an affinity of 1 μM or better, i.e., less than 1 μM. Peptides are found that meet this binding criterion and qualify as HLA class II high affinity binders.
  • DR3 binding epitopes identified in this manner are included in vaccine compositions with DR supermotif-bearing peptide epitopes.
  • Similarly to the case of HLA class I motif-bearing peptides, the class II motif-bearing peptides are analoged to improve affinity or cross-reactivity. For example, aspartic acid at position 4 of the 9-mer core sequence is an optimal residue for DR3 binding, and substitution for that residue often improves DR 3 binding.
  • Example 17 Immunogenicity of 58P1D12-Derived HTL Epitopes
  • This example determines immunogenic DR supermotif- and DR3 motif-bearing epitopes among those identified using the methodology set forth herein.
  • Immunogenicity of HTL epitopes are confirmed in a manner analogous to the determination of immunogenicity of CTL epitopes, by assessing the ability to stimulate HTL responses and/or by using appropriate transgenic mouse models Immunogenicity is determined by screening for: 1.) in vitro primary induction using normal PBMC or 2.) recall responses from patients who have 58P1D12-expressing tumors.
  • Example 18 Calculation of Phenotypic Frequencies of HLA-Supertypes in Various Ethnic Backgrounds to Determine Breadth of Population Coverage
  • This example illustrates the assessment of the breadth of population coverage of a vaccine composition comprised of multiple epitopes comprising multiple supermotifs and/or motifs.
  • In order to analyze population coverage, gene frequencies of HLA alleles are determined. Gene frequencies for each HLA allele are calculated from antigen or allele frequencies utilizing the binomial distribution formulae gf=1-(SQRT(1-af)) (see, e.g., Sidney et al., Human Immunol. 45:79-93, 1996). To obtain overall phenotypic frequencies, cumulative gene frequencies are calculated, and the cumulative antigen frequencies derived by the use of the inverse formula [af=1−(1−Cgf)2].
  • Where frequency data is not available at the level of DNA typing, correspondence to the serologically defined antigen frequencies is assumed. To obtain total potential supertype population coverage no linkage disequilibrium is assumed, and only alleles confirmed to belong to each of the supertypes are included (minimal estimates). Estimates of total potential coverage achieved by inter-loci combinations are made by adding to the A coverage the proportion of the non-A covered population that could be expected to be covered by the B alleles considered (e.g., total=A+B*(1-A)). Confirmed members of the A3-like supertype are A3, A11, A31, A*3301, and A*6801. Although the A3-like supertype may also include A34, A66, and A*7401, these alleles were not included in overall frequency calculations. Likewise, confirmed members of the A2-like supertype family are A*0201, A*0202, A*0203, A*0204, A*0205, A*0206, A*0207, A*6802, and A*6901. Finally, the B7-like supertype-confirmed alleles are: B7, B*3501-03, B51, B*5301, B*5401, B*5501-2, B*5601, B*6701, and B*7801 (potentially also B*1401, B*3504-06, B*4201, and B*5602).
  • Population coverage achieved by combining the A2-, A3- and B7-supertypes is approximately 86% in five major ethnic groups. Coverage may be extended by including peptides bearing the A1 and A24 motifs. On average, A1 is present in 12% and A24 in 29% of the population across five different major ethnic groups (Caucasian, North American Black, Chinese, Japanese, and Hispanic). Together, these alleles are represented with an average frequency of 39% in these same ethnic populations. The total coverage across the major ethnicities when A1 and A24 are combined with the coverage of the A2-, A3- and B7-supertype alleles is >95%, see, e.g., Table IV (G). An analogous approach can be used to estimate population coverage achieved with combinations of class II motif-bearing epitopes.
  • Immunogenicity studies in humans (e.g., Bertoni et al., J. Clin. Invest. 100:503, 1997; Doolan et al., Immunity 7:97, 1997; and Threlkeld et al., J. Immunol. 159:1648, 1997) have shown that highly cross-reactive binding peptides are almost always recognized as epitopes. The use of highly cross-reactive binding peptides is an important selection criterion in identifying candidate epitopes for inclusion in a vaccine that is immunogenic in a diverse population.
  • With a sufficient number of epitopes (as disclosed herein and from the art), an average population coverage is predicted to be greater than 95% in each of five major ethnic populations. The game theory Monte Carlo simulation analysis, which is known in the art (see e.g., Osborne, M.J. and Rubinstein, A. “A course in game theory” MIT Press, 1994), can be used to estimate what percentage of the individuals in a population comprised of the Caucasian, North American Black, Japanese, Chinese, and Hispanic ethnic groups would recognize the vaccine epitopes described herein. A preferred percentage is 90%. A more preferred percentage is 95%.
  • Example 19 CTL Recognition of Endogenously Processed Antigens After Priming
  • This example confirms that CTL induced by native or analoged peptide epitopes identified and selected as described herein recognize endogenously synthesized, i.e., native antigens.
  • Effector cells isolated from transgenic mice that are immunized with peptide epitopes, for example HLA-A2 supermotif-bearing epitopes, are re-stimulated in vitro using peptide-coated stimulator cells. Six days later, effector cells are assayed for cytotoxicity and the cell lines that contain peptide-specific cytotoxic activity are further re-stimulated. An additional six days later, these cell lines are tested for cytotoxic activity on 51Cr labeled Jurkat-A2.1/Kb target cells in the absence or presence of peptide, and also tested on 51Cr labeled target cells bearing the endogenously synthesized antigen, i.e. cells that are stably transfected with 58P1D12 expression vectors.
  • The results demonstrate that CTL lines obtained from animals primed with peptide epitope recognize endogenously synthesized 58P1D12 antigen. The choice of transgenic mouse model to be used for such an analysis depends upon the epitope(s) that are being evaluated. In addition to HLA-A*0201/Kb transgenic mice, several other transgenic mouse models including mice with human A11, which may also be used to evaluate A3 epitopes, and B7 alleles have been characterized and others (e.g., transgenic mice for HLA-A1 and A24) are being developed. HLA-DR1 and HLA-DR3 mouse models have also been developed, which may be used to evaluate HTL epitopes.
  • Example 20 Activity of CTL-HTL Conjugated Epitopes in Transgenic Mice
  • This example illustrates the induction of CTLs and HTLs in transgenic mice, by use of a 58P1D12-derived CTL and HTL peptide vaccine compositions. The vaccine composition used herein comprise peptides to be administered to a patient with a 58P1D12-expressing tumor. The peptide composition can comprise multiple CTL and/or HTL epitopes. The epitopes are identified using methodology as described herein. This example also illustrates that enhanced immunogenicity can be achieved by inclusion of one or more HTL epitopes in a CTL vaccine composition; such a peptide composition can comprise an HTL epitope conjugated to a CTL epitope. The CTL epitope can be one that binds to multiple HLA family members at an affinity of 500 nM or less, or analogs of that epitope. The peptides may be lipidated, if desired.
  • Immunization procedures: Immunization of transgenic mice is performed as described (Alexander et al., J. Immunol. 159:4753-4761, 1997). For example, A2/Kb mice, which are transgenic for the human HLA A2.1 allele and are used to confirm the immunogenicity of HLA-A*0201 motif- or HLA-A2 supermotif-bearing epitopes, and are primed subcutaneously (base of the tail) with a 0.1 ml of peptide in Incomplete Freund's Adjuvant, or if the peptide composition is a lipidated CTL/HTL conjugate, in DMSO/saline, or if the peptide composition is a polypeptide, in PBS or Incomplete Freund's Adjuvant. Seven days after priming, splenocytes obtained from these animals are restimulated with syngenic irradiated LPS-activated lymphoblasts coated with peptide.
  • Cell lines: Target cells for peptide-specific cytotoxicity assays are Jurkat cells transfected with the HLA-A2.1/Kb chimeric gene (e.g., Vitiello et al., J. Exp. Med. 173:1007, 1991)
  • In vitro CTL activation: One week after priming, spleen cells (30×106 cells/flask) are co-cultured at 37° C. with syngeneic, irradiated (3000 rads), peptide coated lymphoblasts (10×106 cells/flask) in 10 ml of culture medium/T25 flask. After six days, effector cells are harvested and assayed for cytotoxic activity.
  • Assay for cytotoxic activity: Target cells (1.0 to 1.5×106) are incubated at 37° C. in the presence of 200 μl of 51Cr. After 60 minutes, cells are washed three times and resuspended in R10 medium. Peptide is added where required at a concentration of 1 μg/ml. For the assay, 104 51Cr-labeled target cells are added to different concentrations of effector cells (final volume of 200 μl) in U-bottom 96-well plates. After a six hour incubation period at 37° C., a 0.1 ml aliquot of supernatant is removed from each well and radioactivity is determined in a Micromedic automatic gamma counter. The percent specific lysis is determined by the formula: percent specific release=100×(experimental release−spontaneous release)/(maximum release−spontaneous release). To facilitate comparison between separate CTL assays run under the same conditions, % 51Cr release data is expressed as lytic units/106 cells. One lytic unit is arbitrarily defined as the number of effector cells required to achieve 30% lysis of 10,000 target cells in a six hour 51Cr release assay. To obtain specific lytic units/106, the lytic units/106 obtained in the absence of peptide is subtracted from the lytic units/106 obtained in the presence of peptide. For example, if 30% 51Cr release is obtained at the effector (E): target (T) ratio of 50:1 (i.e., 5×105 effector cells for 10,000 targets) in the absence of peptide and 5:1 (i.e., 5×104 effector cells for 10,000 targets) in the presence of peptide, the specific lytic units would be: [(1/50,000)−(1/500,000)]×106=18 LU.
  • The results are analyzed to assess the magnitude of the CTL responses of animals injected with the immunogenic CTL/HTL conjugate vaccine preparation and are compared to the magnitude of the CTL response achieved using, for example, CTL epitopes as outlined above in the Example entitled “Confirmation of Immunogenicity.” Analyses similar to this may be performed to confirm the immunogenicity of peptide conjugates containing multiple CTL epitopes and/or multiple HTL epitopes. In accordance with these procedures, it is found that a CTL response is induced, and concomitantly that an HTL response is induced upon administration of such compositions.
  • Example 21 Selection of CTL and HTL Epitopes for Inclusion in a 58P1D12-Specific Vaccine
  • This example illustrates a procedure for selecting peptide epitopes for vaccine compositions of the invention. The peptides in the composition can be in the form of a nucleic acid sequence, either single or one or more sequences (i.e., minigene) that encodes peptide(s), or can be single and/or polyepitopic peptides.
  • The following principles are utilized when selecting a plurality of epitopes for inclusion in a vaccine composition. Each of the following principles is balanced in order to make the selection.
  • Epitopes are selected which, upon administration, mimic immune responses that are correlated with 58P1D12 clearance. The number of epitopes used depends on observations of patients who spontaneously clear 58P1D12. For example, if it has been observed that patients who spontaneously clear 58P1D12-expressing cells generate an immune response to at least three (3) epitopes from 58P1D12 antigen, then at least three epitopes should be included for HLA class I. A similar rationale is used to determine HLA class II epitopes.
  • Epitopes are often selected that have a binding affinity of an IC50 of 500 nM or less for an HLA class I molecule, or for class II, an IC50 of 1000 nM or less; or HLA Class I peptides with high binding scores from the BIMAS web site, at URL bimas.dcrt.nih gov/.
  • In order to achieve broad coverage of the vaccine through out a diverse population, sufficient supermotif bearing peptides, or a sufficient array of allele-specific motif bearing peptides, are selected to give broad population coverage. In one embodiment, epitopes are selected to provide at least 80% population coverage. A Monte Carlo analysis, a statistical evaluation known in the art, can be employed to assess breadth, or redundancy, of population coverage.
  • When creating polyepitopic compositions, or a minigene that encodes same, it is typically desirable to generate the smallest peptide possible that encompasses the epitopes of interest. The principles employed are similar, if not the same, as those employed when selecting a peptide comprising nested epitopes. For example, a protein sequence for the vaccine composition is selected because it has maximal number of epitopes contained within the sequence, i.e., it has a high concentration of epitopes. Epitopes may be nested or overlapping (i.e., frame shifted relative to one another). For example, with overlapping epitopes, two 9-mer epitopes and one 10-mer epitope can be present in a 10 amino acid peptide. Each epitope can be exposed and bound by an HLA molecule upon administration of such a peptide. A multi-epitopic, peptide can be generated synthetically, recombinantly, or via cleavage from the native source. Alternatively, an analog can be made of this native sequence, whereby one or more of the epitopes comprise substitutions that alter the cross-reactivity and/or binding affinity properties of the polyepitopic peptide. Such a vaccine composition is administered for therapeutic or prophylactic purposes. This embodiment provides for the possibility that an as yet undiscovered aspect of immune system processing will apply to the native nested sequence and thereby facilitate the production of therapeutic or prophylactic immune response-inducing vaccine compositions. Additionally such an embodiment provides for the possibility of motif-bearing epitopes for an HLA makeup that is presently unknown. Furthermore, this embodiment (absent the creating of any analogs) directs the immune response to multiple peptide sequences that are actually present in 58P1D12, thus avoiding the need to evaluate any junctional epitopes. Lastly, the embodiment provides an economy of scale when producing nucleic acid vaccine compositions. Related to this embodiment, computer programs can be derived in accordance with principles in the art, which identify in a target sequence, the greatest number of epitopes per sequence length.
  • A vaccine composition comprised of selected peptides, when administered, is safe, efficacious, and elicits an immune response similar in magnitude to an immune response that controls or clears cells that bear or overexpress 58P1D12.
  • Example 22 Construction of “Minigene” Multi-Epitope DNA Plasmids
  • This example discusses the construction of a minigene expression plasmid. Minigene plasmids may, of course, contain various configurations of B cell, CTL and/or HTL epitopes or epitope analogs as described herein.
  • A minigene expression plasmid typically includes multiple CTL and HTL peptide epitopes. In the present example, HLA-A2, -A3, -B7 supermotif-bearing peptide epitopes and HLA-A1 and -A24 motif-bearing peptide epitopes are used in conjunction with DR supermotif-bearing epitopes and/or DR3 epitopes. HLA class I supermotif or motif-bearing peptide epitopes derived 58P1D12, are selected such that multiple supermotifs/motifs are represented to ensure broad population coverage. Similarly, HLA class II epitopes are selected from 58P1D12 to provide broad population coverage, i.e. both HLA DR-1-4-7 supermotif-bearing epitopes and HLA DR-3 motif-bearing epitopes are selected for inclusion in the minigene construct. The selected CTL and HTL epitopes are then incorporated into a minigene for expression in an expression vector.
  • Such a construct may additionally include sequences that direct the HTL epitopes to the endoplasmic reticulum. For example, the Ii protein may be fused to one or more HTL epitopes as described in the art, wherein the CLIP sequence of the Ii protein is removed and replaced with an HLA class II epitope sequence so that HLA class II epitope is directed to the endoplasmic reticulum, where the epitope binds to an HLA class II molecules.
  • This example illustrates the methods to be used for construction of a minigene-bearing expression plasmid. Other expression vectors that may be used for minigene compositions are available and known to those of skill in the art.
  • The minigene DNA plasmid of this example contains a consensus Kozak sequence and a consensus murine kappa Ig-light chain signal sequence followed by CTL and/or HTL epitopes selected in accordance with principles disclosed herein. The sequence encodes an open reading frame fused to the Myc and His antibody epitope tag coded for by the pcDNA 3.1 Myc-His vector.
  • Overlapping oligonucleotides that can, for example, average about 70 nucleotides in length with 15 nucleotide overlaps, are synthesized and HPLC-purified. The oligonucleotides encode the selected peptide epitopes as well as appropriate linker nucleotides, Kozak sequence, and signal sequence. The final multiepitope minigene is assembled by extending the overlapping oligonucleotides in three sets of reactions using PCR. A Perkin/Elmer 9600 PCR machine is used and a total of 30 cycles are performed using the following conditions: 95° C. for 15 sec, annealing temperature (5° below the lowest calculated Tm of each primer pair) for 30 sec, and 72° C. for 1 min
  • For example, a minigene is prepared as follows. For a first PCR reaction, 5 μg of each of two oligonucleotides are annealed and extended: In an example using eight oligonucleotides, i.e., four pairs of primers, oligonucleotides 1+2, 3+4, 5+6, and 7+8 are combined in 100 μl reactions containing Pfu polymerase buffer (1×=10 mM KCL, 10 mM (NH4)2SO4, 20 mM Tris-chloride, pH 8.75, 2 mM MgSO4, 0.1% Triton X-100, 100 μg/ml BSA), 0.25 mM each dNTP, and 2.5 U of Pfu polymerase. The full-length dimer products are gel-purified, and two reactions containing the product of 1+2 and 3+4, and the product of 5+6 and 7+8 are mixed, annealed, and extended for 10 cycles. Half of the two reactions are then mixed, and 5 cycles of annealing and extension carried out before flanking primers are added to amplify the full length product. The full-length product is gel-purified and cloned into pCR-blunt (Invitrogen) and individual clones are screened by sequencing.
  • Example 23 The Plasmid Construct and the Degree to which it Induces Immunogenicity
  • The degree to which a plasmid construct, for example a plasmid constructed in accordance with the previous Example, is able to induce immunogenicity is confirmed in vitro by determining epitope presentation by APC following transduction or transfection of the APC with an epitope-expressing nucleic acid construct. Such a study determines “antigenicity” and allows the use of human APC. The assay determines the ability of the epitope to be presented by the APC in a context that is recognized by a T cell by quantifying the density of epitope-HLA class I complexes on the cell surface. Quantitation can be performed by directly measuring the amount of peptide eluted from the APC (see, e.g., Sijts et al., J. Immunol. 156:683-692, 1996; Demotz et al., Nature 342:682-684, 1989); or the number of peptide-HLA class I complexes can be estimated by measuring the amount of lysis or lymphokine release induced by diseased or transfected target cells, and then determining the concentration of peptide necessary to obtain equivalent levels of lysis or lymphokine release (see, e.g., Kageyama et al., J. Immunol. 154:567-576, 1995).
  • Alternatively, immunogenicity is confirmed through in vivo injections into mice and subsequent in vitro assessment of CTL and HTL activity, which are analyzed using cytotoxicity and proliferation assays, respectively, as detailed e.g., in Alexander et al., Immunity 1:751-761, 1994.
  • For example, to confirm the capacity of a DNA minigene construct containing at least one HLA-A2 supermotif peptide to induce CTLs in vivo, HLA-A2.1/Kb transgenic mice, for example, are immunized intramuscularly with 100 μg of naked cDNA. As a means of comparing the level of CTLs induced by cDNA immunization, a control group of animals is also immunized with an actual peptide composition that comprises multiple epitopes synthesized as a single polypeptide as they would be encoded by the minigene.
  • Splenocytes from immunized animals are stimulated twice with each of the respective compositions (peptide epitopes encoded in the minigene or the polyepitopic peptide), then assayed for peptide-specific cytotoxic activity in a 51Cr release assay. The results indicate the magnitude of the CTL response directed against the A2-restricted epitope, thus indicating the in vivo immunogenicity of the minigene vaccine and polyepitopic vaccine.
  • It is, therefore, found that the minigene elicits immune responses directed toward the HLA-A2 supermotif peptide epitopes as does the polyepitopic peptide vaccine. A similar analysis is also performed using other HLA-A3 and HLA-B7 transgenic mouse models to assess CTL induction by HLA-A3 and HLA-B7 motif or supermotif epitopes, whereby it is also found that the minigene elicits appropriate immune responses directed toward the provided epitopes.
  • To confirm the capacity of a class II epitope-encoding minigene to induce HTLs in vivo, DR transgenic mice, or for those epitopes that cross react with the appropriate mouse MHC molecule, I-Ab-restricted mice, for example, are immunized intramuscularly with 100 μg of plasmid DNA. As a means of comparing the level of HTLs induced by DNA immunization, a group of control animals is also immunized with an actual peptide composition emulsified in complete Freund's adjuvant. CD4+ T cells, i.e. HTLs, are purified from splenocytes of immunized animals and stimulated with each of the respective compositions (peptides encoded in the minigene). The HTL response is measured using a 3H-thymidine incorporation proliferation assay, (see, e.g., Alexander et al. Immunity 1:751-761, 1994). The results indicate the magnitude of the HTL response, thus demonstrating the in vivo immunogenicity of the minigene.
  • DNA minigenes, constructed as described in the previous Example, can also be confirmed as a vaccine in combination with a boosting agent using a prime boost protocol. The boosting agent can consist of recombinant protein (e.g., Barnett et al., Aids Res. and Human Retroviruses 14, Supplement 3:S299-S309, 1998) or recombinant vaccinia, for example, expressing a minigene or DNA encoding the complete protein of interest (see, e.g., Hanke et al., Vaccine 16:439-445, 1998; Sedegah et al., Proc. Natl. Acad. Sci. USA 95:7648-53, 1998; Hanke and McMichael, Immunol. Letters 66:177-181, 1999; and Robinson et al., Nature Med. 5:526-34, 1999).
  • For example, the efficacy of the DNA minigene used in a prime boost protocol is initially evaluated in transgenic mice. In this example, A2.1/Kb transgenic mice are immunized IM with 100 μg of a DNA minigene encoding the immunogenic peptides including at least one HLA-A2 supermotif-bearing peptide. After an incubation period (ranging from 3-9 weeks), the mice are boosted IP with 107 pfu/mouse of a recombinant vaccinia virus expressing the same sequence encoded by the DNA minigene. Control mice are immunized with 100 μg of DNA or recombinant vaccinia without the minigene sequence, or with DNA encoding the minigene, but without the vaccinia boost. After an additional incubation period of two weeks, splenocytes from the mice are immediately assayed for peptide-specific activity in an ELISPOT assay. Additionally, splenocytes are stimulated in vitro with the A2-restricted peptide epitopes encoded in the minigene and recombinant vaccinia, then assayed for peptide-specific activity in an alpha, beta and/or gamma IFN ELISA.
  • It is found that the minigene utilized in a prime-boost protocol elicits greater immune responses toward the HLA-A2 supermotif peptides than with DNA alone. Such an analysis can also be performed using HLA-A11 or HLA-B7 transgenic mouse models to assess CTL induction by HLA-A3 or HLA-B7 motif or supermotif epitopes. The use of prime boost protocols in humans is described below in the Example entitled “Induction of CTL Responses Using a Prime Boost Protocol.”
  • Example 24 Peptide Compositions for Prophylactic Uses
  • Vaccine compositions of the present invention can be used to prevent 58P1D12 expression in persons who are at risk for tumors that bear this antigen. For example, a polyepitopic peptide epitope composition (or a nucleic acid comprising the same) containing multiple CTL and HTL epitopes such as those selected in the above Examples, which are also selected to target greater than 80% of the population, is administered to individuals at risk for a 58P1D12-associated tumor.
  • For example, a peptide-based composition is provided as a single polypeptide that encompasses multiple epitopes. The vaccine is typically administered in a physiological solution that comprises an adjuvant, such as Incomplete Freunds Adjuvant. The dose of peptide for the initial immunization is from about 1 to about 50,000 μg, generally 100-5,000 μg, for a 70 kg patient. The initial administration of vaccine is followed by booster dosages at 4 weeks followed by evaluation of the magnitude of the immune response in the patient, by techniques that determine the presence of epitope-specific CTL populations in a PBMC sample. Additional booster doses are administered as required. The composition is found to be both safe and efficacious as a prophylaxis against 58P1D12-associated disease.
  • Alternatively, a composition typically comprising transfecting agents is used for the administration of a nucleic acid-based vaccine in accordance with methodologies known in the art and disclosed herein.
  • Example 25 Polyepitopic Vaccine Compositions Derived from Native 58P1D12 Sequences
  • A native 58P1D12 polyprotein sequence is analyzed, preferably using computer algorithms defined for each class I and/or class II supermotif or motif, to identify “relatively short” regions of the polyprotein that comprise multiple epitopes. The “relatively short” regions are preferably less in length than an entire native antigen. This relatively short sequence that contains multiple distinct or overlapping, “nested” epitopes can be used to generate a minigene construct. The construct is engineered to express the peptide, which corresponds to the native protein sequence. The “relatively short” peptide is generally less than 250 amino acids in length, often less than 100 amino acids in length, preferably less than 75 amino acids in length, and more preferably less than 50 amino acids in length. The protein sequence of the vaccine composition is selected because it has maximal number of epitopes contained within the sequence, i.e., it has a high concentration of epitopes. As noted herein, epitope motifs may be nested or overlapping (i.e., frame shifted relative to one another). For example, with overlapping epitopes, two 9-mer epitopes and one 10-mer epitope can be present in a 10 amino acid peptide. Such a vaccine composition is administered for therapeutic or prophylactic purposes.
  • The vaccine composition will include, for example, multiple CTL epitopes from 58P1D12 antigen and at least one HTL epitope. This polyepitopic native sequence is administered either as a peptide or as a nucleic acid sequence which encodes the peptide. Alternatively, an analog can be made of this native sequence, whereby one or more of the epitopes comprise substitutions that alter the cross-reactivity and/or binding affinity properties of the polyepitopic peptide.
  • The embodiment of this example provides for the possibility that an as yet undiscovered aspect of immune system processing will apply to the native nested sequence and thereby facilitate the production of therapeutic or prophylactic immune response-inducing vaccine compositions. Additionally, such an embodiment provides for the possibility of motif-bearing epitopes for an HLA makeup(s) that is presently unknown. Furthermore, this embodiment (excluding an analoged embodiment) directs the immune response to multiple peptide sequences that are actually present in native 58P1D12, thus avoiding the need to evaluate any junctional epitopes. Lastly, the embodiment provides an economy of scale when producing peptide or nucleic acid vaccine compositions.
  • Related to this embodiment, computer programs are available in the art which can be used to identify in a target sequence, the greatest number of epitopes per sequence length.
  • Example 26 Polyepitopic Vaccine Compositions from Multiple Antigens
  • The 58P1D12 peptide epitopes of the present invention are used in conjunction with epitopes from other target tumor-associated antigens, to create a vaccine composition that is useful for the prevention or treatment of cancer that expresses 58P1D12 and such other antigens. For example, a vaccine composition can be provided as a single polypeptide that incorporates multiple epitopes from 58P1D12 as well as tumor-associated antigens that are often expressed with a target cancer associated with 58P1D12 expression, or can be administered as a composition comprising a cocktail of one or more discrete epitopes. Alternatively, the vaccine can be administered as a minigene construct or as dendritic cells which have been loaded with the peptide epitopes in vitro.
  • Example 27 Use of Peptides to Evaluate an Immune Response
  • Peptides of the invention may be used to analyze an immune response for the presence of specific antibodies, CTL or HTL directed to 58P1D12. Such an analysis can be performed in a manner described by Ogg et al., Science 279:2103-2106, 1998. In this Example, peptides in accordance with the invention are used as a reagent for diagnostic or prognostic purposes, not as an immunogen.
  • In this example highly sensitive human leukocyte antigen tetrameric complexes (“tetramers”) are used for a cross-sectional analysis of, for example, 58P1D12 HLA-A*0201-specific CTL frequencies from HLA A*0201-positive individuals at different stages of disease or following immunization comprising a 58P1D12 peptide containing an A*0201 motif. Tetrameric complexes are synthesized as described (Musey et al., N. Engl. J. Med. 337:1267, 1997). Briefly, purified HLA heavy chain (A*0201 in this example) and β2-microglobulin are synthesized by means of a prokaryotic expression system. The heavy chain is modified by deletion of the transmembrane-cytosolic tail and COOH-terminal addition of a sequence containing a BirA enzymatic biotinylation site. The heavy chain, β2-microglobulin, and peptide are refolded by dilution. The 45-kD refolded product is isolated by fast protein liquid chromatography and then biotinylated by BirA in the presence of biotin (Sigma, St. Louis, Mo.), adenosine 5′ triphosphate and magnesium. Streptavidin-phycoerythrin conjugate is added in a 1:4 molar ratio, and the tetrameric product is concentrated to 1 mg/ml. The resulting product is referred to as tetramer-phycoerythrin.
  • For the analysis of patient blood samples, approximately one million PBMCs are centrifuged at 300 g for 5 minutes and resuspended in 50 μl of cold phosphate-buffered saline. Tri-color analysis is performed with the tetramer-phycoerythrin, along with anti-CD8-Tricolor, and anti-CD38. The PBMCs are incubated with tetramer and antibodies on ice for 30 to 60 min and then washed twice before formaldehyde fixation. Gates are applied to contain >99.98% of control samples. Controls for the tetramers include both A*0201-negative individuals and A*0201-positive non-diseased donors. The percentage of cells stained with the tetramer is then determined by flow cytometry. The results indicate the number of cells in the PBMC sample that contain epitope-restricted CTLs, thereby readily indicating the extent of immune response to the 58P1D12 epitope, and thus the status of exposure to 58P1D12, or exposure to a vaccine that elicits a protective or therapeutic response.
  • Example 28 Use of Peptide Epitopes to Evaluate Recall Responses
  • The peptide epitopes of the invention are used as reagents to evaluate T cell responses, such as acute or recall responses, in patients. Such an analysis may be performed on patients who have recovered from 58P1D12-associated disease or who have been vaccinated with a 58P1D12 vaccine.
  • For example, the class I restricted CTL response of persons who have been vaccinated may be analyzed. The vaccine may be any 58P1D12 vaccine. PBMC are collected from vaccinated individuals and HLA typed. Appropriate peptide epitopes of the invention that, optimally, bear supermotifs to provide cross-reactivity with multiple HLA supertype family members, are then used for analysis of samples derived from individuals who bear that HLA type.
  • PBMC from vaccinated individuals are separated on Ficoll-Histopaque density gradients (Sigma Chemical Co., St. Louis, Mo.), washed three times in HBSS (GIBCO Laboratories), resuspended in RPMI-1640 (GIBCO Laboratories) supplemented with L-glutamine (2 mM), penicillin (50 U/ml), streptomycin (50 μg/ml), and Hepes (10 mM) containing 10% heat-inactivated human AB serum (complete RPMI) and plated using microculture formats. A synthetic peptide comprising an epitope of the invention is added at 10 μg/ml to each well and HBV core 128-140 epitope is added at 1 μg/ml to each well as a source of T cell help during the first week of stimulation.
  • In the microculture format, 4×105 PBMC are stimulated with peptide in 8 replicate cultures in 96-well round bottom plate in 100 μl/well of complete RPMI. On days 3 and 10, 100 μl of complete RPMI and 20 U/ml final concentration of rIL-2 are added to each well. On day 7 the cultures are transferred into a 96-well flat-bottom plate and restimulated with peptide, rIL-2 and 105 irradiated (3,000 rad) autologous feeder cells. The cultures are tested for cytotoxic activity on day 14. A positive CTL response requires two or more of the eight replicate cultures to display greater than 10% specific 51Cr release, based on comparison with non-diseased control subjects as previously described (Rehermann, et al., Nature Med. 2:1104, 1108, 1996; Rehermann et al., J. Clin. Invest. 97:1655-1665, 1996; and Rehermann et al. J. Clin. Invest. 98:1432-1440, 1996).
  • Target cell lines are autologous and allogeneic EBV-transformed B-LCL that are either purchased from the American Society for Histocompatibility and Immunogenetics (ASHI, Boston, Mass.) or established from the pool of patients as described (Guilhot, et al. J. Virol. 66:2670-2678, 1992).
  • Cytotoxicity assays are performed in the following manner. Target cells consist of either allogeneic HLA-matched or autologous EBV-transformed B lymphoblastoid cell line that are incubated overnight with the synthetic peptide epitope of the invention at 10 μM, and labeled with 100 μCi of 51Cr (Amersham Corp., Arlington Heights, Ill.) for 1 hour after which they are washed four times with HBSS.
  • Cytolytic activity is determined in a standard 4-h, split well 51Cr release assay using U-bottomed 96 well plates containing 3,000 targets/well. Stimulated PBMC are tested at effector/target (E/T) ratios of 20-50:1 on day 14. Percent cytotoxicity is determined from the formula: 100×[(experimental release-spontaneous release)/maximum release-spontaneous release)]. Maximum release is determined by lysis of targets by detergent (2% Triton X-100; Sigma Chemical Co., St. Louis, Mo.). Spontaneous release is <25% of maximum release for all experiments.
  • The results of such an analysis indicate the extent to which HLA-restricted CTL populations have been stimulated by previous exposure to 58P1D12 or a 58P1D12 vaccine.
  • Similarly, Class II restricted HTL responses may also be analyzed. Purified PBMC are cultured in a 96-well flat bottom plate at a density of 1.5×105 cells/well and are stimulated with 10 μg/ml synthetic peptide of the invention, whole 58P1D12 antigen, or PHA. Cells are routinely plated in replicates of 4-6 wells for each condition. After seven days of culture, the medium is removed and replaced with fresh medium containing 10 U/ml IL-2. Two days later, 1 μCi 3H-thymidine is added to each well and incubation is continued for an additional 18 hours. Cellular DNA is then harvested on glass fiber mats and analyzed for 3H-thymidine incorporation. Antigen-specific T cell proliferation is calculated as the ratio of 3H-thymidine incorporation in the presence of antigen divided by the 3H-thymidine incorporation in the absence of antigen.
  • Example 29 Induction of Specific CTL Response In Humans
  • A human clinical trial for an immunogenic composition comprising CTL and HTL epitopes of the invention is set up as an IND Phase I, dose escalation study and carried out as a randomized, double-blind, placebo-controlled trial. Such a trial is designed, for example, as follows:
  • A total of about 27 individuals are enrolled and divided into 3 groups:
  • Group I: 3 subjects are injected with placebo and 6 subjects are injected with 5 μg of peptide composition;
  • Group II: 3 subjects are injected with placebo and 6 subjects are injected with 50 μg peptide composition;
  • Group III: 3 subjects are injected with placebo and 6 subjects are injected with 500 μg of peptide composition.
  • After 4 weeks following the first injection, all subjects receive a booster inoculation at the same dosage.
  • The endpoints measured in this study relate to the safety and tolerability of the peptide composition as well as its immunogenicity. Cellular immune responses to the peptide composition are an index of the intrinsic activity of this the peptide composition, and can therefore be viewed as a measure of biological efficacy. The following summarize the clinical and laboratory data that relate to safety and efficacy endpoints.
  • Safety: The incidence of adverse events is monitored in the placebo and drug treatment group and assessed in terms of degree and reversibility.
  • Evaluation of Vaccine Efficacy: For evaluation of vaccine efficacy, subjects are bled before and after injection. Peripheral blood mononuclear cells are isolated from fresh heparinized blood by Ficoll-Hypaque density gradient centrifugation, aliquoted in freezing media and stored frozen. Samples are assayed for CTL and HTL activity.
  • The vaccine is found to be both safe and efficacious.
  • Example 30 Phase II Trials In Patients Expressing 58P1D12
  • Phase II trials are performed to study the effect of administering the CTL-HTL peptide compositions to patients having cancer that expresses 58P1D12. The main objectives of the trial are to determine an effective dose and regimen for inducing CTLs in cancer patients that express 58P1D12, to establish the safety of inducing a CTL and HTL response in these patients, and to see to what extent activation of CTLs improves the clinical picture of these patients, as manifested, e.g., by the reduction and/or shrinking of lesions. Such a study is designed, for example, as follows:
  • The studies are performed in multiple centers. The trial design is an open-label, uncontrolled, dose escalation protocol wherein the peptide composition is administered as a single dose followed six weeks later by a single booster shot of the same dose. The dosages are 50, 500 and 5,000 micrograms per injection. Drug-associated adverse effects (severity and reversibility) are recorded.
  • There are three patient groupings. The first group is injected with 50 micrograms of the peptide composition and the second and third groups with 500 and 5,000 micrograms of peptide composition, respectively. The patients within each group range in age from 21-65 and represent diverse ethnic backgrounds. All of them have a tumor that expresses 58P1D12.
  • Clinical manifestations or antigen-specific T-cell responses are monitored to assess the effects of administering the peptide compositions. The vaccine composition is found to be both safe and efficacious in the treatment of 58P1D12-associated disease.
  • Example 31 Induction of CTL Responses Using a Prime Boost Protocol
  • A prime boost protocol similar in its underlying principle to that used to confirm the efficacy of a DNA vaccine in transgenic mice, such as described above in the Example entitled “The Plasmid Construct and the Degree to Which It Induces Immunogenicity,” can also be used for the administration of the vaccine to humans. Such a vaccine regimen can include an initial administration of, for example, naked DNA followed by a boost using recombinant virus encoding the vaccine, or recombinant protein/polypeptide or a peptide mixture administered in an adjuvant.
  • For example, the initial immunization may be performed using an expression vector, such as that constructed in the Example entitled “Construction of “Minigene” Multi-Epitope DNA Plasmids” in the form of naked nucleic acid administered IM (or SC or ID) in the amounts of 0.5-5 mg at multiple sites. The nucleic acid (0.1 to 1000 μg) can also be administered using a gene gun. Following an incubation period of 3-4 weeks, a booster dose is then administered. The booster can be recombinant fowlpox virus administered at a dose of 5-107 to 5×109 pfu. An alternative recombinant virus, such as an MVA, canarypox, adenovirus, or adeno-associated virus, can also be used for the booster, or the polyepitopic protein or a mixture of the peptides can be administered. For evaluation of vaccine efficacy, patient blood samples are obtained before immunization as well as at intervals following administration of the initial vaccine and booster doses of the vaccine. Peripheral blood mononuclear cells are isolated from fresh heparinized blood by Ficoll-Hypaque density gradient centrifugation, aliquoted in freezing media and stored frozen. Samples are assayed for CTL and HTL activity.
  • Analysis of the results indicates that a magnitude of response sufficient to achieve a therapeutic or protective immunity against 58P1D12 is generated.
  • Example 32 Administration of Vaccine Compositions Using Dendritic Cells (DC)
  • Vaccines comprising peptide epitopes of the invention can be administered using APCs, or “professional” APCs such as DC. In this example, peptide-pulsed DC are administered to a patient to stimulate a CTL response in vivo. In this method, dendritic cells are isolated, expanded, and pulsed with a vaccine comprising peptide CTL and HTL epitopes of the invention. The dendritic cells are infused back into the patient to elicit CTL and HTL responses in vivo. The induced CTL and HTL then destroy or facilitate destruction, respectively, of the target cells that bear the 58P1D12 protein from which the epitopes in the vaccine are derived.
  • For example, a cocktail of epitope-comprising peptides is administered ex vivo to PBMC, or isolated DC therefrom. A pharmaceutical to facilitate harvesting of DC can be used, such as Progenipoietin™ (Monsanto, St. Louis, Mo.) or GM-CSF/IL-4. After pulsing the DC with peptides, and prior to reinfusion into patients, the DC are washed to remove unbound peptides.
  • As appreciated clinically, and readily determined by one of skill based on clinical outcomes, the number of DC reinfused into the patient can vary (see, e.g., Nature Med. 4:328, 1998; Nature Med. 2:52, 1996 and Prostate 32:272, 1997). Although 2-50×106 DC per patient are typically administered, larger number of DC, such as 107 or 108 can also be provided. Such cell populations typically contain between 50-90% DC.
  • In some embodiments, peptide-loaded PBMC are injected into patients without purification of the DC. For example, PBMC generated after treatment with an agent such as Progenipoietin™ are injected into patients without purification of the DC. The total number of PBMC that are administered often ranges from 108 to 1010. Generally, the cell doses injected into patients is based on the percentage of DC in the blood of each patient, as determined, for example, by immunofluorescence analysis with specific anti-DC antibodies. Thus, for example, if Progenipoietin™ mobilizes 2% DC in the peripheral blood of a given patient, and that patient is to receive 5×106 DC, then the patient will be injected with a total of 2.5×108 peptide-loaded PBMC. The percent DC mobilized by an agent such as Progenipoietin™ is typically estimated to be between 2-10%, but can vary as appreciated by one of skill in the art.
  • Ex Vivo Activation of CTL/HTL Responses
  • Alternatively, ex vivo CTL or HTL responses to 58P1D12 antigens can be induced by incubating, in tissue culture, the patient's, or genetically compatible, CTL or HTL precursor cells together with a source of APC, such as DC, and immunogenic peptides. After an appropriate incubation time (typically about 7-28 days), in which the precursor cells are activated and expanded into effector cells, the cells are infused into the patient, where they will destroy (CTL) or facilitate destruction (HTL) of their specific target cells, i.e., tumor cells.
  • Example 33 An Alternative Method of Identifying and Confirming Motif-Bearing Peptides
  • Another method of identifying and confirming motif-bearing peptides is to elute them from cells bearing defined MHC molecules. For example, EBV transformed B cell lines used for tissue typing have been extensively characterized to determine which HLA molecules they express. In certain cases these cells express only a single type of HLA molecule. These cells can be transfected with nucleic acids that express the antigen of interest, e.g. 58P1D12. Peptides produced by endogenous antigen processing of peptides produced as a result of transfection will then bind to HLA molecules within the cell and be transported and displayed on the cell's surface. Peptides are then eluted from the HLA molecules by exposure to mild acid conditions and their amino acid sequence determined, e.g., by mass spectral analysis (e.g., Kubo et al., J. Immunol. 152:3913, 1994). Because the majority of peptides that bind a particular HLA molecule are motif-bearing, this is an alternative modality for obtaining the motif-bearing peptides correlated with the particular HLA molecule expressed on the cell.
  • Alternatively, cell lines that do not express endogenous HLA molecules can be transfected with an expression construct encoding a single HLA allele. These cells can then be used as described, i.e., they can then be transfected with nucleic acids that encode 58P1D12 to isolate peptides corresponding to 58P1D12 that have been presented on the cell surface. Peptides obtained from such an analysis will bear motif(s) that correspond to binding to the single HLA allele that is expressed in the cell.
  • As appreciated by one in the art, one can perform a similar analysis on a cell bearing more than one HLA allele and subsequently determine peptides specific for each HLA allele expressed. Moreover, one of skill would also recognize that means other than transfection, such as loading with a protein antigen, can be used to provide a source of antigen to the cell.
  • Example 34 Complementary Polynucleotides
  • Sequences complementary to the 58P1D12-encoding sequences, or any parts thereof, are used to detect, decrease, or inhibit expression of naturally occurring 58P1D12. Although use of oligonucleotides comprising from about 15 to 30 base pairs is described, essentially the same procedure is used with smaller or with larger sequence fragments. Appropriate oligonucleotides are designed using, e.g., OLIGO 4.06 software (National Biosciences) and the coding sequence of 58P1D12. To inhibit transcription, a complementary oligonucleotide is designed from the most unique 5′ sequence and used to prevent promoter binding to the coding sequence. To inhibit translation, a complementary oligonucleotide is designed to prevent ribosomal binding to a 58P1D12-encoding transcript.
  • Example 35 Purification of Naturally-Occurring or Recombinant 58P1D12 Using 58P1D12-Specific Antibodies
  • Naturally occurring or recombinant 58P1D12 is substantially purified by immunoaffinity chromatography using antibodies specific for 58P1D12. An immunoaffinity column is constructed by covalently coupling anti-58P1D12 antibody to an activated chromatographic resin, such as CNBr-activated SEPHAROSE (Amersham Pharmacia Biotech). After the coupling, the resin is blocked and washed according to the manufacturer's instructions.
  • Media containing 58P1D12 are passed over the immunoaffinity column, and the column is washed under conditions that allow the preferential absorbance of 58P1D12 (e.g., high ionic strength buffers in the presence of detergent). The column is eluted under conditions that disrupt antibody/58P1D12 binding (e.g., a buffer of pH 2 to pH 3, or a high concentration of a chaotrope, such as urea or thiocyanate ion), and GCR.P is collected.
  • Example 36 Identification of Molecules Which Interact with 58P1D12
  • 58P1D12, or biologically active fragments thereof, are labeled with 121 l Bolton-Hunter reagent. (See, e.g., Bolton et al. (1973) Biochem. J. 133:529.) Candidate molecules previously arrayed in the wells of a multi-well plate are incubated with the labeled 58P1D12, washed, and any wells with labeled 58P1D12 complex are assayed. Data obtained using different concentrations of 58P1D12 are used to calculate values for the number, affinity, and association of 58P1D12 with the candidate molecules.
  • Example 37 In Vivo Assay for 58P1D12 Tumor Growth Promotion
  • In vivo assay of 3T3 cell growth by recombinant expression of 58P1D12.
  • To address the potential for 58P1D12 to accelerate the growth of non-tumorigenic cells in an in vivo mouse model, non-transformed 3T3 cells were prepared by infection with either a virus containing an empty vector control (Neo gene alone) or with a vector containing the 58P1D12 full-length gene. 3T3 cells were selected for survival in G-418, and expression of 58P1D12 confirmed by Northern blot analysis. To assess the growth potential of these cells, 1×106 58P1D12 expressing 3T3 cells or 1×106 Neo control were mixed with Matrigel®, then injected intratibially in SCID mice and allowed to grow for 31 days. The growth of these cells was assessed on day 31 by visual inspection and by necropsy (FIG. 4). Clearly, the 58P1D12 expressing 3T3 cells showed a potent effect in comparison to the 3T3-Neo cells, indicating that the 58P1D12 protein enhanced the growth of the cells in Matrigel®. These data indicate that 58P1D12 promotes the growth of non-tumorigenic cells and provides a growth advantage in vivo that mimics the role of this protein in human malignancies.
  • Example 38 58P1D12 Monoclonal Antibody-mediated Inhibition of Bladder, Prostate, and Ovarian Tumors In Vivo
  • The significant expression of 58P1D12 in cancer tissues, together with its restrictive expression in normal tissues makes 58P1D12 a good target for antibody therapy. Similarly, 58P1D12 is a target for T cell-based immunotherapy. Thus, the therapeutic efficacy of anti-58P1D12 MAbs in human prostate cancer xenograft mouse models is evaluated by using recombinant cell lines such as PC3-58P1D12, and 3T3-58P1D12 (see, e.g., Kaighn, M. E., et al., Invest Urol, 1979. 17(1): p. 16-23), as well as human prostate xenograft models such as LAPC9 (Saffran et al, Proc Natl Acad Sci USA. 2001, 98:2658). Similarly, anti-58P1D12 MAbs are evaluated in human bladder cancer xenograft models using recombinant cell lines such as J82-58P1D12, kidney cancer xenograft models using 769-P cells, or ovarian cancer xenograft models using recombinant cell lines such as OVCAR-58P1D12.
  • Antibody efficacy on tumor growth and metastasis formation is studied, e.g., in a mouse orthotopic bladder cancer xenograft model, and a mouse prostate cancer xenograft model. The antibodies can be unconjugated, as discussed in this Example, or can be conjugated to a therapeutic modality (see below), as appreciated in the art. Anti-58P1D12 MAbs inhibit formation of prostate and bladder xenografts. Anti-58P1D12 MAbs also retard the growth of established orthotopic tumors and prolonged survival of tumor-bearing mice. These results indicate the utility of anti-58P1D12 MAbs in the treatment of local and advanced stages of prostate and bladder cancer (see, e.g., Saffran, D., 2001, et al., PNAS 10:1073-1078 or on the World Wide Web address at pnas.org/cgi/doi/10.1073/pnas.051624698).
  • Administration of the anti-58P1D12 MAbs led to retardation of established orthotopic tumor growth and inhibition of metastasis to distant sites, resulting in a significant prolongation in the survival of tumor-bearing mice. These studies indicate that 58P1D12 is an attractive target for immunotherapy and demonstrate the therapeutic potential of anti-58P1D12 MAbs for the treatment of local and metastatic cancer. This example demonstrates that unconjugated 58P1D12 monoclonal antibodies are effective to inhibit the growth of human bladder and prostate tumor xenografts grown in SCID mice; accordingly, a combination of such efficacious MAbs is also effective.
  • MAb-toxin conjugates: Another embodiment of MAb therapy is through the use of toxin conjugation of MAbs for targeted delivery of cytotoxic agents to cells expressing the protein target. Major advances have been made in the clinical application of MAb toxin conjugates with the development of Mylotarg for acute myeloid leukemia (Bross, P. F., et al., 2001, Clin. Cancer Res. 7:1490-1496). Mylotarg is a humanized MAb directed to CD33 which is conjugated to a highly potent DNA-alkylating agent (calichemicin) via an acid labile hydrazone bond (Hamann, P. R., et al., 2002, Bioconjug. Chem. 13:40-46; ibid., 13:47-58). Additional toxins for MAb conjugation in development include maytansinoid, doxorubicin, taxoids and the potent synthetic dolastatin 10 analogs auristatin E and monomethylauristatin E (Doronina, S. O., et al., 2003, Nature Biotech. 21:778-784; Ross, S., et al., 2002, Cancer Res. 62:2546-2553; Francisco, J. A., et el., 2003, Blood 102 :1458-1465; Mao, W., et al., 2004, Cancer Res. 64:781-788). Such applications have potential to deliver a cytotoxic agent to cells expressing the protein target of the MAb. Internalization of the target protein upon MAb binding is important for toxin delivery, and the mechanism spares the non-targeted tissues from the potentially harmful effects of the cytotoxic agent.
  • 58P1D12 MAbs conjugated to toxins are used to induce cell killing in vitro using established protocols for cytotoxicity assays and clonogenic assays (Doronina, S. O., et al., 2003, Nature Biotech. 21:778-784; Mao, W., et al., 2004, Cancer Res. 64:781-788). Toxin conjugated anti-58P1D12 MAbs induce cytotoxicity of cells expressing endogenous 58P1D12 (769-P cells) and recombinant 58P1D12 (PC3-58P1D12, 3T3-58P1D12, Rat-1-58P1D12 and B300.19-58P1D12). This methodology allows confirmation that the toxin conjugated MAb is functional against cells expressing the 58P1D12 protein on their surface versus those that do not express the target.
  • The MAb toxin conjugates are tested for their ability to inhibit tumor growth in vivo. Antibody efficacy on tumor growth and metastasis formation is studied, e.g., in a mouse orthotopic bladder cancer xenograft model, a mouse prostate cancer xenograft model, or mouse ovarian cancer xenograft model. Administration of the anti-58P1D12 MAbs led to retardation of established orthotopic tumor growth and inhibition of metastasis to distant sites, resulting in a significant prolongation in the survival of tumor-bearing mice. These studies indicate that 58P1D12 is an attractive target for immunotherapy and demonstrate the therapeutic potential of toxin-conjugated anti-58P1D12 MAbs for the treatment of local and metastatic cancer. This example demonstrates that toxin-conjugated 58P1D12 monoclonal antibodies are effective to inhibit the growth of human bladder, prostate and ovarian tumor xenografts grown in SCID mice; accordingly, a combination of such efficacious MAbs is also effective. The methodology allows the targeted delivery of a cytotoxin using a plasma stable linker in a MAb-toxin conjugate. Such a mechanism of action reduces the potential harmful effects of the toxin on non-targeted tissues.
  • Tumor Inhibition Using Multiple Unconjugated or Toxin-Conjugated 58P1D12 MAbs
  • Materials and Methods
  • 58P1D12 Monoclonal Antibodies:
  • Monoclonal antibodies are raised against 58P1D12 as described in the Example entitled “Generation of 58P1D12 Monoclonal Antibodies (MAbs).” The antibodies are characterized by ELISA, Western blot, FACS, and immunoprecipitation for their capacity to bind 58P1D12. Epitope mapping data for the anti-58P1D12 MAbs, as determined by ELISA and Western analysis, recognize epitopes on the 58P1D12 protein Immunohistochemical analysis of prostate cancer tissues and cells with these antibodies is performed.
  • The monoclonal antibodies are purified from ascites or hybridoma tissue culture supernatants by Protein-G Sepharose chromatography, dialyzed against PBS, filter sterilized, and stored at −20° C. Protein determinations are performed by a Bradford assay (Bio-Rad, Hercules, Calif.). A therapeutic monoclonal antibody or a cocktail comprising a mixture of individual monoclonal antibodies is prepared and used for the treatment of mice receiving subcutaneous or orthotopic injections of SCABER, J82, A498, 769-P, CaOvl, OVCAR or PA1 tumor xenografts.
  • The MAbs to 58P1D12 are conjugated to various different toxins (listed above) using any of a variety of methods described elsewhere in the art (Hamann, P. R., et al., 2002, Bioconjug. Chem. 13:40-46; ibid., 13:47-58; Doronina, S. O., et al., 2003, Nature Biotech. 21:778-784; Ojima, I., et al. 2002, J. Med. Chem. 45:5620-5623; Dubowchik, G. M., et al., 2002, Bioconjug. Chem. 13:855-869; King, H. D., 2002, J. Med. Chem. 45:4336-4343; Ross, S., et al., 2002, Cancer Res. 62:2546-2553; Francisco, J. A., et el., 2003, Blood 102 :1458-1465; Mao, W., et al., 2004, Cancer Res. 64:781-788).
  • Cell Lines
  • The bladder and prostate carcinoma cell lines, J82 and PC3 as well as the fibroblast line NIH 3T3 (American Type Culture Collection) are maintained in media supplemented with L-glutamine and 10% FBS. J82-58P1D12, PC3-58P1D12 and 3T3-58P1D12 cell populations are generated by retroviral gene transfer as described in Hubert, R. S., et al., Proc. Natl. Acad. Sci. USA, 1999, 96(25):14523.
  • Xenograft Mouse Models
  • Subcutaneous (s.c.) tumors are generated by injection of 1×106 cancer cells mixed at a 1:1 dilution with Matrigel® (Collaborative Research) in the right flank of male SCID mice. To test antibody efficacy on tumor formation, i.p. antibody injections are started on the same day as tumor-cell injections. As a control, mice are injected with either purified mouse IgG (ICN) or PBS; or a purified monoclonal antibody that recognizes an irrelevant protein not expressed in human cells. Tumor sizes are determined by caliper measurements, and the tumor volume is calculated as: Length×Width×Height. Mice with s.c. tumors greater than 1.5 cm in diameter are sacrificed.
  • Orthotopic injections are performed under anesthesia by using ketamine/xylazine. For bladder orthotopic studies, an incision is made through the abdomen to expose the bladder, and tumor cells (5×105) mixed with Matrigel® are injected into the bladder wall in a 10-μl volume. To monitor tumor growth, mice are palpated and blood is collected on a weekly basis to measure BTA levels. For prostate orthopotic models, an incision is made through the abdominal muscles to expose the bladder and seminal vesicles, which then are delivered through the incision to expose the dorsal prostate. Tumor cells, e.g. LAPC-9 cells (5×105) mixed with Matrigel® are injected into the prostate in a 10-μl volume (Yoshida Y et al, Anticancer Res. 1998, 18:327; Ahn et al, Tumour Biol. 2001, 22:146). To monitor tumor growth, blood is collected on a weekly basis measuring PSA levels. The mice are segregated into groups for the appropriate treatments, with anti-58P1D12 or control MAbs being injected i.p.
  • Anti-58P1D12 MAbs Inhibit Growth of 58P1D12-Expressing Xenograft-Cancer Tumors
  • The effect of anti-58P1D12 MAbs on tumor formation is tested on the growth and progression of bladder, and prostate cancer xenografts using J82-58P1D12, and PC3-58P1D12 orthotopic models. As compared with the s.c. tumor model, the orthotopic model, which requires injection of tumor cells directly in the mouse bladder, and prostate, respectively, results in a local tumor growth, development of metastasis in distal sites, deterioration of mouse health, and subsequent death (Saffran, D., et al., PNAS supra; Fu, X., et al., Int J Cancer, 1992. 52(6): p. 987-90; Kubota, T., J Cell Biochem, 1994. 56(1): p. 4-8). The features make the orthotopic model more representative of human disease progression and allowed us to follow the therapeutic effect of MAbs on clinically relevant end points.
  • Accordingly, tumor cells are injected into the mouse bladder, or prostate, and 2 days later, the mice are segregated into two groups and treated with either: a) 200-500 μg, of anti-58P1D12 Ab, or b) PBS three times per week for two to five weeks.
  • A major advantage of the orthotopic cancer models is the ability to study the development of metastases. Formation of metastasis in mice bearing established orthotopic tumors is studies by IHC analysis on lung sections using an antibody against a tumor-specific cell-surface protein such as anti-CK20 for bladder cancer, anti-STEAP-1 for prostate cancer models (Lin S et al, Cancer Detect Prey. 2001; 25:202; Saffran, D., et al., PNAS supra).
  • Mice bearing established orthotopic tumors are administered 1000 μg injections of either anti-58P1D12 MAb or PBS over a 4-week period. Mice in both groups are allowed to establish a high tumor burden, to ensure a high frequency of metastasis formation in mouse lungs. Mice then are killed and their bladders, livers, bone and lungs are analyzed for the presence of tumor cells by IHC analysis.
  • These studies demonstrate a broad anti-tumor efficacy of anti-58P1D12 antibodies on initiation and progression of prostate, kidney, bladder and ovarian cancer in xenograft mouse models. Anti-58P1D12 antibodies inhibit formation of tumors as well as retarding the growth of already established tumors and prolong the survival of treated mice. Moreover, anti-58P1D12 MAbs demonstrate a dramatic inhibitory effect on the spread of local bladder and prostate tumor to distal sites, even in the presence of a large tumor burden. Thus, anti-58P1D12 MAbs are efficacious on major clinically relevant end points (tumor growth), prolongation of survival, and health.
  • Example 39 Therapeutic and Diagnostic use of Anti-58P1D12 Antibodies in Humans
  • Anti-58P1D12 monoclonal antibodies are safely and effectively used for diagnostic, prophylactic, prognostic and/or therapeutic purposes in humans. Western blot and immunohistochemical analysis of cancer tissues and cancer xenografts with anti-58P1D12 mAb show strong extensive staining in carcinoma but significantly lower or undetectable levels in normal tissues. Detection of 58P1D12 in carcinoma and in metastatic disease demonstrates the usefulness of the mAb as a diagnostic and/or prognostic indicator. Anti-58P1D12 antibodies are therefore used in diagnostic applications such as immunohistochemistry of kidney biopsy specimens to detect cancer from suspect patients.
  • As determined by flow cytometry, anti-58P1D12 mAb specifically binds to carcinoma cells. Thus, anti-58P1D12 antibodies are used in diagnostic whole body imaging applications, such as radioimmunoscintigraphy and radioimmunotherapy, (see, e.g., Potamianos S., et. al. Anticancer Res 20(2A):925-948 (2000)) for the detection of localized and metastatic cancers that exhibit expression of 58P1D12. Shedding or release of an extracellular domain of 58P1D12 into the extracellular milieu, such as that seen for alkaline phosphodiesterase B10 (Meerson, N. R., Hepatology 27:563-568 (1998)), allows diagnostic detection of 58P1D12 by anti-58P1D12 antibodies in serum and/or urine samples from suspect patients.
  • Anti-58P1D12 antibodies that specifically bind 58P1D12 are used in therapeutic applications for the treatment of cancers that express 58P1D12. Anti-58P1D12 antibodies are used as an unconjugated modality and as conjugated form in which the antibodies are attached to one of various therapeutic or imaging modalities well known in the art, such as a prodrugs, enzymes or radioisotopes. In preclinical studies, unconjugated and conjugated anti-58P1D12 antibodies are tested for efficacy of tumor prevention and growth inhibition in the SCID mouse cancer xenograft models, e.g., kidney cancer models AGS-K3 and AGS-K6, (see, e.g., the Example entitled “58P1D12 Monoclonal Antibody-mediated Inhibition of Bladder and Lung Tumors In Vivo”). Either conjugated and unconjugated anti-58P1D12 antibodies are used as a therapeutic modality in human clinical trials either alone or in combination with other treatments as described in following Examples.
  • Example 40 Human Clinical Trials for the Treatment and Diagnosis of Human Carcinomas Through Use of Human Anti-58P1D12 Antibodies In Vivo
  • Antibodies are used in accordance with the present invention which recognize an epitope on 58P1D12, and are used in the treatment of certain tumors such as those listed in Table I. Based upon a number of factors, including 58P1D12 expression levels, tumors such as those listed in Table I are presently preferred indications. In connection with each of these indications, three clinical approaches are successfully pursued.
  • I.) Adjunctive therapy: In adjunctive therapy, patients are treated with anti-58P1D12 antibodies in combination with a chemotherapeutic or antineoplastic agent and/or radiation therapy. Primary cancer targets, such as those listed in Table I, are treated under standard protocols by the addition anti-58P1D12 antibodies to standard first and second line therapy. Protocol designs address effectiveness as assessed by reduction in tumor mass as well as the ability to reduce usual doses of standard chemotherapy. These dosage reductions allow additional and/or prolonged therapy by reducing dose-related toxicity of the chemotherapeutic agent. Anti-58P1D12 antibodies are utilized in several adjunctive clinical trials in combination with the chemotherapeutic or antineoplastic agents adriamycin (advanced prostrate carcinoma), cisplatin (advanced head and neck and lung carcinomas), taxol (breast cancer), and doxorubicin (preclinical).
  • II.) Monotherapy: In connection with the use of the anti-58P1D12 antibodies in monotherapy of tumors, the antibodies are administered to patients without a chemotherapeutic or antineoplastic agent. In one embodiment, monotherapy is conducted clinically in end stage cancer patients with extensive metastatic disease. Patients show some disease stabilization. Trials demonstrate an effect in refractory patients with cancerous tumors.
  • III.) Imaging Agent: Through binding a radionuclide (e.g., iodine or yttrium (I131, Y90) to anti-58P1D12 antibodies, the radiolabeled antibodies are utilized as a diagnostic and/or imaging agent. In such a role, the labeled antibodies localize to both solid tumors, as well as, metastatic lesions of cells expressing 58P1D12. In connection with the use of the anti-58P1D12 antibodies as imaging agents, the antibodies are used as an adjunct to surgical treatment of solid tumors, as both a pre-surgical screen as well as a post-operative follow-up to determine what tumor remains and/or returns. In one embodiment, a (111In)-58P1D12 antibody is used as an imaging agent in a Phase I human clinical trial in patients having a carcinoma that expresses 58P1D12 (by analogy see, e.g., Divgi et al. J. Natl. Cancer Inst. 83:97-104 (1991)). Patients are followed with standard anterior and posterior gamma camera. The results indicate that primary lesions and metastatic lesions are identified.
  • Dose and Route of Administration
  • As appreciated by those of ordinary skill in the art, dosing considerations can be determined through comparison with the analogous products that are in the clinic. Thus, anti-58P1D12 antibodies can be administered with doses in the range of 5 to 400 mg/m2, with the lower doses used, e.g., in connection with safety studies. The affinity of anti-58P1D12 antibodies relative to the affinity of a known antibody for its target is one parameter used by those of skill in the art for determining analogous dose regimens. Further, anti-58P1D12 antibodies that are fully human antibodies, as compared to the chimeric antibody, have slower clearance; accordingly, dosing in patients with such fully human anti-58P1D12 antibodies can be lower, perhaps in the range of 50 to 300 mg/m2, and still remain efficacious. Dosing in mg/m2, as opposed to the conventional measurement of dose in mg/kg, is a measurement based on surface area and is a convenient dosing measurement that is designed to include patients of all sizes from infants to adults.
  • Three distinct delivery approaches are useful for delivery of anti-58P1D12 antibodies. Conventional intravenous delivery is one standard delivery technique for many tumors. However, in connection with tumors in the peritoneal cavity, such as tumors of the ovaries, biliary duct, other ducts, and the like, intraperitoneal administration may prove favorable for obtaining high dose of antibody at the tumor and to also minimize antibody clearance. In a similar manner, certain solid tumors possess vasculature that is appropriate for regional perfusion. Regional perfusion allows for a high dose of antibody at the site of a tumor and minimizes short term clearance of the antibody.
  • Clinical Development Plan (CDP)
  • Overview: The CDP follows and develops treatments of anti-58P1D12 antibodies in connection with adjunctive therapy, monotherapy, and as an imaging agent. Trials initially demonstrate safety and thereafter confirm efficacy in repeat doses. Trails are open label comparing standard chemotherapy with standard therapy plus anti-58P1D12 antibodies. As will be appreciated, one criteria that can be utilized in connection with enrollment of patients is 58P1D12 expression levels in their tumors as determined by biopsy.
  • As with any protein or antibody infusion-based therapeutic, safety concerns are related primarily to (i) cytokine release syndrome, i.e., hypotension, fever, shaking, chills; (ii) the development of an immunogenic response to the material (i.e., development of human antibodies by the patient to the antibody therapeutic, or HAHA response); and, (iii) toxicity to normal cells that express 58P1D12. Standard tests and follow-up are utilized to monitor each of these safety concerns. Anti-58P1D12 antibodies are found to be safe upon human administration.
  • Example 41 Human Clinical Trial Adjunctive Therapy with Human Anti-58P1D12 Antibody and Chemotherapeutic Agent
  • A phase I human clinical trial is initiated to assess the safety of six intravenous doses of a human anti-58P1D12 antibody in connection with the treatment of a solid tumor, e.g., a cancer of a tissue listed in Table I. In the study, the safety of single doses of anti-58P1D12 antibodies when utilized as an adjunctive therapy to an antineoplastic or chemotherapeutic agent as defined herein, such as, without limitation: cisplatin, topotecan, doxorubicin, adriamycin, taxol, or the like, is assessed. The trial design includes delivery of six single doses of an anti-58P1D12 antibody with dosage of antibody escalating from approximately about 25 mg/m2 to about 275 mg/m2 over the course of the treatment in accordance with the following schedule:
  • Day 0 Day 7 Day 14 Day 21 Day 28 Day 35
    mAb Dose 25 mg/m2 75 mg/m2 125 mg/m2 175 mg/m2 225 mg/m2 275 mg/m2
    Chemotherapy + + + + + +
    (standard dose)
  • Patients are closely followed for one-week following each administration of antibody and chemotherapy. In particular, patients are assessed for the safety concerns mentioned above: (i) cytokine release syndrome, i.e., hypotension, fever, shaking, chills; (ii) the development of an immunogenic response to the material (i.e., development of human antibodies by the patient to the human antibody therapeutic, or HAHA response); and, (iii) toxicity to normal cells that express 58P1D12. Standard tests and follow-up are utilized to monitor each of these safety concerns. Patients are also assessed for clinical outcome, and particularly reduction in tumor mass as evidenced by MRI or other imaging.
  • The anti-58P1D12 antibodies are demonstrated to be safe and efficacious, Phase II trials confirm the efficacy and refine optimum dosing.
  • Example 42 Human Clinical Trial: Monotherapy with Human Anti-58P1D12 Antibody
  • Anti-58P1D12 antibodies are safe in connection with the above-discussed adjunctive trial, a Phase II human clinical trial confirms the efficacy and optimum dosing for monotherapy. Such trial is accomplished, and entails the same safety and outcome analyses, to the above-described adjunctive trial with the exception being that patients do not receive chemotherapy concurrently with the receipt of doses of anti-58P1D12 antibodies.
  • Example 43 Human Clinical Trial: Diagnostic Imaging with Anti-58P1D12 Antibody
  • Once again, as the adjunctive therapy discussed above is safe within the safety criteria discussed above, a human clinical trial is conducted concerning the use of anti-58P1D12 antibodies as a diagnostic imaging agent. The protocol is designed in a substantially similar manner to those described in the art, such as in Divgi et al. J. Natl. Cancer Inst. 83:97-104 (1991). The antibodies are found to be both safe and efficacious when used as a diagnostic modality.
  • Example 44 I. Enhanced Angiogenesis (Tube Formation) of Human Umbilical Vein Endothelial Cells by Recombinant Extracellular Domain (ECD) of 58P1D12
  • Angiogenesis is a critical event in the maintenance of tumors for their growth, nutrition and oxygenation. Growth and migration of endothelial cells is a hallmark of angiogenesis, and the stimulation of such cells requires multiple serum growth factors including vascular endothelial growth factor (VEGF). To address whether 58P1D12 facilitates angiogenesis of primary endothelium, an assay was established to detect the effect of the extracellular domain (ECD) (amino acids x-y) of 58P1D12 on the formation of endothelial matrices (tubes) when plated on Matrigel®. Human umbilical vein endothelial cells (HUVEC) or human lung microvascular endothelial cells (HMVEC) were grown in 10% FBS in media to 70% confluency (passage 2-passage 6). Cells were detached in 1:1 trypsin:PBS or 10 mM EDTA, washed and resuspended in EBM-0.1% FBS. Recombinant 58P1D12 ECD (described in Example entitled “Production of Recombinant 58P1D12 in Eukaryotic Systems”) was added to the cells and then incubated on 200 □l Matrigel® for 8-16 hours at 37° C. Tube formation was quantitated and captured by photography. FIG. 23 shows that the 58P1D12 ECD induced the formation of endothelial tubes similarly as generated by treatment with purified VEGF protein. FIG. 24 shows that similar results were obtained when treating mature lung-derived microvascular endothelial cells (HMVEC) with 58P1D12 ECD. Together, these results indicate that the 58P1D12 protein is involved in promoting angiogenesis that facilitates tumor growth, activation of endothelium for tumor vascularization or tumor cell invasion. FIG. 25 shows inhibition of endothelial tubes induced by 58P1D12 ECD with monoclonal antibody M8-143(5)10 at 30 □g/ml. An isotype control monoclonal antibody at the same concentration did not inhibit tube formation.
  • II. Enhanced Survival and Proliferation of Endothelial Cells by Recombinant Extracellular Domain (ECD) of 58P1D12
  • Enhanced proliferation, entry into S-phase and survival of endothelial cells is a hallmark of both angiogenesis and the cancer cell phenotype. To address the effect of the 58P1D12 protein on the survival and proliferation of normal endothelial cells, both HUVEC and HMVEC were treated with the 58P1D12 ECD and assayed in low serum conditions. The cells were grown overnight in 0.5% FBS and then compared to cells treated with VEGF or 58P1D12 ECD. The cells were evaluated for survival by the Alamar blue assay. The results in FIG. 26 indicate that HUVEC cell viability was increased by the endothelial growth factor VEGF (positive control) as well as increasing concentrations of recombinant 58P1D12 ECD. Further, the results in FIG. 27 show that proliferation of HMVEC in response to 58P1D12 ECD was enhanced at 72 hr post-treatment measured by a 3H-thymidine incorporation assay. These results confirm those measured by Alamar blue assay, and indicate that cells that are treated with 58P1D12 protein have an enhanced proliferative capacity and survive in low serum conditions. Accordingly, 58P1D12 expressing cells induce increased potential for growth of endothelium in vivo.
  • Together, these data indicate that the 58P1D12 protein has growth promoting, survival potentiating and angiogenic activities for primary endothelium. Such activities are vital for the establishment and maintenance of tumors by stimulating capillary bed formation by endothelial cells. This increased angiogenesis provides a means of increased nutrition and oxygenation of growing tumors, and affords a therapeutic intervention strategy for targeting of 58P1D12 in malignancies. The 58P1D12 protein also plays a role in cell migration, particularly for cells that express the protein, and also serves as an attractant for endothelial cells and other cells expressing its ligand. Cell migration is important for the metastatic potential of tumor cells, and facilitates their homing to remote tissues. Expression of 58P1D12 protein in bone metastases indicates that the migratory phenotype of 58P1D12-expressing cells imparts a strong potential for such cells to metastasize.
  • Example 45 58P1D12 RNA Interference (RNAi)
  • RNA interference (RNAi) technology is implemented to a variety of cell assays relevant to oncology. RNAi is a post-transcriptional gene silencing mechanism activated by double-stranded RNA (dsRNA). RNAi induces specific mRNA degradation leading to changes in protein expression and subsequently in gene function. In mammalian cells, these dsRNAs called short interfering RNA (siRNA) have the correct composition to activate the RNAi pathway targeting for degradation, specifically some mRNAs. See, Elbashir S. M., et. al., Duplexes of 21-nucleotide RNAs Mediate RNA interference in Cultured Mammalian Cells, Nature 411(6836):494-8 (2001). Thus, RNAi technology is used successfully in mammalian cells to silence targeted genes.
  • Loss of cell proliferation control is a hallmark of cancerous cells; thus, assessing the role of 58P1D12 in cell survival/proliferation assays is relevant. Accordingly, RNAi was used to investigate the function of the 58P1D12 antigen. To generate siRNA for 58P1D12, algorithms were used that predict oligonucleotides that exhibit the critical molecular parameters (G:C content, melting temperature, etc.) and have the ability to significantly reduce the expression levels of the 58P1D12 protein when introduced into cells. In accordance with this Example, 58P1D12 siRNA compositions are used that comprise siRNA (double stranded, short interfering RNA) that correspond to the nucleic acid ORF sequence of the 58P1D12 protein or subsequences thereof. Thus, siRNA subsequences are used in this manner are generally 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35 or more than 35 contiguous RNA nucleotides in length. These siRNA sequences are complementary and non-complementary to at least a portion of the mRNA coding sequence. In a preferred embodiment, the subsequences are 19-25 nucleotides in length, most preferably 21-23 nucleotides in length. In preferred embodiments, these siRNA achieve knockdown of 58P1D12 antigen in cells expressing the protein and have functional effects as described below.
  • Mammalian siRNA transfections: The day before siRNA transfection, the different cell lines are plated in media (RPMI 1640 with 10% FBS w/o antibiotics) at 2×103 cells/well in 80μ (96 well plate format) for the proliferation assay. In parallel with the 58P1D12 specific siRNA oligo, the following sequences are included in every experiment as controls: a) Mock transfected cells with Lipofectamine 2000 (Invitrogen, Carlsbad, Calif.) and annealing buffer (no siRNA); b) Luciferase-4 specific siRNA (targeted sequence: 5′-AAGGGACGAAGACGAACACUUCTT-3′) (SEQ ID NO:35); and, c) Eg5 specific siRNA (targeted sequence: 5′-AACTGAAGACCTGAAGACAATAA-3′) (SEQ ID NO:36). SiRNAs are used at 10 nM and μg/ml Lipofectamine 2000 final concentration.
  • The procedure is as follows: The siRNAs are first diluted in OPTIMEM (serum-free transfection media, Invitrogen) at 0.1 μM (10-fold concentrated) and incubated 5-10 min RT. Lipofectamine 2000 was diluted at 10 μg/ml (10-fold concentrated) for the total number transfections and incubated 5-10 minutes at room temperature (RT). Appropriate amounts of diluted 10-fold concentrated Lipofectamine 2000 were mixed 1:1 with diluted 10-fold concentrated siRNA and incubated at RT for 20-30″ (5-fold concentrated transfection solution). 20 μls of the 5-fold concentrated transfection solutions were added to the respective samples and incubated at 37° C. for 96 hours before analysis.
  • 3H-Thymidine incorporation assay: The proliferation assay is a 3H-thymidine incorporation method for determining the proliferation of viable cells by uptake and incorporation of label into DNA.
  • The procedure was as follows: Cells growing in log phase are trypsinized, washed, counted and plated in 96-well plates at 1000-4000 cells/well in 10% FBS. After 4-8 hrs, the media is replaced. The cells are incubated for 24-72 hrs, pulsed with 3H-Thy at 1.5 μCi/ml for 14 hrs, harvested onto a filtermat and counted in scintillation cocktail on a Microbeta trilux or other counter.
  • These data indicate that 58P1D12 plays an important role in the proliferation of cancer cells and that the lack of 58P1D12 clearly decreases the survival potential of these cells. It is to be noted that 58P1D12 is constitutively expressed in many tumor cell lines. 58P1D12 serves a role in malignancy; its expression is a primary indicator of disease, where such disease is often characterized by high rates of uncontrolled cell proliferation and diminished apoptosis. Correlating cellular phenotype with gene knockdown following RNAi treatments is important, and allows one to draw valid conclusions and rule out toxicity or other non-specific effects of these reagents. To this end, assays to measure the levels of expression of both protein and mRNA for the target after RNAi treatments are important, including Western blotting, FACS staining with antibody, immunoprecipitation, Northern blotting or RT-PCR (Taqman or standard methods). Any phenotypic effect of the siRNAs in these assays should be correlated with the protein and/or mRNA knockdown levels in the same cell lines. 58P1D12 protein is reduced after treatment with siRNA oligos described above (e.g., 58P1D12.a, etc.).
  • A method to analyze 58P1D12 related cell proliferation is the measurement of DNA synthesis as a marker for proliferation. Labeled DNA precursors (i.e. 3H-Thymidine) are used and their incorporation to DNA is quantified. Incorporation of the labeled precursor into DNA is directly proportional to the amount of cell division occurring in the culture. Another method used to measure cell proliferation is performing clonogenic assays. In these assays, a defined number of cells are plated onto the appropriate matrix and the number of colonies formed after a period of growth following siRNA treatment is counted.
  • In 58P1D12 cancer target validation, complementing the cell survival/proliferation analysis with apoptosis and cell cycle profiling studies are considered. The biochemical hallmark of the apoptotic process is genomic DNA fragmentation, an irreversible event that commits the cell to die. A method to observe fragmented DNA in cells is the immunological detection of histone-complexed DNA fragments by an immunoassay (i.e. cell death detection ELISA) which measures the enrichment of histone-complexed DNA fragments (mono- and oligo-nucleosomes) in the cytoplasm of apoptotic cells. This assay does not require pre-labeling of the cells and can detect DNA degradation in cells that do not proliferate in vitro (i.e. freshly isolated tumor cells).
  • The most important effector molecules for triggering apoptotic cell death are caspases. Caspases are proteases that when activated cleave numerous substrates at the carboxy-terminal site of an aspartate residue mediating very early stages of apoptosis upon activation. All caspases are synthesized as pro-enzymes and activation involves cleavage at aspartate residues. In particular, caspase 3 seems to play a central role in the initiation of cellular events of apoptosis. Assays for determination of caspase 3 activation detect early events of apoptosis. Following RNAi treatments, Western blot detection of active caspase 3 presence or proteolytic cleavage of products (i.e. PARP) found in apoptotic cells further support an active induction of apoptosis. Because the cellular mechanisms that result in apoptosis are complex, each has its advantages and limitations. Consideration of other criteria/endpoints such as cellular morphology, chromatin condensation, membrane blebbing, apoptotic bodies help to further support cell death as apoptotic. Since not all the gene targets that regulate cell growth are anti-apoptotic, the DNA content of permeabilized cells is measured to obtain the profile of DNA content or cell cycle profile. Nuclei of apoptotic cells contain less DNA due to the leaking out to the cytoplasm (sub-G1 population). In addition, the use of DNA stains (i.e., propidium iodide) also differentiate between the different phases of the cell cycle in the cell population due to the presence of different quantities of DNA in G0/G1, S and G2/M. In these studies the subpopulations can be quantified.
  • For the 58P1D12 gene, RNAi studies facilitate the understanding of the contribution of the gene product in cancer pathways. Such active RNAi molecules have use in identifying assays to screen for mAbs that are active anti-tumor therapeutics. Further, siRNA are administered as therapeutics to cancer patients for reducing the malignant growth of several cancer types, including those listed in Table I. When 58P1D12 plays a role in cell survival, cell proliferation, tumorigenesis, or apoptosis, it is used as a target for diagnostic, prognostic, preventative and/or therapeutic purposes.
  • Example 46 Homology Comparison of 58P1D12 to known Sequences
  • The 58P1D12 protein has 273 amino acids with a calculated molecular weight of 30.47 kDa and a pI of 6.38. 58P1D12 is predicted to be a plasma membrane protein (PSORT at the World Wide Web address psort.nibb.ac.jp/form.html). 58P1D12 contains a single transmembrane region from amino acids 218-240 with high probability that the amino-terminus resides outside, consistent with the topology of a Type 1 transmembrane protein (at the World Wide Web address cbs.dtu.dk/services/TMHMM). Also visualized is a short hydrophobic stretch from amino acids 1-21, consistent with the existence of an amino-terminal signal peptide (at the World Wide Web address cbs.dtu.dk/services/SignalP). Based on the TMpred algorithm of Hofmann and Stoffel which utilizes TMBASE (K. Hofmann, W. Stoffel, TMBASE—A database of membrane spanning protein segments Biol. Chem. Hoppe-Seyler 374:166, 1993), 58P1D12 contains a primary transmembrane region from amino acids 220-243 and a secondary transmembrane region from amino acids 1-21 (contiguous amino acids with values greater than 0 on the plot have high probability of being transmembrane regions) with an orientation in which the amino terminus resides inside and the carboxyl terminus outside. An alternative model is also predicted that 58P1D12 is a Type 1 transmembrane protein in which the amino-terminus resides outside and the protein contains a secondary transmembrane domain signal peptide from amino acids 1-21 and a primary transmembrane domain from aa214-aa246. The transmembrane prediction algorithms are accessed through the ExPasy molecular biology server (at the World Wide Web address expasy.ch/tools/).
  • The C-type lectins are a family of both transmembrane and secreted proteins belonging to the lectin superfamily (8 different types of lectins) of carbohydrate recognition/adhesion molecules. They have related carbohydrate recognition domains (CRDs) in their extracellular domains, but for the transmembrane anchored proteins, they contain unique cytoplasmic domains. There are 14 identified families of C-type lectins, and they are all believed to have carbohydrate recognition properties, and have roles in various cell functions including cell adhesion, glycoprotein clearance and innate immunity. The adhesion function of C-type lectins is well-established for the selectin family. Additional functions of C-type lectins include signal transduction, and may provide a platform for protein associations. Some C-type lectins have been shown to facilitate glycoprotein clearance, elimination of foreign pathogens during innate immune responses and migration of cells. In mammals, C-type lectins are widely expressed, yet play roles in other species.
  • 58P1D12 belongs to the type VIII family of C-type lectins, which includes one other member, layilin. Layilin has ˜45% homology with 58P1D12, and interacts with the carbohydrate ligand hyaluronate. Hyaluronate, an extracellular matrix component consisting of repeating disaccharide units, is a ligand for several C-type lectins and is a putative ligand for 58P1D12, but was not detected in a binding reaction with 58P1D12 using the cetylpyridinium precipitation technique (Weng, L. et al, 2002, Genomics 80:62-70). Layilin localizes in membrane ruffles during cell migration, and interacts with the cytoskeletal protein talin (Borowsky, M.L. and Hynes, R. O., 1998, J. Cell Biol. 143:429-442). 58P1D12 also is observed in membrane ruffles and associates with talin through its cytoplasmic domain.
  • 58P1D12 also interacts with other cytoplasmic signaling molecules, including serine/threonine and dual kinases that phosphorylate its intracellular serine and threonine residues. Such phosphorylation leads to the interaction of 58P1D12 with signaling molecules that are regulated by their interaction with the plasma membrane through association with 58P1D12. Such molecules induce signals that are critical to the function of 58P1D12 protein, which mediate the growth pathways that are stimulated in tumor cells expressing 58P1D12. Further, the adhesion properties of the 58P1D12 protein expressed on tumor cells enhances their ability to migrate during metastases, facilitates their homing to distal sites (lymph nodes), and promotes interactions with, and activation of, other cells such as endothelia during the formation of tumor masses. The adhesive and growth signals of 58P1D12 protein significantly promote unregulated growth of cancer cells, contributing to their growth advantage in vivo.
  • Similar to other C-type lectins, 58P1D12 is membrane associated. Functional analysis shows that a recombinant soluble form of 58P1D12 has both angiogenic and proliferative enhancing properties for endothelial cells (see Example 44). The membrane-associated form of the 58P1D12 protein or a secreted portion promotes establishment and maintenance of the malignant state of tumors expressing the 58P1D12 antigen. The formation of capillary beds surrounding tumors through the angiogenic function of 58P1D12 protein as well as the proliferative signals that are imparted by the protein promote survival of tumors which require nutrition and oxygenation.
  • Throughout this application, various website data content, publications, patent applications and patents are referenced. (Websites are referenced by their Uniform Resource Locator, or URL, addresses on the World Wide Web.) The disclosures of each of these references are hereby incorporated by reference herein in their entireties.
  • The present invention is not to be limited in scope by the embodiments disclosed herein, which are intended as single illustrations of individual aspects of the invention, and any that are functionally equivalent are within the scope of the invention. Various modifications to the models and methods of the invention, in addition to those described herein, will become apparent to those skilled in the art from the foregoing description and teachings, and are similarly intended to fall within the scope of the invention. Such modifications or other embodiments can be practiced without departing from the true scope and spirit of the invention.
  • TABLE 1
    Tissues that express 58P1D12 when malignant.
    Prostate
    Bladder
    Lung
    Ovary
    Cervix
  • TABLE II
    Amino Acid Abbreviations
    SINGLE LETTER THREE LETTER FULL NAME
    F Phe phenylalanine
    L Leu leucine
    S Ser serine
    Y Tyr tyrosine
    C Cys cysteine
    W Trp tryptophan
    P Pro proline
    H His histidine
    Q Gln glutamine
    R Arg arginine
    I Ile isoleucine
    M Met methionine
    T Thr threonine
    N Asn asparagine
    K Lys lysine
    V Val valine
    A Ala alanine
    D Asp aspartic acid
    E Glu glutamic acid
    G Gly glycine
  • TABLE III
    Amino Acid Substitution Matrix
    Adapted from the GCG Software 9.0 BLOSUM62 amino acid substitution matrix
    (block substitution matrix). The higher the value, the more likely a substitution is found
    in related, natural proteins. (See world wide web URL ikp.unibe.ch/manual/blosum62.html)
    A C D E F G H I K L M N P Q R S T V W Y .
    4 0 −2 −1 −2 0 −2 −1 −1 −1 −1 −2 −1 −1 −1 1 0 0 −3 −2 A
    9 −3 −4 −2 −3 −3 −1 −3 −1 −1 −3 −3 −3 −3 −1 −1 −1 −2 −2 C
    6 2 −3 −1 −1 −3 −1 −4 −3 1 −1 0 −2 0 −1 −3 −4 −3 D
    5 −3 −2 0 −3 1 −3 −2 0 −1 2 0 0 −1 −2 −3 −2 E
    6 −3 −1 0 −3 0 0 −3 −4 −3 −3 −2 −2 −1 1 3 F
    6 −2 −4 −2 −4 −3 0 −2 −2 −2 0 −2 −3 −2 −3 G
    8 −3 −1 −3 −2 1 −2 0 0 −1 −2 −3 −2 2 H
    4 −3 2 1 −3 −3 −3 −3 −2 −1 3 −3 −1 I
    5 −2 −1 0 −1 1 2 0 −1 −2 −3 −2 K
    4 2 −3 −3 −2 −2 −2 −1 1 −2 −1 L
    5 −2 −2 0 −1 −1 −1 1 −1 −1 M
    6 −2 0 0 1 0 −3 −4 −2 N
    7 −1 −2 −1 −1 −2 −4 −3 P
    5 1 0 −1 −2 −2 −1 Q
    5 −1 −1 −3 −3 −2 R
    4 1 −2 −3 −2 S
    5 0 −2 −2 T
    4 −3 −1 V
    11 2 W
    7 Y
  • TABLE IV
    HLA Class I/II Motifs/Supermotifs
    TABLE IV (A): HLA Class I Supermotifs/Motifs
    POSITION POSITION POSITION
    2 (Primary Anchor) 3 (Primary Anchor) C Terminus (Primary Anchor)
    SUPERMOTIF
    A1 TI LVMS FWY
    A2 LIVM ATQ IV MATL
    A3 VSMA TLI RK
    A24 YF WIVLMT FI YWLM
    B7 P VILF MWYA
    B27 RHK FYL WMIVA
    B44 E D FWY LIMVA
    B58 ATS FWYLIVMA
    B62 QL IVMP FWY MIVLA
    MOTIFS
    A1 TSM Y
    A1 DE AS Y
    A2.1 LM VQIAT V LIMAT
    A3 LMVISATF CGD KYR HFA
    A11 VTMLISAGN CDF K RYH
    A24 YFWM FLIW
    A*3101 MVT ALIS R K
    A*3301 MVALF IST RK
    A*6801 AVT MSLI RK
    B*0702 P LMF WYAIV
    B*3501 P LMFWY IVA
    B51 P LIVF WYAM
    B*5301 P IMFWY ALV
    B*5401 P ATIV LMFWY
    Bolded residues are preferred, italicized residues are less preferred: A peptide is considered
    motif-bearing if it has primary anchors at each primary anchor position for a motif or
    supermotif as specified in the above table.
    TABLE IV (B): HLA Class II Supermotif
    1 6 9
    W, F, Y, V, I, L A, V, I, L, P, C, S, T A, V, I, L, C, S, T, M, Y
    TABLE IV (C): HLA Class II Motifs
    MOTIFS 1° anchor 1 2 3 4 5 1° anchor 6 7 8 9
    DR4 preferred FMYLIVW M T I VSTCPALIM MH MH
    deleterious W R WDE
    DR1 preferred MFLIVWY PAMQ VMATSPLIC M AVM
    deleterious C CH FD CWD GDE D
    DR7 preferred MFLIVWY M W A IVMSACTPL M IV
    deleterious C G GRD N G
    DR3 MOTIFS 1° anchor 1 2 3 1° anchor 4 5 1° anchor 6
    Motif a preferred LIVMFY D
    Motif b preferred LIVMFAY DNQEST KRH
    DR Supermotif MFLIVWY VMSTACPLI
    Italicized residues indicate less preferred or “tolerated” residues
    TABLE IV (D): HLA Class I Supermotifs
    SUPER- POSITION:
    MOTIFS 1 2 3 4 5 6 7 8 C-terminus
    A1 1° Anchor 1° Anchor
    TILVMS FWY
    A2 1° Anchor 1° Anchor
    LIVMATQ LIVMAT
    A3 Preferred 1° Anchor YFW YFW YFW P 1° Anchor
    VSMATLI (4/5) (3/5) (4/5) (4/5) RK
    deleterious DE (3/5); DE
    P (5/5) (4/5)
    A24 1° Anchor 1° Anchor
    YFWIVLMT FIYWLM
    B7 Preferred FWY (5/5) 1° Anchor FWY FWY 1° Anchor
    LIVM (3/5) P (4/5) (3/5) VILFMWYA
    deleterious DE (3/5); DE G QN DE
    P(5/5); (3/5) (4/5) (4/5) (4/5)
    G(4/5);
    A(3/5);
    QN(3/5)
    B27 1° Anchor 1° Anchor
    RHK FYLWMIVA
    B44 1° Anchor 1° Anchor
    ED FWYLIMVA
    B58 1° Anchor 1° Anchor
    ATS FWYLIVMA
    B62 1° Anchor 1° Anchor
    QLIVMP FWYMIVLA
    Italicized residues indicate less preferred or “tolerated” residues
    TABLE IV (E): HLA Class I Motifs
    POSITION
    9
    or C- C-
    1 2 3 4 5 6 7 8 terminus terminus
    A1 preferred GFYW 1° Anchor DEA YFW P DEQN YFW 1° Anchor
    9-mer STM Y
    deleterious DE RHKLIVMP A G A
    A1 preferred GRHK ASTCLIVM 1° Anchor GSTC ASTC LIVM DE 1° Anchor
    9-mer DEAS Y
    deleterious A RHKDEPYFW DE PQN RHK PG GP
    A1 preferred YFW 1° Anchor DEAQN A YFWQN PASTC GDE P 1° Anchor
    10- STM Y
    mer
    deleterious GP RHKGLIVM DE RHK QNA RHKYFW RHK A
    A1 preferred YFW STCLIVM 1° Anchor A YFW PG G YFW 1° Anchor
    10- DEAS Y
    mer deleterious RHK RHKDEPYFW P G PRHK QN
    A2.1 preferred YFW 1° Anchor YFW STC YFW A P 1° Anchor
    9-mer LMIVQAT VLIMAT
    deleterious DEP DERKH RKH DERKH
    A2.1 preferred AYFW 1° Anchor LVIM G G FYWL 1° Anchor
    10- LMIVQAT VIM VLIMAT
    mer deleterious DEP DE RKHA P RKH DERK
    HRKH
    A3 preferred RHK 1° Anchor YFW PRHKYFW A YFW P 1° Anchor
    LMVISATFCGD KYRHFA
    deleterious DEP DE
    A11 preferred A 1° Anchor YFW YFW A YFW YFW P 1° Anchor
    VTLMISAGNCDF KRYH
    deleterious DEP A G
    A24 preferred YFWRHK 1° Anchor STC YFW YFW 1° Anchor
    9-mer YFWM FLIW
    deleterious DEG DE G QNP DERHKG AQN
    A24 Preferred 1° Anchor P YFWP P 1° Anchor
    10- YFWM FLIW
    mer Deleterious GDE QN RHK DE A QN DEA
    A3101 Preferred RHK 1° Anchor YFW P YFW YFW AP 1° Anchor
    MVTALIS RK
    Deleterious DEP DE ADE DE DE DE
    A3301 Preferred 1° Anchor YFW AYFW 1° Anchor
    MVALFIST RK
    Deleterious GP DE
    A6801 Preferred YFWSTC 1° Anchor YFWLIVM YFW P 1° Anchor
    AVTMSLI RK
    deleterious GP DEG RHK A
    B0702 Preferred RHKFWY 1° Anchor RHK RHK RHK RHK PA 1° Anchor
    P LMFWYAIV
    deleterious DEQNP DEP DE DE GDE QN DE
    A1 preferred GFYW 1° Anchor DEA YFW P DEQN YFW 1° Anchor
    9-mer STM Y
    deleterious DE RHKLIVMP A G A
    A1 preferred GRHK ASTCLIVM 1° Anchor GSTC ASTC LIVM DE 1° Anchor
    9-mer DEAS Y
    deleterious A RHKDEPYFW DE PQN RHK PG GP
    B3501 Preferred FWYLIVM 1° Anchor FWY FWY 1° Anchor
    P LMFWYIVA
    deleterious AGP G G
    B51 Preferred LIVMFWY 1° Anchor FWY STC FWY G FWY 1° Anchor
    P LIVFWYAM
    deleterious AGPDER DE G DEQN GDE
    HKSTC
    B5301 preferred LIVMFWY 1° Anchor FWY STC FWY LIVMF FWY 1° Anchor
    P WY IMFWYALV
    deleterious AGPQN G RHKQN DE
    B5401 preferred FWY 1° Anchor FWYLIVM LIVM ALIVM FWYA 1° Anchor
    P P ATIVLMFWY
    deleterious GPQNDE GDESTC RHKDE DE QNDGE DE
    TABLE IV (F):
    Summary of HLA-supertypes
    Overall phenotypic frequencies of HLA-supertypes in different ethnic populations
    Specificity Phenotypic frequency
    Supertype Position 2 C-Terminus Caucasian N.A. Black Japanese Chinese Hispanic Average
    B7 P AILMVFWY 43.2 55.1 57.1 43.0 49.3 49.5
    A3 AILMVST RK 37.5 42.1 45.8 52.7 43.1 44.2
    A2 AILMVT AILMVT 45.8 39.0 42.4 45.9 43.0 42.2
    A24 YF (WIVLMT) FI (YWLM) 23.9 38.9 58.6 40.1 38.3 40.0
    B44 E (D) FWYLIMVA 43.0 21.2 42.9 39.1 39.0 37.0
    A1 TI (LVMS) FWY 47.1 16.1 21.8 14.7 26.3 25.2
    B27 RHK FYL (WMI) 28.4 26.1 13.3 13.9 35.3 23.4
    B62 QL (IVMP) FWY (MIV) 12.6 4.8 36.5 25.4 11.1 18.1
    B58 ATS FWY (LIV) 10.0 25.1 1.6 9.0 5.9 10.3
    TABLE IV (G):
    Calculated population coverage afforded by different HLA-supertype combinations
    Phenotypic frequency
    HLA-supertypes Caucasian N.A Blacks Japanese Chinese Hispanic Average
    A2, A3 and B7 83.0 86.1 87.5 88.4 86.3 86.2
    A2, A3, B7, A24, 99.5 98.1 100.0 99.5 99.4 99.3
    B44 and A1 99.9 99.6 100.0 99.8 99.9 99.8
    A2, A3, B7, A24,
    B44, A1, B27, B62,
    and B 58
    Motifs indicate the residues defining supertype specificites. The motifs incorporate residues
    determined on the basis of published data to be recognized by multiple alleles within the
    supertype. Residues within brackets are additional residues also predicted to be tolerated
    by multiple alleles within the supertype.
  • TABLE V
    Frequently Occurring Motifs
    avrg. %
    Name identity Description Potential Function
    zf-C2H2 34% Zinc finger, C2H2 type Nucleic acid-binding protein functions as
    transcription factor, nuclear location
    probable
    cytochrome_b_N 68% Cytochrome b(N- membrane bound oxidase, generate
    terminal)/b6/petB superoxide
    Ig
    19% Immunoglobulin domain domains are one hundred amino acids
    long and include a conserved
    intradomain disulfide bond.
    WD40 18% WD domain, G-beta repeat tandem repeats of about 40 residues,
    each containing a Trp-Asp motif.
    Function in signal transduction and
    protein interaction
    PDZ
    23% PDZ domain may function in targeting signaling
    molecules to sub-membranous sites
    LRR
    28% Leucine Rich Repeat short sequence motifs involved in
    protein-protein interactions
    Pkinase 23% Protein kinase domain conserved catalytic core common to
    both serine/threonine and tyrosine
    protein kinases containing an ATP
    binding site and a catalytic site
    PH
    16% PH domain pleckstrin homology involved in
    intracellular signaling or as constituents
    of the cytoskeleton
    EGF
    34% EGF-like domain 30-40 amino-acid long found in the
    extracellular domain of membrane-
    bound proteins or in secreted proteins
    Rvt 49% Reverse transcriptase
    (RNA-dependent DNA
    polymerase)
    Ank 25% Ank repeat Cytoplasmic protein, associates integral
    membrane proteins to the cytoskeleton
    Oxidored_q1
    32% NADH- membrane associated. Involved in
    Ubiquinone/plastoquinone proton translocation across the
    (complex I), various chains membrane
    Efhand
    24% EF hand calcium-binding domain, consists of a12
    residue loop flanked on both sides by a
    12 residue alpha-helical domain
    Rvp 79% Retroviral aspartyl Aspartyl or acid proteases, centered on
    protease a catalytic aspartyl residue
    Collagen
    42% Collagen triple helix repeat extracellular structural proteins involved
    (20 copies) in formation of connective tissue. The
    sequence consists of the G-X-Y and the
    polypeptide chains forms a triple helix.
    Fn3 20% Fibronectin type III domain Located in the extracellular ligand-
    binding region of receptors and is about
    200 amino acid residues long with two
    pairs of cysteines involved in disulfide
    bonds
    7tm_1 19% 7 transmembrane receptor seven hydrophobic transmembrane
    (rhodopsin family) regions, with the N-terminus located
    extracellularly while the C-terminus is
    cytoplasmic. Signal through G proteins
  • TABLE VI
    Motif-bearing subsequences of 58P1D12
    N-glycosylation sites
    86 NLTK (SEQ ID NO: 37)
    185 NPTA (SEQ ID NO: 38)
    O-glycosylation sites
    S 2
    S 6
    S 26
    S 49
    S 50
    S 53
    S 63
    S 70
    S 82
    T 88
    T 92
    S 95
    T 111
    S 112
    S 122
    S 125
    S 127
    T 134
    S 138
    S 141
    T 152
    T 187
    T 196
    T 202
    T 209
    T 224
    T 237
    S 246
    T 250
    T 252
    S 253
    S 257
    T 258
    S 262
    S 264
    T 265
    S 269
    Phosphorylation sites
    Serine residues
    Pos. Context
     53 SSRVSFQEA (SEQ ID NO: 39)
     70 GVLLSLENE (SEQ ID NO: 40)
     95 GTGISDGDF (SEQ ID NO: 41)
    112 DGQTSGACP (SEQ ID NO: 42)
    127 DGSNSQYRN (SEQ ID NO: 43)
    141 PSCGSEKCV (SEQ ID NO: 44)
    246 MLHKSKGRT (SEQ ID NO: 45)
    253 RTKTSPNQS (SEQ ID NO: 46)
    269 TRKESGMEV (SEQ ID NO: 47)
    Threonine residue
    Pos Context
    265 ISKSTRKES (SEQ ID NO: 48)
    Pos Context
    Tyrosine residues
    119 CPDLYQWSD (SEQ ID NO: 49)
    129 SNSQYRNWY (SEQ ID NO: 50)
    133 YRNWYTDEP (SEQ ID NO: 51)
    176 MKHNYICKY (SEQ ID NO: 52)
    194 VEKPYLTNQ (SEQ ID NO: 53)
    219 PNLIYVVIP (SEQ ID NO: 54)
    SUMO-1 binding site
    K36 VCFAD F K HP CYKMA (SEQ ID NO: 55)
  • TABLE VII
    Search Peptides
    58P1D12 Variant
     1, aa 1-273; (SEQ ID NO: 56)
    9-mer;
    10-mer;
    15-mer.
    MSRVVSLLLG AALLCGHGAF CRRVVSGQKV CFADFKHPCY KMAYFHELSS RVSFQEARLA
    CESEGGVLLS LENEAEQKLI ESMLQNLTKP GTGISDGDFW IGLWRNGDGQ TSGACPDLYQ
    WSDGSNSQYR NWYTDEPSCG SEKCVVMYHQ PTANPGLGGP YLYQWNDDRC NMKHNYICKY
    EPEINPTAPV EKPYLTNQPG DTHQNVVVTE AGIIPNLIYV VIPTIPLLLL ILVAFGTCCF
    QMLHKSKGRT KTSPNQSTLW ISKSTRKESG MEV
    58P1D12 Variant
     4
    9-mers, aa 163-236 (SEQ ID NO: 57)
    QNVVVTEAVKEEQKLVQTSLHCGFQRVPEKKVAWKYNNSLTWFQNFVILDLYKEWHQN
    NSLEWLEITKDLQDEL
    10-mers, aa 162-236 (SEQ ID NO: 58)
    HQNVVVTEAVKEEQKLVQTSLHCGFQRVPEKKVAWKYNNSLTWFQNFVILDLYKEWHQN
    NSLEWLEITKDLQDEL
    15-mers, aa 157-236 (SEQ ID NO: 59)
    QPGDTHQNVVVTEAVKEEQKLVQTSLHCGFQRVPEKKVAWKYNNSLTWFQNFVILDLYKEWHQN
    NSLEWLEITKDLQDEL
  • TABLE VIII
    58P1D12v.1-A1-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    32 FADFKHPCY 50.000
    121 WSDGSNSQY 37.500
    70 SLENEAEQK 18.000
    140 GSEKCVVMY 13.500
    208 VTEAGIIPN 11.250
    62 ESEGGVLLS 6.750
    74 EAEQKLIES 4.500
    165 WNDDRCNMK 2.500
    81 ESMLQNLTK 1.500
    186 PTAPVEKPY 1.250
    133 YTDEPSCGS 1.250
    237 TCCFQMLHK 1.000
    60 ACESEGGVL 0.900
    79 LIESMLQNL 0.900
    94 ISDGDFWIG 0.750
    111 TSGACPDLY 0.750
    211 AGIIPNLIY 0.625
    212 GIIPNLIYV 0.500
    14 LCGHGAFCR 0.500
    179 KYEPEINPT 0.450
    251 KTSPNQSTL 0.250
    91 GTGISDGDF 0.250
    72 ENEAEQKLI 0.225
    220 VVIPTIPLL 0.200
    227 LLLLILVAF 0.200
    113 GACPDLYQW 0.200
    24 VVSGQKVCF 0.200
    255 NQSTLWISK 0.150
    106 NGDGQTSGA 0.125
    236 GTCCFQMLH 0.125
    153 ANPGLGGPY 0.125
    96 DGDFWIGLW 0.125
    223 PTIPLLLLI 0.125
    125 SNSQYRNWY 0.125
    36 KHPCYKMAY 0.125
    184 INPTAPVEK 0.100
    161 YLYQWNDDR 0.100
    12 ALLCGHGAF 0.100
    232 LVAFGTCCF 0.100
    8 LLGAALLCG 0.050
    224 TIPLLLLIL 0.050
    156 GLGGPYLYQ 0.050
    3 RVVSLLLGA 0.050
    7 LLLGAALLC 0.050
    29 KVCFADFKH 0.050
    201 DTHQNVVVT 0.050
    93 GISDGDFWI 0.050
    78 KLIESMLQN 0.050
    157 LGGPYLYQW 0.050
    37 HPCYKMAYF 0.050
    216 NLIYVVIPT 0.050
    219 YVVIPTIPL 0.050
    110 QTSGACPDL 0.050
    42 MAYFHELSS 0.050
    171 NMKHNYICK 0.050
    221 VIPTIPLLL 0.050
    229 LLILVAFGT 0.050
    241 QMLHKSKGR 0.050
    134 TDEPSCGSE 0.045
    45 FHELSSRVS 0.045
    189 PVEKPYLTN 0.045
    5 VSLLLGAAL 0.030
    69 LSLENEAEQ 0.030
    54 FQEARLACE 0.027
    168 DRCNMKHNY 0.025
    257 STLWISKST 0.025
    97 GDFWIGLWR 0.025
    196 TNQPGDTHQ 0.025
    195 LTNQPGDTH 0.025
    15 CGHGAFCRR 0.025
    87 LTKPGTGIS 0.025
    172 MKHNYICKY 0.025
    155 PGLGGPYLY 0.025
    181 EPEINPTAP 0.022
    18 GAFCRRVVS 0.020
    213 IIPNLIYVV 0.020
    187 TAPVEKPYL 0.020
    145 VVMYHQPTA 0.020
    6 SLLLGAALL 0.020
    228 LLLILVAFG 0.020
    13 LLCGHGAFC 0.020
    51 RVSFQEARL 0.020
    183 EINPTAPVE 0.020
    205 NVVVTEAGI 0.020
    217 LIYVVIPTI 0.020
    124 GSNSQYRNW 0.015
    126 NSQYRNWYT 0.015
    25 VSGQKVCFA 0.015
    52 VSFQEARLA 0.015
    256 QSTLWISKS 0.015
    252 TSPNQSTLW 0.015
    115 CPDLYQWSD 0.013
    199 PGDTHQNVV 0.013
    225 IPLLLLILV 0.013
    151 PTANPGLGG 0.013
    112 SGACPDLYQ 0.013
    222 IPTIPLLLL 0.013
    253 SPNQSTLWI 0.013
    166 NDDRCNMKH 0.013
    65 GGVLLSLEN 0.013
  • TABLE IX
    58P1D12v.1-A1-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    32 FADFkHPCYK 10.000
    134 TDEPsCGSEK 9.000
    121 WSDGsNSQYR 7.500
    62 ESEGgVLLSL 6.750
    96 DGDFwIGLWR 6.250
    152 TANPgLGGPY 5.000
    208 VTEAgIIPNL 4.500
    183 EINPtAPVEK 4.000
    94 ISDGdFWIGL 3.750
    210 EAGIiPNLIY 2.500
    236 GTCCfQMLHK 2.500
    60 ACESeGGVLL 1.800
    110 QTSGaCPDLY 1.250
    35 FKHPcYKMAY 1.250
    70 SLENeAEQKL 0.900
    45 FHELsSRVSF 0.900
    79 LIESmLQNLT 0.900
    74 EAEQkLIESM 0.900
    179 KYEPeINPTA 0.900
    124 GSNSqYRNWY 0.750
    140 GSEKcVVMYH 0.675
    165 WNDDrCNMKH 0.625
    13 LLCGhGAFCR 0.500
    220 VVIPtIPLLL 0.500
    181 EPEInPTAPV 0.450
    69 LSLEnEAEQK 0.300
    257 STLWiSKSTR 0.250
    251 KTSPnQSTLW 0.250
    139 CGSEkCVVMY 0.250
    154 NPGLgGPYLY 0.250
    212 GIIPnLIYVV 0.200
    238 CCFQmLHKSK 0.200
    23 RVVSgQKVCF 0.200
    54 FQEArLACES 0.135
    106 NGDGqTSGAC 0.125
    185 NPTApVEKPY 0.125
    223 PTIPlLLLIL 0.125
    115 CPDLyQWSDG 0.125
    133 YTDEpSCGSE 0.125
    72 ENEAeQKLIE 0.113
    11 AALLcGHGAF 0.100
    113 GACPdLYQWS 0.100
    14 LCGHgAFCRR 0.100
    42 MAYFhELSSR 0.100
    231 ILVAfGTCCF 0.100
    111 TSGAcPDLYQ 0.075
    52 VSFQeARLAC 0.075
    252 TSPNqSTLWI 0.075
    233 VAFGtCCFQM 0.050
    221 VIPTiPLLLL 0.050
    80 IESMlQNLTK 0.050
    207 VVTEaGIIPN 0.050
    7 LLLGaALLCG 0.050
    224 TIPLlLLILV 0.050
    6 SLLLgAALLC 0.050
    31 CFADfKHPCY 0.050
    186 PTAPvEKPYL 0.050
    228 LLLIlVAFGT 0.050
    5 VSLLlGAALL 0.030
    196 TNQPgDTHQN 0.025
    157 LGGPyLYQWN 0.025
    170 CNMKhNYICK 0.025
    254 PNQStLWISK 0.025
    199 PGDThQNVVV 0.025
    41 KMAYfHELSS 0.025
    91 GTGIsDGDFW 0.025
    195 LTNQpGDTHQ 0.025
    120 QWSDgSNSQY 0.025
    171 NMKHnYICKY 0.025
    144 CVVMyHQPTA 0.020
    216 NLIYvVIPTI 0.020
    83 MLQNlTKPGT 0.020
    169 RCNMkHNYIC 0.020
    18 GAFCrRVVSG 0.020
    258 TLWIsKSTRK 0.020
    213 IIPNlIYVVI 0.020
    242 MLHKsKGRTK 0.020
    219 YVVIpTIPLL 0.020
    68 LLSLeNEAEQ 0.020
    4 VVSLlLGAAL 0.020
    12 ALLCgHGAFC 0.020
    138 SCGSeKCVVM 0.020
    226 PLLLlILVAF 0.020
    227 LLLLiLVAFG 0.020
    187 TAPVeKPYLT 0.020
    156 GLGGpYLYQW 0.020
    49 SSRVsFQEAR 0.015
    256 QSTLwISKST 0.015
    240 FQMLhKSKGR 0.015
    225 IPLLlLILVA 0.013
    87 LTKPgTGISD 0.013
    150 QPTAnPGLGG 0.013
    188 APVEkPYLTN 0.013
    235 FGTCcFQMLH 0.013
    222 IPTIpLLLLI 0.013
    64 EGGVlLSLEN 0.013
    92 TGISdGDFWI 0.013
    211 AGIIpNLIYV 0.013
    145 VVMYhQPTAN 0.010
    93 GISDgDFWIG 0.010
  • TABLE X
    58P1D12v.1-A0201-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    93 GISDGDFWI 187.620
    212 GIIPNLIYV 120.977
    6 SLLLGAALL 79.041
    229 LLILVAFGT 73.477
    13 LLCGHGAFC 46.451
    231 ILVAFGTCC 32.093
    7 LLLGAALLC 31.249
    67 VLLSLENEA 31.249
    217 LIYVVIPTI 17.949
    216 NLIYVVIPT 17.140
    213 IIPNLIYVV 15.331
    86 NLTKPGTGI 10.433
    219 YVVIPTIPL 8.598
    220 VVIPTIPLL 7.309
    176 YICKYEPEI 6.599
    225 IPLLLLILV 6.568
    228 LLLILVAFG 5.929
    221 VIPTIPLLL 4.993
    194 YLTNQPGDT 4.456
    71 LENEAEQKL 2.895
    224 TIPLLLLIL 2.770
    145 VVMYHQPTA 2.734
    78 KLIESMLQN 2.460
    126 NSQYRNWYT 2.007
    51 RVSFQEARL 1.869
    198 QPGDTHQNV 1.861
    180 YEPEINPTA 1.822
    61 CESEGGVLL 1.703
    242 MLHKSKGRT 1.647
    187 TAPVEKPYL 1.632
    84 LQNLTKPGT 1.284
    235 FGTCCFQML 1.219
    227 LLLLILVAF 1.078
    230 LILVAFGTC 1.061
    251 KTSPNQSTL 1.038
    3 RVVSLLLGA 1.000
    40 YKMAYFHEL 0.959
    209 TEAGIIPNL 0.955
    63 SEGGVLLSL 0.955
    205 NVVVTEAGI 0.913
    44 YFHELSSRV 0.911
    5 VSLLLGAAL 0.877
    59 LACESEGGV 0.875
    95 SDGDFWIGL 0.786
    154 NPGLGGPYL 0.767
    79 LIESMLQNL 0.735
    25 VSGQKVCFA 0.697
    144 CVVMYHQPT 0.652
    222 IPTIPLLLL 0.545
    257 STLWISKST 0.383
    226 PLLLLILVA 0.381
    169 RCNMKHNYI 0.375
    253 SPNQSTLWI 0.375
    41 KMAYFHELS 0.351
    110 QTSGACPDL 0.297
    146 VMYHQPTAN 0.293
    8 LLGAALLCG 0.291
    119 YQWSDGSNS 0.288
    11 AALLCGHGA 0.255
    80 IESMLQNLT 0.246
    4 VVSLLLGAA 0.221
    188 APVEKPYLT 0.199
    138 SCGSEKCVV 0.186
    23 RVVSGQKVC 0.178
    29 KVCFADFKH 0.132
    240 FQMLHKSKG 0.129
    83 MLQNLTKPG 0.127
    258 TLWISKSTR 0.124
    156 GLGGPYLYQ 0.123
    263 KSTRKESGM 0.114
    75 AEQKLIESM 0.109
    161 YLYQWNDDR 0.107
    82 SMLQNLTKP 0.092
    206 VVVTEAGII 0.083
    149 HQPTANPGL 0.074
    214 IPNLIYVVI 0.071
    102 GLWRNGDGQ 0.061
    163 YQWNDDRCN 0.058
    139 CGSEKCVVM 0.055
    31 CFADFKHPC 0.054
    170 CNMKHNYIC 0.049
    35 FKHPCYKMA 0.048
    52 VSFQEARLA 0.041
    241 QMLHKSKGR 0.040
    200 GDTHQNVVV 0.040
    203 HQNVVVTEA 0.039
    16 GHGAFCRRV 0.037
    58 RLACESEGG 0.037
    12 ALLCGHGAF 0.036
    182 PEINPTAPV 0.030
    234 AFGTCCFQM 0.020
    1 MSRVVSLLL 0.018
    114 ACPDLYQWS 0.016
    53 SFQEARLAC 0.014
    47 ELSSRVSFQ 0.014
    106 NGDGQTSGA 0.013
    68 LLSLENEAE 0.012
    232 LVAFGTCCF 0.011
    24 VVSGQKVCF 0.011
    210 EAGIIPNLI 0.011
  • TABLE XI
    58P1D12v.1-A0201-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    78 KLIEsMLQNL 705.066
    228 LLLIlVAFGT 271.864
    12 ALLCgHGAFC 101.099
    161 YLYQwNDDRC 35.833
    216 NLIYvVIPTI 23.995
    224 TIPLlLLILV 21.996
    219 YVVIpTIPLL 19.776
    163 YQWNdDRCNM 19.231
    6 SLLLgAALLC 18.382
    230 LILVaFGTCC 16.327
    83 MLQNlTKPGT 12.668
    221 VIPTiPLLLL 11.485
    24 VVSGqKVCFA 9.348
    58 RLACeSEGGV 9.042
    212 GIIPnLIYVV 9.018
    102 GLWRnGDGQT 8.041
    197 NQPGdTHQNV 7.052
    227 LLLLiLVAFG 5.929
    211 AGIIpNLIYV 5.743
    4 VVSLlLGAAL 3.178
    220 VVIPtIPLLL 3.178
    241 QMLHkSKGRT 2.589
    233 VAFGtCCFQM 2.517
    92 TGISdGDFWI 2.172
    109 GQTSgACPDL 2.166
    229 LLILvAFGTC 2.086
    125 SNSQyRNWYT 2.007
    66 GVLLsLENEA 1.608
    213 IIPNlIYVVI 1.500
    71 LENEaEQKLI 1.414
    70 SLENeAEQKL 1.367
    7 LLLGaALLCG 1.078
    5 VSLLlGAALL 0.877
    30 VCFAdFKHPC 0.775
    153 ANPGlGGPYL 0.767
    198 QPGDtHQNVV 0.763
    94 ISDGdFWIGL 0.613
    144 CVVMyHQPTA 0.435
    75 AEQKlIESML 0.415
    252 TSPNqSTLWI 0.375
    15 CGHGaFCRRV 0.345
    178 CKYEpEINPT 0.312
    8 LLGAaLLCGH 0.291
    130 RNWYtDEPSC 0.269
    222 IPTIpLLLLI 0.266
    85 QNLTkPGTGI 0.252
    93 GISDgDFWIG 0.245
    52 VSFQeARLAC 0.204
    209 TEAGiIPNLI 0.203
    82 SMLQnLTKPG 0.199
    225 IPLLlLILVA 0.192
    41 KMAYfHELSS 0.188
    156 GLGGpYLYQW 0.171
    143 KCVVmYHQPT 0.170
    194 YLTNqPGDTH 0.168
    13 LLCGhGAFCR 0.147
    105 RNGDgQTSGA 0.133
    3 RVVSlLLGAA 0.130
    231 ILVAfGTCCF 0.127
    258 TLWIsKSTRK 0.124
    79 LIESmLQNLT 0.111
    59 LACEsEGGVL 0.110
    48 LSSRvSFQEA 0.105
    207 VVTEaGIIPN 0.105
    187 TAPVeKPYLT 0.104
    51 RVSFqEARLA 0.087
    208 VTEAgIIPNL 0.074
    264 STRKeSGMEV 0.073
    10 GAALlCGHGA 0.069
    148 YHQPtANPGL 0.068
    204 QNVVvTEAGI 0.068
    146 VMYHqPTANP 0.059
    119 YQWSdGSNSQ 0.058
    205 NVVVtEAGII 0.049
    256 QSTLwISKST 0.049
    260 WISKsTRKES 0.047
    67 VLLSlENEAE 0.046
    145 VVMYhQPTAN 0.041
    186 PTAPvEKPYL 0.036
    43 AYFHeLSSRV 0.036
    136 EPSCgSEKCV 0.034
    250 TKTSpNQSTL 0.030
    234 AFGTcCFQML 0.028
    240 FQMLhKSKGR 0.026
    33 ADFKhPCYKM 0.026
    68 LLSLeNEAEQ 0.025
    255 NQSTlWISKS 0.017
    226 PLLLlILVAF 0.014
    73 NEAEqKLIES 0.014
    61 CESEgGVLLS 0.014
    169 RCNMkHNYIC 0.013
    138 SCGSeKCVVM 0.013
    232 LVAFgTCCFQ 0.012
    200 GDTHqNVVVT 0.010
    176 YICKyEPEIN 0.009
    215 PNLIyVVIPT 0.009
    60 ACESeGGVLL 0.009
    18 GAFCrRVVSG 0.009
    29 KVCFaDFKHP 0.009
    113 GACPdLYQWS 0.009
  • TABLE XII
    58P1D12v.1-A3-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    161 YLYQWNDDR 60.000
    171 NMKHNYICK 40.000
    258 TLWISKSTR 20.000
    70 SLENEAEQK 20.000
    227 LLLLILVAF 6.750
    241 QMLHKSKGR 3.000
    216 NLIYVVIPT 1.350
    255 NQSTLWISK 1.200
    231 ILVAFGTCC 0.900
    6 SLLLGAALL 0.900
    12 ALLCGHGAF 0.900
    86 NLTKPGTGI 0.900
    7 LLLGAALLC 0.900
    217 LIYVVIPTI 0.675
    229 LLILVAFGT 0.675
    237 TCCFQMLHK 0.600
    27 GQKVCFADF 0.540
    93 GISDGDFWI 0.540
    212 GIIPNLIYV 0.405
    78 KLIESMLQN 0.405
    41 KMAYFHELS 0.360
    24 VVSGQKVCF 0.300
    13 LLCGHGAFC 0.300
    102 GLWRNGDGQ 0.300
    67 VLLSLENEA 0.300
    156 GLGGPYLYQ 0.270
    219 YVVIPTIPL 0.270
    220 VVIPTIPLL 0.203
    232 LVAFGTCCF 0.200
    97 GDFWIGLWR 0.180
    29 KVCFADFKH 0.180
    236 GTCCFQMLH 0.180
    117 DLYQWSDGS 0.180
    221 VIPTIPLLL 0.180
    224 TIPLLLLIL 0.180
    140 GSEKCVVMY 0.180
    176 YICKYEPEI 0.180
    251 KTSPNQSTL 0.135
    14 LCGHGAFCR 0.120
    146 VMYHQPTAN 0.100
    33 ADFKHPCYK 0.100
    28 QKVCFADFK 0.090
    91 GTGISDGDF 0.090
    226 PLLLLILVA 0.090
    3 RVVSLLLGA 0.090
    213 IIPNLIYVV 0.090
    205 NVVVTEAGI 0.090
    79 LIESMLQNL 0.090
    228 LLLILVAFG 0.090
    81 ESMLQNLTK 0.090
    36 KHPCYKMAY 0.072
    51 RVSFQEARL 0.060
    8 LLGAALLCG 0.060
    144 CVVMYHQPT 0.045
    82 SMLQNLTKP 0.045
    110 QTSGACPDL 0.045
    113 GACPDLYQW 0.041
    32 FADFKHPCY 0.040
    165 WNDDRCNMK 0.040
    184 INPTAPVEK 0.040
    259 LWISKSTRK 0.030
    242 MLHKSKGRT 0.030
    121 WSDGSNSQY 0.030
    145 VVMYHQPTA 0.030
    37 HPCYKMAYF 0.030
    43 AYFHELSSR 0.030
    194 YLTNQPGDT 0.030
    222 IPTIPLLLL 0.027
    206 VVVTEAGII 0.027
    203 HQNVVVTEA 0.027
    230 LILVAFGTC 0.027
    159 GPYLYQWND 0.027
    125 SNSQYRNWY 0.024
    186 PTAPVEKPY 0.022
    223 PTIPLLLLI 0.020
    68 LLSLENEAE 0.020
    58 RLACESEGG 0.020
    111 TSGACPDLY 0.020
    50 SRVSFQEAR 0.018
    172 MKHNYICKY 0.018
    211 AGIIPNLIY 0.018
    15 CGHGAFCRR 0.018
    149 HQPTANPGL 0.018
    135 DEPSCGSEK 0.018
    47 ELSSRVSFQ 0.018
    23 RVVSGQKVC 0.015
    195 LTNQPGDTH 0.015
    66 GVLLSLENE 0.013
    63 SEGGVLLSL 0.012
    253 SPNQSTLWI 0.012
    239 CFQMLHKSK 0.010
    83 MLQNLTKPG 0.010
    21 CRRVVSGQK 0.009
    225 IPLLLLILV 0.009
    46 HELSSRVSF 0.009
    1 MSRVVSLLL 0.009
    214 IPNLIYVVI 0.009
    127 SQYRNWYTD 0.009
    4 VVSLLLGAA 0.009
    40 YKMAYFHEL 0.008
  • TABLE XIII
    58P1D12v.1-A3-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    258 TLWIsKSTRK 100.00
    13 LLCGhGAFCR 18.000
    171 NMKHnYICKY 18.000
    156 GLGGpYLYQW 12.150
    236 GTCCfQMLHK 9.000
    78 KLIEsMLQNL 6.075
    27 GQKVcFADFK 5.400
    231 ILVAfGTCCF 3.000
    216 NLIYvVIPTI 2.025
    242 MLHKsKGRTK 2.000
    161 YLYQwNDDRC 1.000
    183 EINPtAPVEK 0.900
    228 LLLIlVAFGT 0.675
    226 PLLLlILVAF 0.675
    212 GIIPnLIYVV 0.608
    70 SLENeAEQKL 0.600
    6 SLLLgAALLC 0.600
    238 CCFQmLHKSK 0.500
    102 GLWRnGDGQT 0.450
    220 VVIPtIPLLL 0.405
    257 STLWiSKSTR 0.300
    23 RVVSgQKVCF 0.300
    42 MAYFhELSSR 0.300
    194 YLTNqPGDTH 0.300
    110 QTSGaCPDLY 0.300
    12 ALLCgHGAFC 0.300
    8 LLGAaLLCGH 0.300
    229 LLILvAFGTC 0.270
    221 VIPTiPLLLL 0.270
    41 KMAYfHELSS 0.240
    69 LSLEnEAEQK 0.225
    32 FADFkHPCYK 0.200
    124 GSNSqYRNWY 0.180
    146 VMYHqPTANP 0.150
    219 YVVIpTIPLL 0.135
    7 LLLGaALLCG 0.135
    170 CNMKhNYICK 0.120
    14 LCGHgAFCRR 0.120
    154 NPGLgGPYLY 0.120
    80 IESMlQNLTK 0.120
    83 MLQNlTKPGT 0.100
    240 FQMLhKSKGR 0.090
    66 GVLLsLENEA 0.090
    227 LLLLiLVAFG 0.090
    230 LILVaFGTCC 0.090
    213 IIPNlIYVVI 0.090
    24 VVSGqKVCFA 0.090
    20 FCRRvVSGQK 0.090
    208 VTEAgIIPNL 0.068
    224 TIPLlLLILV 0.060
    49 SSRVsFQEAR 0.060
    4 VVSLlLGAAL 0.060
    58 RLACeSEGGV 0.060
    93 GISDgDFWIG 0.054
    109 GQTSgACPDL 0.054
    251 KTSPnQSTLW 0.045
    241 QMLHkSKGRT 0.045
    233 VAFGtCCFQM 0.045
    35 FKHPcYKMAY 0.036
    210 EAGIiPNLIY 0.036
    134 TDEPsCGSEK 0.030
    30 VCFAdFKHPC 0.030
    144 CVVMyHQPTA 0.030
    67 VLLSlENEAE 0.030
    163 YQWNdDRCNM 0.030
    121 WSDGsNSQYR 0.030
    91 GTGIsDGDFW 0.030
    185 NPTApVEKPY 0.030
    47 ELSSrVSFQE 0.027
    94 ISDGdFWIGL 0.027
    205 NVVVtEAGII 0.027
    140 GSEKcVVMYH 0.027
    223 PTIPlLLLIL 0.020
    68 LLSLeNEAEQ 0.020
    264 STRKeSGMEV 0.020
    164 QWNDdRCNMK 0.020
    139 CGSEkCVVMY 0.018
    152 TANPgLGGPY 0.018
    222 IPTIpLLLLI 0.018
    217 LIYVvIPTIP 0.015
    82 SMLQnLTKPG 0.015
    3 RVVSlLLGAA 0.013
    143 KCVVmYHQPT 0.013
    86 NLTKpGTGIS 0.012
    52 VSFQeARLAC 0.010
    79 LIESmLQNLT 0.010
    36 KHPCyKMAYF 0.009
    11 AALLcGHGAF 0.009
    197 NQPGdTHQNV 0.009
    127 SQYRnWYTDE 0.009
    18 GAFCrRVVSG 0.009
    207 VVTEaGIIPN 0.009
    117 DLYQwSDGSN 0.009
    225 IPLLlLILVA 0.009
    29 KVCFaDFKHP 0.009
    254 PNQStLWISK 0.008
    249 RTKTsPNQST 0.007
    62 ESEGgVLLSL 0.006
    26 SGQKvCFADF 0.006
    252 TSPNqSTLWI 0.006
  • TABLE XIV
    58P1D12v.1-A1101-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    255 NQSTLWISK 1.200
    171 NMKHNYICK 0.800
    70 SLENEAEQK 0.400
    237 TCCFQMLHK 0.400
    3 RVVSLLLGA 0.180
    29 KVCFADFKH 0.180
    161 YLYQWNDDR 0.160
    43 AYFHELSSR 0.160
    258 TLWISKSTR 0.160
    14 LCGHGAFCR 0.120
    239 CFQMLHKSK 0.100
    51 RVSFQEARL 0.060
    236 GTCCFQMLH 0.060
    219 YVVIPTIPL 0.060
    241 QMLHKSKGR 0.060
    97 GDFWIGLWR 0.048
    165 WNDDRCNMK 0.040
    33 ADFKHPCYK 0.040
    184 INPTAPVEK 0.040
    145 VVMYHQPTA 0.040
    212 GIIPNLIYV 0.036
    93 GISDGDFWI 0.036
    259 LWISKSTRK 0.030
    28 QKVCFADFK 0.030
    206 VVVTEAGII 0.030
    91 GTGISDGDF 0.030
    205 NVVVTEAGI 0.030
    251 KTSPNQSTL 0.030
    220 VVIPTIPLL 0.030
    81 ESMLQNLTK 0.024
    4 VVSLLLGAA 0.020
    232 LVAFGTCCF 0.020
    24 VVSGQKVCF 0.020
    21 CRRVVSGQK 0.020
    135 DEPSCGSEK 0.018
    27 GQKVCFADF 0.018
    113 GACPDLYQW 0.012
    110 QTSGACPDL 0.010
    195 LTNQPGDTH 0.010
    66 GVLLSLENE 0.009
    221 VIPTIPLLL 0.008
    213 IIPNLIYVV 0.008
    224 TIPLLLLIL 0.008
    217 LIYVVIPTI 0.008
    50 SRVSFQEAR 0.006
    12 ALLCGHGAF 0.006
    234 AFGTCCFQM 0.006
    203 HQNVVVTEA 0.006
    169 RCNMKHNYI 0.006
    225 IPLLLLILV 0.006
    67 VLLSLENEA 0.006
    149 HQPTANPGL 0.006
    6 SLLLGAALL 0.006
    227 LLLLILVAF 0.006
    23 RVVSGQKVC 0.005
    253 SPNQSTLWI 0.004
    222 IPTIPLLLL 0.004
    79 LIESMLQNL 0.004
    207 VVTEAGIIP 0.004
    176 YICKYEPEI 0.004
    15 CGHGAFCRR 0.004
    86 NLTKPGTGI 0.004
    122 SDGSNSQYR 0.004
    78 KLIESMLQN 0.004
    249 RTKTSPNQS 0.003
    223 PTIPLLLLI 0.003
    11 AALLCGHGA 0.003
    144 CVVMYHQPT 0.003
    156 GLGGPYLYQ 0.002
    102 GLWRNGDGQ 0.002
    127 SQYRNWYTD 0.002
    159 GPYLYQWND 0.002
    243 LHKSKGRTK 0.002
    198 QPGDTHQNV 0.002
    214 IPNLIYVVI 0.002
    32 FADFKHPCY 0.002
    59 LACESEGGV 0.002
    60 ACESEGGVL 0.002
    44 YFHELSSRV 0.002
    208 VTEAGIIPN 0.002
    37 HPCYKMAYF 0.002
    154 NPGLGGPYL 0.002
    138 SCGSEKCVV 0.002
    187 TAPVEKPYL 0.002
    229 LLILVAFGT 0.002
    109 GQTSGACPD 0.002
    76 EQKLIESML 0.002
    7 LLLGAALLC 0.001
    39 CYKMAYFHE 0.001
    240 FQMLHKSKG 0.001
    216 NLIYVVIPT 0.001
    36 KHPCYKMAY 0.001
    41 KMAYFHELS 0.001
    179 KYEPEINPT 0.001
    119 YQWSDGSNS 0.001
    192 KPYLTNQPG 0.001
    58 RLACESEGG 0.001
    226 PLLLLILVA 0.001
    18 GAFCRRVVS 0.001
    63 SEGGVLLSL 0.001
  • TABLE XV
    58P1D12v.1-A1101-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    236 GTCCfQMLHK 6.000
    27 GQKVcFADFK 1.800
    258 TLWIsKSTRK 0.800
    257 STLWiSKSTR 0.300
    13 LLCGhGAFCR 0.240
    20 FCRRvVSGQK 0.200
    238 CCFQmLHKSK 0.200
    32 FADFkHPCYK 0.200
    170 CNMKhNYICK 0.160
    80 IESMlQNLTK 0.120
    240 FQMLhKSKGR 0.120
    183 EINPtAPVEK 0.120
    3 RVVSlLLGAA 0.090
    66 GVLLsLENEA 0.090
    23 RVVSgQKVCF 0.090
    42 MAYFhELSSR 0.080
    220 VVIPtIPLLL 0.060
    14 LCGHgAFCRR 0.040
    242 MLHKsKGRTK 0.040
    212 GIIPnLIYVV 0.036
    69 LSLEnEAEQK 0.030
    205 NVVVtEAGII 0.030
    219 YVVIpTIPLL 0.030
    91 GTGIsDGDFW 0.030
    144 CVVMyHQPTA 0.030
    251 KTSPnQSTLW 0.030
    156 GLGGpYLYQW 0.024
    4 VVSLlLGAAL 0.020
    24 VVSGqKVCFA 0.020
    134 TDEPsCGSEK 0.020
    164 QWNDdRCNMK 0.020
    264 STRKeSGMEV 0.020
    78 KLIEsMLQNL 0.018
    109 GQTSgACPDL 0.018
    160 PYLYqWNDDR 0.012
    218 IYVViPTIPL 0.012
    233 VAFGtCCFQM 0.012
    179 KYEPeINPTA 0.012
    58 RLACeSEGGV 0.012
    163 YQWNdDRCNM 0.012
    208 VTEAgIIPNL 0.010
    110 QTSGaCPDLY 0.010
    254 PNQStLWISK 0.008
    224 TIPLlLLILV 0.008
    43 AYFHeLSSRV 0.008
    221 VIPTiPLLLL 0.008
    51 RVSFqEARLA 0.006
    225 IPLLlLILVA 0.006
    175 NYICkYEPEI 0.006
    10 GAALlCGHGA 0.006
    206 VVVTeAGIIP 0.006
    197 NQPGdTHQNV 0.006
    231 ILVAfGTCCF 0.006
    216 NLIYvVIPTI 0.006
    39 CYKMaYFHEL 0.004
    145 VVMYhQPTAN 0.004
    213 IIPNlIYVVI 0.004
    49 SSRVsFQEAR 0.004
    70 SLENeAEQKL 0.004
    207 VVTEaGIIPN 0.004
    222 IPTIpLLLLI 0.004
    171 NMKHnYICKY 0.004
    194 YLTNqPGDTH 0.004
    154 NPGLgGPYLY 0.004
    37 HPCYkMAYFH 0.004
    8 LLGAaLLCGH 0.004
    121 WSDGsNSQYR 0.004
    11 AALLcGHGAF 0.003
    223 PTIPlLLLIL 0.003
    29 KVCFaDFKHP 0.003
    249 RTKTsPNQST 0.003
    41 KMAYfHELSS 0.002
    102 GLWRnGDGQT 0.002
    93 GISDgDFWIG 0.002
    96 DGDFwIGLWR 0.002
    232 LVAFgTCCFQ 0.002
    198 QPGDtHQNVV 0.002
    60 ACESeGGVLL 0.002
    87 LTKPgTGISD 0.002
    152 TANPgLGGPY 0.002
    234 AFGTcCFQML 0.002
    138 SCGSeKCVVM 0.002
    59 LACEsEGGVL 0.002
    31 CFADfKHPCY 0.002
    228 LLLIlVAFGT 0.002
    6 SLLLgAALLC 0.001
    105 RNGDgQTSGA 0.001
    140 GSEKcVVMYH 0.001
    192 KPYLtNQPGD 0.001
    169 RCNMkHNYIC 0.001
    18 GAFCrRVVSG 0.001
    127 SQYRnWYTDE 0.001
    119 YQWSdGSNSQ 0.001
    7 LLLGaALLCG 0.001
    159 GPYLyQWNDD 0.001
    210 EAGIiPNLIY 0.001
    185 NPTApVEKPY 0.001
    133 YTDEpSCGSE 0.001
    186 PTAPvEKPYL 0.001
    195 LTNQpGDTHQ 0.001
  • TABLE XVI
    58P1D12v.1-A24-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    179 KYEPEINPT 21.600
    251 KTSPNQSTL 9.600
    221 VIPTIPLLL 8.400
    51 RVSFQEARL 8.000
    162 LYQWNDDRC 7.500
    118 LYQWSDGSN 7.500
    5 VSLLLGAAL 7.200
    224 TIPLLLLIL 7.200
    79 LIESMLQNL 7.200
    220 VVIPTIPLL 7.200
    187 TAPVEKPYL 6.000
    6 SLLLGAALL 6.000
    219 YVVIPTIPL 6.000
    149 HQPTANPGL 6.000
    60 ACESEGGVL 6.000
    235 FGTCCFQML 5.760
    76 EQKLIESML 5.600
    1 MSRVVSLLL 5.600
    227 LLLLILVAF 4.200
    222 IPTIPLLLL 4.000
    110 QTSGACPDL 4.000
    154 NPGLGGPYL 4.000
    169 RCNMKHNYI 3.600
    12 ALLCGHGAF 3.000
    34 DFKHPCYKM 2.750
    234 AFGTCCFQM 2.500
    27 GQKVCFADF 2.400
    214 IPNLIYVVI 2.100
    232 LVAFGTCCF 2.000
    91 GTGISDGDF 2.000
    37 HPCYKMAYF 2.000
    24 VVSGQKVCF 2.000
    210 EAGIIPNLI 1.680
    72 ENEAEQKLI 1.500
    253 SPNQSTLWI 1.500
    206 VVVTEAGII 1.500
    205 NVVVTEAGI 1.500
    217 LIYVVIPTI 1.400
    93 GISDGDFWI 1.200
    176 YICKYEPEI 1.100
    218 IYVVIPTIP 1.050
    263 KSTRKESGM 1.000
    86 NLTKPGTGI 1.000
    71 LENEAEQKL 0.950
    164 QWNDDRCNM 0.900
    53 SFQEARLAC 0.900
    40 YKMAYFHEL 0.792
    175 NYICKYEPE 0.750
    44 YFHELSSRV 0.720
    31 CFADFKHPC 0.600
    139 CGSEKCVVM 0.600
    132 WYTDEPSCG 0.600
    63 SEGGVLLSL 0.560
    209 TEAGIIPNL 0.560
    128 QYRNWYTDE 0.500
    43 AYFHELSSR 0.500
    98 DFWIGLWRN 0.500
    39 CYKMAYFHE 0.500
    147 MYHQPTANP 0.500
    95 SDGDFWIGL 0.480
    61 CESEGGVLL 0.480
    3 RVVSLLLGA 0.360
    78 KLIESMLQN 0.360
    23 RVVSGQKVC 0.300
    46 HELSSRVSF 0.300
    114 ACPDLYQWS 0.259
    249 RTKTSPNQS 0.240
    67 VLLSLENEA 0.238
    203 HQNVVVTEA 0.231
    223 PTIPLLLLI 0.216
    158 GGPYLYQWN 0.216
    257 STLWISKST 0.210
    229 LLILVAFGT 0.210
    216 NLIYVVIPT 0.210
    41 KMAYFHELS 0.200
    130 RNWYTDEPS 0.200
    247 KGRTKTSPN 0.200
    245 KSKGRTKTS 0.200
    65 GGVLLSLEN 0.198
    212 GIIPNLIYV 0.180
    213 IIPNLIYVV 0.180
    144 CVVMYHQPT 0.180
    225 IPLLLLILV 0.180
    188 APVEKPYLT 0.180
    230 LILVAFGTC 0.180
    153 ANPGLGGPY 0.180
    74 EAEQKLIES 0.165
    49 SSRVSFQEA 0.158
    124 GSNSQYRNW 0.150
    170 CNMKHNYIC 0.150
    92 TGISDGDFW 0.150
    231 ILVAFGTCC 0.150
    252 TSPNQSTLW 0.150
    7 LLLGAALLC 0.150
    11 AALLCGHGA 0.150
    197 NQPGDTHQN 0.150
    62 ESEGGVLLS 0.150
    140 GSEKCVVMY 0.150
    211 AGIIPNLIY 0.150
    145 VVMYHQPTA 0.150
  • TABLE XVII
    58P1D12v.1-A24-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    218 IYVViPTIPL 300.000
    39 CYKMaYFHEL 264.000
    175 NYICkYEPEI 82.500
    234 AFGTcCFQML 28.800
    179 KYEPeINPTA 25.200
    78 KLIEsMLQNL 17.280
    220 VVIPtIPLLL 10.080
    208 VTEAgIIPNL 8.400
    62 ESEGgVLLSL 8.400
    70 SLENeAEQKL 7.920
    118 LYQWsDGSNS 7.500
    162 LYQWnDDRCN 7.500
    60 ACESeGGVLL 6.000
    43 AYFHeLSSRV 6.000
    153 ANPGlGGPYL 6.000
    219 YVVIpTIPLL 6.000
    23 RVVSgQKVCF 6.000
    221 VIPTiPLLLL 6.000
    5 VSLLlGAALL 6.000
    132 WYTDePSCGS 6.000
    59 LACEsEGGVL 4.800
    4 VVSLlLGAAL 4.800
    94 ISDGdFWIGL 4.000
    109 GQTSgACPDL 4.000
    26 SGQKvCFADF 3.600
    11 AALLcGHGAF 3.000
    231 ILVAfGTCCF 3.000
    213 IIPNIlYVVI 2.100
    216 NLIYvVIPTI 2.100
    204 QNVVvTEAGI 1.500
    205 NVVVtEAGII 1.500
    85 QNLTkPGTGI 1.500
    252 TSPNqSTLWI 1.500
    92 TGISdGDFWI 1.500
    222 IPTIpLLLLI 1.200
    74 EAEQkLIESM 0.900
    223 PTIPlLLLIL 0.864
    75 AEQKlIESML 0.840
    193 PYLTnQPGDT 0.750
    148 YHQPtANPGL 0.720
    44 YFHElSSRVS 0.600
    147 MYHQpTANPG 0.600
    50 SRVSfQEARL 0.600
    31 CFADfKHPCY 0.600
    36 KHPCyKMAYF 0.600
    128 QYRNwYTDEP 0.550
    233 VAFGtCCFQM 0.500
    34 DFKHpCYKMA 0.500
    163 YQWNdDRCNM 0.500
    138 SCGSeKCVVM 0.500
    186 PTAPvEKPYL 0.480
    226 PLLLlILVAF 0.420
    250 TKTSpNQSTL 0.400
    3 RVVSlLLGAA 0.360
    143 KCVVmYHQPT 0.360
    169 RCNMkHNYIC 0.300
    45 FHELsSRVSF 0.300
    105 RNGDgQTSGA 0.240
    249 RTKTsPNQST 0.240
    251 KTSPnQSTLW 0.240
    66 GVLLsLENEA 0.238
    79 LIESmLQNLT 0.216
    212 GIIPnLIYVV 0.216
    152 TANPgLGGPY 0.216
    228 LLLIlVAFGT 0.210
    130 RNWYtDEPSC 0.200
    51 RVSFqEARLA 0.200
    90 PGTGiSDGDF 0.200
    41 KMAYfHELSS 0.200
    58 RLACeSEGGV 0.200
    54 FQEArLACES 0.198
    71 LENEaEQKLI 0.180
    188 APVEkPYLTN 0.180
    196 TNQPgDTHQN 0.180
    224 TIPLlLLILV 0.180
    197 NQPGdTHQNV 0.180
    225 IPLLlLILVA 0.180
    229 LLILvAFGTC 0.180
    124 GSNSqYRNWY 0.180
    113 GACPdLYQWS 0.173
    157 LGGPyLYQWN 0.173
    209 TEAGiIPNLI 0.168
    48 LSSRvSFQEA 0.158
    6 SLLLgAALLC 0.150
    181 EPEInPTAPV 0.150
    187 TAPVeKPYLT 0.150
    253 SPNQsTLWIS 0.150
    12 ALLCgHGAFC 0.150
    83 MLQNlTKPGT 0.150
    211 AGIIpNLIYV 0.150
    230 LILVaFGTCC 0.150
    241 QMLHkSKGRT 0.150
    145 VVMYhQPTAN 0.150
    144 CVVMyHQPTA 0.150
    198 QPGDtHQNVV 0.144
    256 QSTLwISKST 0.140
    185 NPTApVEKPY 0.140
    64 EGGVlLSLEN 0.132
    103 LWRNgDGQTS 0.120
    139 CGSEkCVVMY 0.120
  • TABLE XVIII
    58P1D12v.1-B7-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    222 IPTIPLLLL 120.000
    154 NPGLGGPYL 80.000
    1 MSRVVSLLL 40.000
    219 YVVIPTIPL 30.000
    220 VVIPTIPLL 20.000
    51 RVSFQEARL 20.000
    187 TAPVEKPYL 12.000
    214 IPNLIYVVI 8.000
    253 SPNQSTLWI 8.000
    188 APVEKPYLT 6.000
    110 QTSGACPDL 4.000
    221 VIPTIPLLL 4.000
    149 HQPTANPGL 4.000
    224 TIPLLLLIL 4.000
    225 IPLLLLILV 4.000
    76 EQKLIESML 4.000
    235 FGTCCFQML 4.000
    198 QPGDTHQNV 4.000
    6 SLLLGAALL 4.000
    5 VSLLLGAAL 4.000
    251 KTSPNQSTL 4.000
    60 ACESEGGVL 3.600
    205 NVVVTEAGI 2.000
    136 EPSCGSEKC 2.000
    206 VVVTEAGII 2.000
    145 VVMYHQPTA 1.500
    79 LIESMLQNL 1.200
    210 EAGIIPNLI 1.200
    40 YKMAYFHEL 1.200
    139 CGSEKCVVM 1.000
    263 KSTRKESGM 1.000
    49 SSRVSFQEA 1.000
    11 AALLCGHGA 0.900
    86 NLTKPGTGI 0.600
    59 LACESEGGV 0.600
    23 RVVSGQKVC 0.500
    3 RVVSLLLGA 0.500
    4 VVSLLLGAA 0.500
    144 CVVMYHQPT 0.500
    217 LIYVVIPTI 0.400
    63 SEGGVLLSL 0.400
    176 YICKYEPEI 0.400
    209 TEAGIIPNL 0.400
    61 CESEGGVLL 0.400
    93 GISDGDFWI 0.400
    37 HPCYKMAYF 0 400
    71 LENEAEQKL 0.400
    95 SDGDFWIGL 0.400
    169 RCNMKHNYI 0.400
    75 AEQKLIESM 0.300
    56 EARLACESE 0.300
    185 NPTAPVEKP 0.300
    17 HGAFCRRVV 0.300
    170 CNMKHNYIC 0.300
    234 AFGTCCFQM 0.300
    138 SCGSEKCVV 0.200
    150 QPTANPGLG 0.200
    89 KPGTGISDG 0.200
    247 KGRTKTSPN 0.200
    213 IIPNLIYVV 0.200
    212 GIIPNLIYV 0.200
    192 KPYLTNQPG 0.200
    159 GPYLYQWND 0.200
    164 QWNDDRCNM 0.150
    34 DFKHPCYKM 0.150
    72 ENEAEQKLI 0.120
    25 VSGQKVCFA 0.100
    203 HQNVVVTEA 0.100
    257 STLWISKST 0.100
    232 LVAFGTCCF 0.100
    67 VLLSLENEA 0.100
    242 MLHKSKGRT 0.100
    229 LLILVAFGT 0.100
    52 VSFQEARLA 0.100
    103 LWRNGDGQT 0.100
    201 DTHQNVVVT 0.100
    231 ILVAFGTCC 0.100
    84 LQNLTKPGT 0.100
    24 VVSGQKVCF 0.100
    7 LLLGAALLC 0.100
    20 FCRRVVSGQ 0.100
    216 NLIYVVIPT 0.100
    13 LLCGHGAFC 0.100
    264 STRKESGME 0.100
    126 NSQYRNWYT 0.100
    194 YLTNQPGDT 0.100
    230 LILVAFGTC 0.100
    211 AGIIPNLIY 0.090
    18 GAFCRRVVS 0.090
    114 ACPDLYQWS 0.060
    115 CPDLYQWSD 0.060
    181 EPEINPTAP 0.060
    42 MAYFHELSS 0.060
    12 ALLCGHGAF 0.060
    153 ANPGLGGPY 0.060
    113 GACPDLYQW 0.060
    66 GVLLSLENE 0.050
    29 KVCFADFKH 0.050
    207 VVTEAGIIP 0.050
    223 PTIPLLLLI 0.040
  • TABLE XIX
    58P1D12v.1-B7-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    220 VVIPtIPLLL 20.000
    219 YVVIpTIPLL 20.000
    4 VVSLlLGAAL 20.000
    153 ANPGlGGPYL 12.000
    59 LACEsEGGVL 12.000
    222 IPTIpLLLLI 8.000
    221 VIPTiPLLLL 6.000
    5 VSLLlGAALL 4.000
    109 GQTSgACPDL 4.000
    136 EPSCgSEKCV 4.000
    198 QPGDtHQNVV 4.000
    78 KLIEsMLQNL 4.000
    60 ACESeGGVLL 3.600
    233 VAFGtCCFQM 3.000
    205 NVVVtEAGII 2.000
    225 IPLLlLILVA 2.000
    264 STRKeSGMEV 2.000
    181 EPEInPTAPV 1.800
    188 APVEkPYLTN 1.800
    163 YQWNdDRCNM 1.500
    94 ISDGdFWIGL 1.200
    70 SLENeAEQKL 1.200
    208 VTEAgIIPNL 1.200
    75 AEQKlIESML 1.200
    62 ESEGgVLLSL 1.200
    234 AFGTcCFQML 1.200
    138 SCGSeKCVVM 1.000
    74 EAEQkLIESM 0.900
    211 AGIIpNLIYV 0.600
    218 IYVViPTIPL 0.600
    85 QNLTkPGTGI 0.600
    24 VVSGqKVCFA 0.500
    144 CVVMyHQPTA 0.500
    3 RVVSlLLGAA 0.500
    51 RVSFqEARLA 0.500
    66 GVLLsLENEA 0.500
    33 ADFKhPCYKM 0.450
    223 PTIPlLLLIL 0.400
    92 TGISdGDFWI 0.400
    253 SPNQsTLWIS 0.400
    154 NPGLgGPYLY 0.400
    250 TKTSpNQSTL 0.400
    148 YHQPtANPGL 0.400
    204 QNVVvTEAGI 0.400
    185 NPTApVEKPY 0.400
    50 SRVSfQEARL 0.400
    39 CYKMaYFHEL 0.400
    213 IIPNlIYVVI 0.400
    216 NLIYvVIPTI 0.400
    252 TSPNqSTLWI 0.400
    186 PTAPvEKPYL 0.400
    145 VVMYhQPTAN 0.300
    10 GAALlCGHGA 0.300
    150 QPTAnPGLGG 0.300
    187 TAPVeKPYLT 0.300
    56 EARLaCESEG 0.300
    12 ALLCgHGAFC 0.300
    37 HPCYkMAYFH 0.200
    224 TIPLlLLILV 0.200
    58 RLACeSEGGV 0.200
    192 KPYLtNQPGD 0.200
    15 CGHGaFCRRV 0.200
    214 IPNLiYVVIP 0.200
    89 KPGTgISDGD 0.200
    197 NQPGdTHQNV 0.200
    212 GIIPnLIYVV 0.200
    159 GPYLyQWNDD 0.200
    21 CRRVvSGQKV 0.200
    11 AALLcGHGAF 0.180
    249 RTKTsPNQST 0.150
    52 VSFQeARLAC 0.150
    83 MLQNlTKPGT 0.100
    1 MSRVvSLLLG 0.100
    229 LLILvAFGTC 0.100
    49 SSRVsFQEAR 0.100
    143 KCVVmYHQPT 0.100
    105 RNGDgQTSGA 0.100
    125 SNSQyRNWYT 0.100
    6 SLLLgAALLC 0.100
    207 VVTEaGIIPN 0.100
    102 GLWRnGDGQT 0.100
    262 SKSTrKESGM 0.100
    161 YLYQwNDDRC 0.100
    23 RVVSgQKVCF 0.100
    30 VCFAdFKHPC 0.100
    169 RCNMkHNYIC 0.100
    48 LSSRvSFQEA 0.100
    247 KGRTkTSPNQ 0.100
    228 LLLIlVAFGT 0.100
    230 LILVaFGTCC 0.100
    130 RNWYtDEPSC 0.100
    20 FCRRvVSGQK 0.100
    256 QSTLwISKST 0.100
    241 QMLHkSKGRT 0.100
    210 EAGIiPNLIY 0.090
    113 GACPdLYQWS 0.060
    152 TANPgLGGPY 0.060
    115 CPDLyQWSDG 0.060
    43 AYFHeLSSRV 0.060
    232 LVAFgTCCFQ 0.050
  • TABLE XX
    58P1D12v.1-B3501-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position isspecified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    263 KSTRKESGM 20.000
    222 IPTIPLLLL 20.000
    37 HPCYKMAYF 20.000
    154 NPGLGGPYL 20.000
    1 MSRVVSLLL 15.000
    111 TSGACPDLY 10.000
    214 IPNLIYVVI 8.000
    198 QPGDTHQNV 8.000
    253 SPNQSTLWI 8.000
    5 VSLLLGAAL 5.000
    187 TAPVEKPYL 4.500
    225 IPLLLLILV 4.000
    139 CGSEKCVVM 4.000
    188 APVEKPYLT 4.000
    245 KSKGRTKTS 3.000
    121 WSDGSNSQY 3.000
    76 EQKLIESML 3.000
    27 GQKVCFADF 3.000
    140 GSEKCVVMY 3.000
    252 TSPNQSTLW 2.500
    124 GSNSQYRNW 2.500
    113 GACPDLYQW 2.250
    211 AGIIPNLIY 2.000
    51 RVSFQEARL 2.000
    251 KTSPNQSTL 2.000
    136 EPSCGSEKC 2.000
    153 ANPGLGGPY 2.000
    125 SNSQYRNWY 2.000
    32 FADFKHPCY 1.800
    261 ISKSTRKES 1.500
    49 SSRVSFQEA 1.500
    59 LACESEGGV 1.200
    210 EAGIIPNLI 1.200
    6 SLLLGAALL 1.000
    219 YVVIPTIPL 1.000
    149 HQPTANPGL 1.000
    24 VVSGQKVCF 1.000
    227 LLLLILVAF 1.000
    12 ALLCGHGAF 1.000
    235 FGTCCFQML 1.000
    91 GTGISDGDF 1.000
    110 QTSGACPDL 1.000
    221 VIPTIPLLL 1.000
    220 VVIPTIPLL 1.000
    232 LVAFGTCCF 1.000
    224 TIPLLLLIL 1.000
    93 GISDGDFWI 0.800
    169 RCNMKHNYI 0.800
    92 TGISDGDFW 0.750
    52 VSFQEARLA 0.750
    249 RTKTSPNQS 0.600
    206 VVVTEAGII 0.600
    34 DFKHPCYKM 0.600
    164 QWNDDRCNM 0.600
    247 KGRTKTSPN 0.600
    25 VSGQKVCFA 0.500
    256 QSTLWISKS 0.500
    126 NSQYRNWYT 0.500
    157 LGGPYLYQW 0.500
    60 ACESEGGVL 0.450
    177 ICKYEPEIN 0.450
    217 LIYVVIPTI 0.400
    86 NLTKPGTGI 0.400
    36 KHPCYKMAY 0.400
    205 NVVVTEAGI 0.400
    89 KPGTGISDG 0.400
    176 YICKYEPEI 0.400
    78 KLIESMLQN 0.400
    192 KPYLTNQPG 0.400
    11 AALLCGHGA 0.300
    42 MAYFHELSS 0.300
    87 LTKPGTGIS 0.300
    18 GAFCRRVVS 0.300
    79 LIESMLQNL 0.300
    138 SCGSEKCVV 0.300
    172 MKHNYICKY 0.200
    114 ACPDLYQWS 0.200
    212 GIIPNLIYV 0.200
    95 SDGDFWIGL 0.200
    41 KMAYFHELS 0.200
    159 GPYLYQWND 0.200
    3 RVVSLLLGA 0.200
    234 AFGTCCFQM 0.200
    17 HGAFCRRVV 0.200
    71 LENEAEQKL 0.200
    61 CESEGGVLL 0.200
    168 DRCNMKHNY 0.200
    75 AEQKLIESM 0.200
    213 IIPNLIYVV 0.200
    23 RVVSGQKVC 0.200
    150 QPTANPGLG 0.200
    155 PGLGGPYLY 0.200
    130 RNWYTDEPS 0.200
    186 PTAPVEKPY 0.200
    185 NPTAPVEKP 0.200
    72 ENEAEQKLI 0.180
    96 DGDFWIGLW 0.150
    197 NQPGDTHQN 0.150
    62 ESEGGVLLS 0.150
    119 YQWSDGSNS 0.150
  • TABLE XXI
    58P1D12v.1-B3501-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    185 NPTApVEKPY 40.000
    154 NPGLgGPYLY 40.000
    124 GSNSqYRNWY 10.000
    59 LACEsEGGVL 9.000
    222 IPTIpLLLLI 8.000
    198 QPGDtHQNVV 8.000
    210 EAGIiPNLIY 6.000
    233 VAFGtCCFQM 6.000
    152 TANPgLGGPY 6.000
    171 NMKHnYICKY 6.000
    5 VSLLlGAALL 5.000
    136 EPSCgSEKCV 4.000
    139 CGSEkCVVMY 4.000
    78 KLIEsMLQNL 4.000
    188 APVEkPYLTN 4.000
    163 YQWNdDRCNM 3.000
    11 AALLcGHGAF 3.000
    225 IPLLlLILVA 2.000
    138 SCGSeKCVVM 2.000
    252 TSPNqSTLWI 2.000
    110 QTSGaCPDLY 2.000
    253 SPNQsTLWIS 2.000
    23 RVVSgQKVCF 2.000
    74 EAEQkLIESM 1.800
    94 ISDGdFWIGL 1.500
    62 ESEGgVLLSL 1.500
    181 EPEInPTAPV 1.200
    109 GQTSgACPDL 1.000
    219 YVVIpTIPLL 1.000
    4 VVSLlLGAAL 1.000
    221 VIPTiPLLLL 1.000
    26 SGQKvCFADF 1.000
    231 ILVAfGTCCF 1.000
    220 VVIPtIPLLL 1.000
    153 ANPGlGGPYL 1.000
    251 KTSPnQSTLW 1.000
    112 SGACpDLYQW 0.750
    91 GTGIsDGDFW 0.750
    205 NVVVtEAGII 0.600
    249 RTKTsPNQST 0.600
    167 DDRCnMKHNY 0.600
    264 STRKeSGMEV 0.600
    256 QSTLwISKST 0.500
    52 VSFQeARLAC 0.500
    123 DGSNsQYRNW 0.500
    48 LSSRvSFQEA 0.500
    156 GLGGpYLYQW 0.500
    105 RNGDgQTSGA 0.400
    192 KPYLtNQPGD 0.400
    89 KPGTgISDGD 0.400
    85 QNLTkPGTGI 0.400
    120 QWSDgSNSQY 0.400
    216 NLIYvVIPTI 0.400
    58 RLACeSEGGV 0.400
    92 TGISdGDFWI 0.400
    204 QNVVvTEAGI 0.400
    213 IIPNlIYVVI 0.400
    31 CFADfKHPCY 0.400
    245 KSKGrTKTSP 0.300
    51 RVSFqEARLA 0.300
    208 VTEAgIIPNL 0.300
    39 CYKMaYFHEL 0.300
    187 TAPVeKPYLT 0.300
    113 GACPdLYQWS 0.300
    70 SLENeAEQKL 0.300
    60 ACESeGGVLL 0.300
    10 GAALlCGHGA 0.300
    130 RNWYtDEPSC 0.300
    35 FKHPcYKMAY 0.200
    143 KCVVmYHQPT 0.200
    214 IPNLiYVVIP 0.200
    159 GPYLyQWNDD 0.200
    15 CGHGaFCRRV 0.200
    207 VVTEaGIIPN 0.200
    37 HPCYkMAYFH 0.200
    33 ADFKhPCYKM 0.200
    150 QPTAnPGLGG 0.200
    212 GIIPnLIYVV 0.200
    3 RVVSlLLGAA 0.200
    224 TIPLlLLILV 0.200
    211 AGIIpNLIYV 0.200
    262 SKSTrKESGM 0.200
    36 KHPCyKMAYF 0.200
    169 RCNMkHNYIC 0.200
    197 NQPGdTHQNV 0.200
    41 KMAYfHELSS 0.200
    186 PTAPvEKPYL 0.150
    261 ISKStRKESG 0.150
    263 KSTRkESGME 0.150
    137 PSCGsEKCVV 0.150
    196 TNQPgDTHQN 0.150
    1 MSRVvSLLLG 0.150
    49 SSRVsFQEAR 0.150
    69 LSLEnEAEQK 0.150
    176 YICKyEPEIN 0.150
    71 LENEaEQKLI 0.120
    228 LLLIlVAFGT 0.100
    237 TCCFqMLHKS 0.100
    161 YLYQwNDDRC 0.100
    229 LLILvAFGTC 0.100
  • TABLE VIII
    58P1D12v.4-A1-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    60 SLEWLEITK 90.000
    5 VTEAVKEEQ 2.250
    27 VPEKKVAWK 0.900
    63 WLEITKDLQ 0.900
    18 TSLHCGFQR 0.750
    48 ILDLYKEWH 0.500
    2 NVVVTEAVK 0.400
    9 VKEEQKLVQ 0.225
    38 NSLTWFQNF 0.150
    44 QNFVILDLY 0.125
    22 CGFQRVPEK 0.100
    10 KEEQKLVQT 0.090
    14 KLVQTSLHC 0.050
    45 NFVILDLYK 0.050
    52 YKEWHQNNS 0.045
    17 QTSLHCGFQ 0.025
    26 RVPEKKVAW 0.020
    59 NSLEWLEIT 0.015
    16 VQTSLHCGF 0.015
    37 NNSLTWFQN 0.013
    58 NNSLEWLEI 0.013
    40 LTWFQNFVI 0.013
    31 KVAWKYNNS 0.010
    39 SLTWFQNFV 0.010
    15 LVQTSLHCG 0.010
    6 TEAVKEEQK 0.010
    47 VILDLYKEW 0.010
    19 SLHCGFQRV 0.010
    43 FQNFVILDL 0.007
    25 QRVPEKKVA 0.005
    35 KYNNSLTWF 0.005
    55 WHQNNSLEW 0.003
    66 ITKDLQDEL 0.003
    34 WKYNNSLTW 0.003
    36 YNNSLTWFQ 0.003
    1 QNVVVTEAV 0.003
    46 FVILDLYKE 0.002
    3 VVVTEAVKE 0.002
    21 HCGFQRVPE 0.002
    56 HQNNSLEWL 0.002
    23 GFQRVPEKK 0.001
    30 KKVAWKYNN 0.001
    41 TWFQNFVIL 0.001
    8 AVKEEQKLV 0.001
    4 VVTEAVKEE 0.001
    7 EAVKEEQKL 0.001
    50 DLYKEWHQN 0.001
    13 QKLVQTSLH 0.001
    49 LDLYKEWHQ 0.001
    62 EWLEITKDL 0.001
    64 LEITKDLQD 0.000
    42 WFQNFVILD 0.000
    57 QNNSLEWLE 0.000
    12 EQKLVQTSL 0.000
    24 FQRVPEKKV 0.000
    32 VAWKYNNSL 0.000
    65 EITKDLQDE 0.000
    29 EKKVAWKYN 0.000
    54 EWHQNNSLE 0.000
    51 LYKEWHQNN 0.000
    20 LHCGFQRVP 0.000
    11 EEQKLVQTS 0.000
    33 AWKYNNSLT 0.000
    53 KEWHQNNSL 0.000
    28 PEKKVAWKY 0.000
    61 LEWLEITKD 0.000
  • TABLE IX
    58P1D12v.4-A1-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    6 VTEAvKEEQK 45.000
    60 NSLEwLEITK 1.500
    18 QTSLhCGFQR 1.250
    28 VPEKkVAWKY 1.125
    49 ILDLyKEWHQ 0.500
    64 WLEItKDLQD 0.450
    27 RVPEkKVAWK 0.400
    22 HCGFqRVPEK 0.400
    45 QNFViLDLYK 0.250
    16 LVQTsLHCGF 0.100
    2 QNVVvTEAVK 0.100
    10 VKEEqKLVQT 0.090
    61 SLEWlEITKD 0.090
    44 FQNFvILDLY 0.075
    40 SLTWfQNFVI 0.050
    23 CGFQrVPEKK 0.050
    53 YKEWhQNNSL 0.045
    11 KEEQkLVQTS 0.045
    38 NNSLtWFQNF 0.025
    19 TSLHcGFQRV 0.015
    39 NSLTwFQNFV 0.015
    58 QNNSlEWLEI 0.013
    37 YNNSlTWFQN 0.013
    32 KVAWkYNNSL 0.010
    48 VILDlYKEWH 0.010
    26 QRVPeKKVAW 0.010
    15 KLVQtSLHCG 0.010
    47 FVILdLYKEW 0.010
    5 VVTEaVKEEQ 0.010
    41 LTWFqNFVIL 0.005
    9 AVKEeQKLVQ 0.005
    35 WKYNnSLTWF 0.005
    59 NNSLeWLEIT 0.003
    14 QKLVqTSLHC 0.003
    43 WFQNfVILDL 0.003
    55 EWHQnNSLEW 0.003
    3 NVVVtEAVKE 0.002
    17 VQTSlHCGFQ 0.002
    1 HQNVvVTEAV 0.002
    8 EAVKeEQKLV 0.001
    66 EITKdLQDEL 0.001
    20 SLHCgFQRVP 0.001
    51 DLYKeWHQNN 0.001
    4 VVVTeAVKEE 0.001
    31 KKVAwKYNNS 0.001
    36 KYNNsLTWFQ 0.001
    56 WHQNnSLEWL 0.001
    50 LDLYkEWHQN 0.001
    63 EWLEiTKDLQ 0.001
    7 TEAVkEEQKL 0.001
    34 AWKYnNSLTW 0.000
    42 TWFQnFVILD 0.000
    57 HQNNsLEWLE 0.000
    25 FORVpEKKVA 0.000
    13 EQKLvQTSLH 0.000
    46 NFVIlDLYKE 0.000
    21 LHCGfQRVPE 0.000
    33 VAWKyNNSLT 0.000
    30 EKKVaWKYNN 0.000
    52 LYKEwHQNNS 0.000
    24 GFQRvPEKKV 0.000
    65 LEITkDLQDE 0.000
    12 EEQKlVQTSL 0.000
    62 LEWLeITKDL 0.000
    54 KEWHqNNSLE 0.000
    29 PEKKvAWKYN 0.000
  • TABLE X
    58P1D12v.4-A0201-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    39 SLTWFQNFV 1453.637
    19 SLHCGFQRV 132.149
    43 FQNFVILDL 22.915
    14 KLVQTSLHC 17.388
    53 KEWHQNNSL 12.067
    40 LTWFQNFVI 4.862
    32 VAWKYNNSL 4.186
    24 FQRVPEKKV 2.465
    8 AVKEEQKLV 0.873
    56 HQNNSLEWL 0.456
    47 VILDLYKEW 0.396
    59 NSLEWLEIT 0.341
    1 QNVVVTEAV 0.222
    48 ILDLYKEWH 0.137
    58 NNSLEWLEI 0.116
    10 KEEQKLVQT 0.075
    66 ITKDLQDEL 0.035
    31 KVAWKYNNS 0.034
    50 DLYKEWHQN 0.030
    26 RVPEKKVAW 0.027
    15 LVQTSLHCG 0.025
    7 EAVKEEQKL 0.022
    4 VVTEAVKEE 0.021
    36 YNNSLTWFQ 0.018
    46 FVILDLYKE 0.014
    16 VQTSLHCGF 0.013
    61 LEWLEITKD 0.009
    41 TWFQNFVIL 0.009
    12 EQKLVQTSL 0.006
    37 NNSLTWFQN 0.005
    38 NSLTWFQNF 0.003
    62 EWLEITKDL 0.003
    3 VVVTEAVKE 0.002
    63 WLEITKDLQ 0.002
    18 TSLHCGFQR 0.002
    44 QNFVILDLY 0.002
    34 WKYNNSLTW 0.002
    49 LDLYKEWHQ 0.001
    60 SLEWLEITK 0.001
    22 CGFQRVPEK 0.001
    57 QNNSLEWLE 0.001
    2 NVVVTEAVK 0.001
    64 LEITKDLQD 0.000
    35 KYNNSLTWF 0.000
    42 WFQNFVILD 0.000
    13 QKLVQTSLH 0.000
    30 KKVAWKYNN 0.000
    65 EITKDLQDE 0.000
    25 QRVPEKKVA 0.000
    11 EEQKLVQTS 0.000
    55 WHQNNSLEW 0.000
    17 QTSLHCGFQ 0.000
    6 TEAVKEEQK 0.000
    27 VPEKKVAWK 0.000
    45 NFVILDLYK 0.000
    33 AWKYNNSLT 0.000
    52 YKEWHQNNS 0.000
    5 VTEAVKEEQ 0.000
    28 PEKKVAWKY 0.000
    20 LHCGFQRVP 0.000
    23 GFQRVPEKK 0.000
    9 VKEEQKLVQ 0.000
    21 HCGFQRVPE 0.000
    29 EKKVAWKYN 0.000
    51 LYKEWHQNN 0.000
    54 EWHQNNSLE 0.000
  • TABLE XI
    58P1D12v.4-A0201-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    39 NSLTwFQNFV 35.109
    40 SLTWfQNFVI 24.809
    32 KVAWkYNNSL 6.542
    62 LEWLeITKDL 5.861
    41 LTWFqNFVIL 3.261
    19 TSLHcGFQRV 3.192
    33 VAWKyNNSLT 1.460
    48 VILDlYKEWH 0.712
    15 KLVQtSLHCG 0.600
    56 WHQNnSLEWL 0.423
    7 TEAVkEEQKL 0.415
    43 WFQNfVILDL 0.157
    1 HQNVvVTEAV 0.117
    58 QNNSlEWLEI 0.116
    66 EITKdLQDEL 0.108
    49 ILDLyKEWHQ 0.076
    8 EAVKeEQKLV 0.072
    25 FQRVpEKKVA 0.072
    27 RVPEkKVAWK 0.068
    24 GFQRvPEKKV 0.048
    47 FVILdLYKEW 0.045
    44 FQNFvILDLY 0.039
    12 EEQKlVQTSL 0.031
    14 QKLVqTSLHC 0.026
    59 NNSLeWLEIT 0.022
    37 YNNSlTWFQN 0.022
    35 WKYNnSLTWF 0.019
    51 DLYKeWHQNN 0.018
    16 LVQTsLHCGF 0.011
    10 VKEEqKLVQT 0.011
    53 YKEWhQNNSL 0.009
    54 KEWHqNNSLE 0.008
    4 VVVTeAVKEE 0.005
    20 SLHCgFQRVP 0.005
    5 VVTEaVKEEQ 0.004
    64 WLEltKDLQD 0.004
    61 SLEWlEITKD 0.002
    45 QNFViLDLYK 0.002
    17 VQTSIHCGFQ 0.002
    60 NSLEwLEITK 0.001
    3 NVVVtEAVKE 0.001
    18 QTSLhCGFQR 0.001
    65 LEITkDLQDE 0.001
    50 LDLYkEWHQN 0.001
    38 NNSLtWFQNF 0.001
    11 KEEQkLVQTS 0.001
    31 KKVAwKYNNS 0.001
    28 VPEKkVAWKY 0.000
    57 HQNNsLEWLE 0.000
    36 KYNNsLTWFQ 0.000
    23 CGFQrVPEKK 0.000
    9 AVKEeQKLVQ 0.000
    42 TWFQnFVILD 0.000
    2 QNVVvTEAVK 0.000
    46 NFVIlDLYKE 0.000
    13 EQKLvQTSLH 0.000
    21 LHCGfQRVPE 0.000
    6 VTEAvKEEQK 0.000
    29 PEKKvAWKYN 0.000
    26 QRVPeKKVAW 0.000
    22 HCGFqRVPEK 0.000
    52 LYKEwHQNNS 0.000
    63 EWLEiTKDLQ 0.000
    55 EWHQnNSLEW 0.000
    34 AWKYnNSLTW 0.000
    30 EKKVaWKYNN 0.000
  • TABLE XII
    58P1D12v.4-A3-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    60 SLEWLEITK 40.000
    2 NVVVTEAVK 3.000
    14 KLVQTSLHC 1.800
    39 SLTWFQNFV 0.600
    48 ILDLYKEWH 0.600
    19 SLHCGFQRV 0.600
    27 VPEKKVAWK 0.600
    40 LTWFQNFVI 0.450
    22 CGFQRVPEK 0.300
    38 NSLTWFQNF 0.135
    23 GFQRVPEKK 0.090
    18 TSLHCGFQR 0.090
    32 VAWKYNNSL 0.090
    44 QNFVILDLY 0.090
    43 FQNFVILDL 0.081
    45 NFVILDLYK 0.060
    6 TEAVKEEQK 0.060
    16 VQTSLHCGF 0.060
    66 ITKDLQDEL 0.030
    50 DLYKEWHQN 0.030
    26 RVPEKKVAW 0.030
    56 HQNNSLEWL 0.027
    53 KEWHQNNSL 0.027
    63 WLEITKDLQ 0.020
    8 AVKEEQKLV 0.015
    47 VILDLYKEW 0.015
    46 FVILDLYKE 0.013
    31 KVAWKYNNS 0.012
    35 KYNNSLTWF 0.009
    41 TWFQNFVIL 0.009
    12 EQKLVQTSL 0.008
    28 PEKKVAWKY 0.005
    59 NSLEWLEIT 0.005
    24 FQRVPEKKV 0.005
    4 VVTEAVKEE 0.005
    3 VVVTEAVKE 0.003
    15 LVQTSLHCG 0.003
    7 EAVKEEQKL 0.003
    58 NNSLEWLEI 0.002
    34 WKYNNSLTW 0.002
    5 VTEAVKEEQ 0.002
    10 KEEQKLVQT 0.001
    1 QNVVVTEAV 0.001
    65 EITKDLQDE 0.001
    30 KKVAWKYNN 0.001
    61 LEWLEITKD 0.000
    55 WHQNNSLEW 0.000
    42 WFQNFVILD 0.000
    62 EWLEITKDL 0.000
    13 QKLVQTSLH 0.000
    17 QTSLHCGFQ 0.000
    25 QRVPEKKVA 0.000
    21 HCGFQRVPE 0.000
    64 LEITKDLQD 0.000
    37 NNSLTWFQN 0.000
    36 YNNSLTWFQ 0.000
    57 QNNSLEWLE 0.000
    11 EEQKLVQTS 0.000
    33 AWKYNNSLT 0.000
    49 LDLYKEWHQ 0.000
    51 LYKEWHQNN 0.000
    9 VKEEQKLVQ 0.000
    52 YKEWHQNNS 0.000
    20 LHCGFQRVP 0.000
    54 EWHQNNSLE 0.000
    29 EKKVAWKYN 0.000
  • TABLE XIII
    58P1D12v.4-A3-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    27 RVPEkKVAWK 9.000
    40 SLTWfQNFVI 1.800
    6 VTEAvKEEQK 1.000
    18 QTSLhCGFQR 0.600
    22 HCGFqRVPEK 0.600
    32 KVAWkYNNSL 0.540
    41 LTWFqNFVIL 0.450
    60 NSLEwLEITK 0.450
    45 QNFViLDLYK 0.400
    44 FQNFvILDLY 0.270
    16 LVQTsLHCGF 0.200
    28 VPEKkVAWKY 0.180
    23 CGFQrVPEKK 0.150
    15 KLVQtSLHCG 0.135
    51 DLYKeWHQNN 0.090
    48 VILDlYKEWH 0.090
    2 QNVVvTEAVK 0.060
    49 ILDLyKEWHQ 0.060
    64 WLEItKDLQD 0.040
    38 NNSLtWFQNF 0.036
    61 SLEWlEITKD 0.030
    66 EITKdLQDEL 0.018
    47 FVILdLYKEW 0.015
    35 WKYNnSLTWF 0.015
    1 HQNVvVTEAV 0.009
    62 LEWLeITKDL 0.007
    9 AVKEeQKLVQ 0.006
    20 SLHCgFQRVP 0.006
    33 VAWKyNNSLT 0.005
    5 VVTEaVKEEQ 0.005
    19 TSLHcGFQRV 0.005
    4 VVVTeAVKEE 0.005
    39 NSLTwFQNFV 0.005
    3 NVVVtEAVKE 0.003
    25 FQRVpEKKVA 0.003
    43 WFQNfVILDL 0.003
    58 QNNSlEWLEI 0.002
    42 TWFQnFVILD 0.002
    57 HQNNsLEWLE 0.002
    7 TEAVkEEQKL 0.002
    13 EQKLvQTSLH 0.002
    11 KEEQkLVQTS 0.001
    56 WHQNnSLEWL 0.001
    59 NNSLeWLEIT 0.001
    54 KEWHqNNSLE 0.001
    12 EEQKlVQTSL 0.001
    53 YKEWhQNNSL 0.001
    14 QKLVqTSLHC 0.001
    26 QRVPeKKVAW 0.000
    24 GFQRvPEKKV 0.000
    8 EAVKeEQKLV 0.000
    34 AWKYnNSLTW 0.000
    10 VKEEqKLVQT 0.000
    31 KKVAwKYNNS 0.000
    36 KYNNsLTWFQ 0.000
    17 VQTSlHCGFQ 0.000
    46 NFVIlDLYKE 0.000
    65 LEITkDLQDE 0.000
    37 YNNSlTWFQN 0.000
    55 EWHQnNSLEW 0.000
    52 LYKEwHQNNS 0.000
    30 EKKVaWKYNN 0.000
    50 LDLYkEWHQN 0.000
    21 LHCGfQRVPE 0.000
    63 EWLEiTKDLQ 0.000
    29 PEKKvAWKYN 0.000
  • TABLE XIV
    58P1D12v.4-A1101-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    2 NVVVTEAVK 3.000
    60 SLEWLEITK 0.800
    45 NFVILDLYK 0.600
    23 GFQRVPEKK 0.600
    27 VPEKKVAWK 0.200
    40 LTWFQNFVI 0.060
    26 RVPEKKVAW 0.060
    6 TEAVKEEQK 0.060
    22 CGFQRVPEK 0.040
    18 TSLHCGFQR 0.018
    43 FQNFVILDL 0.012
    35 KYNNSLTWF 0.012
    8 AVKEEQKLV 0.010
    66 ITKDLQDEL 0.010
    46 FVILDLYKE 0.006
    16 VQTSLHCGF 0.006
    31 KVAWKYNNS 0.006
    56 HQNNSLEWL 0.006
    39 SLTWFQNFV 0.004
    32 VAWKYNNSL 0.004
    48 ILDLYKEWH 0.004
    19 SLHCGFQRV 0.004
    14 KLVQTSLHC 0.004
    53 KEWHQNNSL 0.004
    3 VVVTEAVKE 0.003
    47 VILDLYKEW 0.003
    24 FQRVPEKKV 0.003
    15 LVQTSLHCG 0.002
    12 EQKLVQTSL 0.002
    4 VVTEAVKEE 0.001
    5 VTEAVKEEQ 0.001
    17 QTSLHCGFQ 0.001
    7 EAVKEEQKL 0.001
    44 QNFVILDLY 0.001
    58 NNSLEWLEI 0.001
    34 WKYNNSLTW 0.001
    1 QNVVVTEAV 0.001
    63 WLEITKDLQ 0.000
    42 WFQNFVILD 0.000
    51 LYKEWHQNN 0.000
    55 WHQNNSLEW 0.000
    41 TWFQNFVIL 0.000
    13 QKLVQTSLH 0.000
    38 NSLTWFQNF 0.000
    50 DLYKEWHQN 0.000
    21 HCGFQRVPE 0.000
    30 KKVAWKYNN 0.000
    10 KEEQKLVQT 0.000
    64 LEITKDLQD 0.000
    25 QRVPEKKVA 0.000
    37 NNSLTWFQN 0.000
    61 LEWLEITKD 0.000
    28 PEKKVAWKY 0.000
    65 EITKDLQDE 0.000
    57 QNNSLEWLE 0.000
    36 YNNSLTWFQ 0.000
    49 LDLYKEWHQ 0.000
    62 EWLEITKDL 0.000
    9 VKEEQKLVQ 0.000
    59 NSLEWLEIT 0.000
    33 AWKYNNSLT 0.000
    52 YKEWHQNNS 0.000
    11 EEQKLVQTS 0.000
    54 EWHQNNSLE 0.000
    29 EKKVAWKYN 0.000
    20 LHCGFQRVP 0.000
  • TABLE XV
    58P1D12v.4-A1101-10-mers
    Each peptide is a portion of SEQID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    27 RVPEkKVAWK 6.000
    6 VTEAvKEEQK 1.000
    18 QTSLhCGFQR 0.600
    22 HCGFqRVPEK 0.200
    45 QNFViLDLYK 0.160
    60 NSLEwLEITK 0.060
    2 QNVVvTEAVK 0.060
    32 KVAWkYNNSL 0.060
    23 CGFQrVPEKK 0.040
    41 LTWFqNFVIL 0.020
    16 LVQTsLHCGF 0.020
    47 FVILdLYKEW 0.015
    40 SLTWfQNFVI 0.012
    48 VILDlYKEWH 0.006
    1 HQNVvVTEAV 0.006
    44 FQNFvILDLY 0.006
    28 VPEKkVAWKY 0.004
    43 WFQNfVILDL 0.004
    9 AVKEeQKLVQ 0.004
    24 GFQRvPEKKV 0.003
    3 NVVVtEAVKE 0.003
    25 FQRVpEKKVA 0.003
    36 KYNNsLTWFQ 0.002
    5 VVTEaVKEEQ 0.002
    15 KLVQtSLHCG 0.002
    13 EQKLvQTSLH 0.002
    4 VVVTeAVKEE 0.002
    57 HQNNsLEWLE 0.001
    66 EITKdLQDEL 0.001
    49 ILDLyKEWHQ 0.001
    58 QNNSlEWLEI 0.001
    64 WLEItKDLQD 0.001
    46 NFVIlDLYKE 0.001
    17 VQTSlHCGFQ 0.001
    62 LEWLeITKDL 0.001
    7 TEAVkEEQKL 0.001
    8 EAVKeEQKLV 0.000
    52 LYKEwHQNNS 0.000
    35 WKYNnSLTWF 0.000
    61 SLEWlEITKD 0.000
    38 NNSLtWFQNF 0.000
    34 AWKYnNSLTW 0.000
    33 VAWKyNNSLT 0.000
    54 KEWHqNNSLE 0.000
    19 TSLHcGFQRV 0.000
    26 QRVPeKKVAW 0.000
    39 NSLTwFQNFV 0.000
    51 DLYKeWHQNN 0.000
    56 WHQNnSLEWL 0.000
    53 YKEWhQNNSL 0.000
    12 EEQKlVQTSL 0.000
    11 KEEQkLVQTS 0.000
    37 YNNSlTWFQN 0.000
    55 EWHQnNSLEW 0.000
    31 KKVAwKYNNS 0.000
    65 LEITkDLQDE 0.000
    42 TWFQnFVILD 0.000
    14 QKLVqTSLHC 0.000
    59 NNSLeWLEIT 0.000
    20 SLHCgFQRVP 0.000
    50 LDLYkEWHQN 0.000
    10 VKEEqKLVQT 0.000
    21 LHCGfQRVPE 0.000
    30 EKKVaWKYNN 0.000
    63 EWLEiTKDLQ 0.000
    29 PEKKvAWKYN 0.000
  • TABLE XVI
    58P1D12v.4-A24-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    35 KYNNSLTWF 300.000
    62 EWLEITKDL 10.080
    43 FQNFVILDL 8.400
    7 EAVKEEQKL 7.920
    51 LYKEWHQNN 7.200
    66 ITKDLQDEL 6.336
    56 HQNNSLEWL 6.000
    12 EQKLVQTSL 5.600
    32 VAWKYNNSL 4.800
    41 TWFQNFVIL 4.800
    38 NSLTWFQNF 4.320
    16 VQTSLHCGF 2.400
    58 NNSLEWLEI 1.100
    40 LTWFQNFVI 1.000
    53 KEWHQNNSL 0.960
    26 RVPEKKVAW 0.360
    14 KLVQTSLHC 0.300
    31 KVAWKYNNS 0.240
    47 VILDLYKEW 0.238
    1 QNVVVTEAV 0.210
    59 NSLEWLEIT 0.180
    19 SLHCGFQRV 0.144
    44 QNFVILDLY 0.140
    39 SLTWFQNFV 0.120
    8 AVKEEQKLV 0.120
    23 GFQRVPEKK 0.116
    24 FQRVPEKKV 0.110
    50 DLYKEWHQN 0.100
    37 NNSLTWFQN 0.100
    33 AWKYNNSLT 0.100
    45 NFVILDLYK 0.090
    42 WFQNFVILD 0.075
    10 KEEQKLVQT 0.030
    30 KKVAWKYNN 0.030
    5 VTEAVKEEQ 0.023
    11 EEQKLVQTS 0.022
    25 QRVPEKKVA 0.018
    52 YKEWHQNNS 0.018
    60 SLEWLEITK 0.018
    36 YNNSLTWFQ 0.018
    57 QNNSLEWLE 0.018
    46 FVILDLYKE 0.017
    55 WHQNNSLEW 0.017
    3 VVVTEAVKE 0.017
    15 LVQTSLHCG 0.015
    63 WLEITKDLQ 0.015
    27 VPEKKVAWK 0.015
    2 NVVVTEAVK 0.015
    18 TSLHCGFQR 0.015
    4 VVTEAVKEE 0.013
    65 EITKDLQDE 0.012
    22 CGFQRVPEK 0.011
    48 ILDLYKEWH 0.010
    34 WKYNNSLTW 0.010
    21 HCGFQRVPE 0.010
    29 EKKVAWKYN 0.010
    17 QTSLHCGFQ 0.010
    54 EWHQNNSLE 0.010
    9 VKEEQKLVQ 0.002
    28 PEKKVAWKY 0.002
    13 QKLVQTSLH 0.002
    49 LDLYKEWHQ 0.002
    64 LEITKDLQD 0.002
    61 LEWLEITKD 0.001
    20 LHCGFQRVP 0.001
    6 TEAVKEEQK 0.001
  • TABLE XVII
    58P1D12v.4-A24-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    43 WFQNfVILDL 42.000
    32 KVAWkYNNSL 9.600
    52 LYKEwHQNNS 7.200
    66 EITKdLQDEL 5.280
    41 LTWFqNFVIL 4.800
    16 LVQTsLHCGF 3.600
    38 NNSLtWFQNF 2.880
    36 KYNNsLTWFQ 1.800
    58 QNNSlEWLEI 1.650
    40 SLTWfQNFVI 1.000
    12 EEQKlVQTSL 0.840
    24 GFQRvPEKKV 0.825
    53 YKEWhQNNSL 0.720
    56 WHQNnSLEWL 0.600
    62 LEWLeITKDL 0.560
    7 TEAVkEEQKL 0.528
    28 VPEKkVAWKY 0.231
    19 TSLHcGFQRV 0.216
    44 FQNFvILDLY 0.210
    1 HQNVvVTEAV 0.210
    35 WKYNnSLTWF 0.200
    47 FVILdLYKEW 0.198
    39 NSLTwFQNFV 0.180
    8 EAVKeEQKLV 0.150
    37 YNNSlTWFQN 0.150
    51 DLYKeWHQNN 0.120
    55 EWHQnNSLEW 0.110
    34 AWKYnNSLTW 0.100
    25 FQRVpEKKVA 0.100
    33 VAWKyNNSLT 0.100
    59 NNSLeWLEIT 0.100
    46 NFVIlDLYKE 0.083
    11 KEEQkLVQTS 0.043
    31 KKVAwKYNNS 0.036
    27 RVPEkKVAWK 0.036
    15 KLVQtSLHCG 0.030
    60 NSLEwLEITK 0.022
    5 VVTEaVKEEQ 0.018
    26 QRVPeKKVAW 0.018
    10 VKEEqKLVQT 0.018
    48 VILDlYKEWH 0.018
    63 EWLEiTKDLQ 0.018
    57 HQNNsLEWLE 0.018
    4 VVVTeAVKEE 0.017
    3 NVVVtEAVKE 0.017
    61 SLEWlEITKD 0.017
    23 CGFQrVPEKK 0.015
    50 LDLYkEWHQN 0.015
    2 QNVVvTEAVK 0.015
    14 QKLVqTSLHC 0.015
    6 VTEAvKEEQK 0.015
    64 WLEItKDLQD 0.015
    9 AVKEeQKLVQ 0.012
    45 QNFViLDLYK 0.012
    22 HCGFqRVPEK 0.011
    17 VQTSlHCGFQ 0.010
    18 QTSLhCGFQR 0.010
    30 EKKVaWKYNN 0.010
    49 ILDLyKEWHQ 0.010
    20 SLHCgFQRVP 0.010
    42 TWFQnFVILD 0.010
    13 EQKLvQTSLH 0.010
    54 KEWHqNNSLE 0.002
    65 LEITkDLQDE 0.002
    21 LHCGfQRVPE 0.001
    29 PEKKvAWKYN 0.001
  • TABLE XVIII
    58P1D12v.4-B7-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    32 VAWKYNNSL 12.00
    7 EAVKEEQKL 12.00
    43 FQNFVILDL 4.000
    66 ITKDLQDEL 4.000
    56 HQNNSLEWL 4.000
    12 EQKLVQTSL 4.000
    24 FQRVPEKKV 3.000
    8 AVKEEQKLV 3.000
    58 NNSLEWLEI 0.400
    41 TWFQNFVIL 0.400
    53 KEWHQNNSL 0.400
    62 EWLEITKDL 0.400
    40 LTWFQNFVI 0.400
    1 QNVVVTEAV 0.200
    39 SLTWFQNFV 0.200
    19 SLHCGFQRV 0.200
    31 KVAWKYNNS 0.100
    26 RVPEKKVAW 0.100
    59 NSLEWLEIT 0.100
    14 KLVQTSLHC 0.100
    27 VPEKKVAWK 0.060
    3 VVVTEAVKE 0.050
    15 LVQTSLHCG 0.050
    4 VVTEAVKEE 0.050
    2 NVVVTEAVK 0.050
    46 FVILDLYKE 0.050
    33 AWKYNNSLT 0.030
    47 VILDLYKEW 0.020
    50 DLYKEWHQN 0.020
    38 NSLTWFQNF 0.020
    37 NNSLTWFQN 0.020
    16 VQTSLHCGF 0.020
    44 QNFVILDLY 0.020
    21 HCGFQRVPE 0.015
    18 TSLHCGFQR 0.010
    22 CGFQRVPEK 0.010
    25 QRVPEKKVA 0.010
    65 EITKDLQDE 0.010
    57 QNNSLEWLE 0.010
    17 QTSLHCGFQ 0.010
    36 YNNSLTWFQ 0.010
    48 ILDLYKEWH 0.003
    10 KEEQKLVQT 0.003
    63 WLEITKDLQ 0.003
    5 VTEAVKEEQ 0.003
    60 SLEWLEITK 0.003
    35 KYNNSLTWF 0.002
    30 KKVAWKYNN 0.002
    34 WKYNNSLTW 0.002
    11 EEQKLVQTS 0.002
    55 WHQNNSLEW 0.002
    29 EKKVAWKYN 0.002
    51 LYKEWHQNN 0.002
    45 NFVILDLYK 0.001
    54 EWHQNNSLE 0.001
    61 LEWLEITKD 0.001
    42 WFQNFVILD 0.001
    20 LHCGFQRVP 0.001
    23 GFQRVPEKK 0.001
    64 LEITKDLQD 0.001
    6 TEAVKEEQK 0.001
    49 LDLYKEWHQ 0.001
    13 QKLVQTSLH 0.001
    52 YKEWHQNNS 0.001
    9 VKEEQKLVQ 0.000
    28 PEKKVAWKY 0.000
  • TABLE XIX
    58P1D12v.4-B7-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    32 KVAWkYNNSL 20.000
    41 LTWFqNFVIL 4.000
    66 EITKdLQDEL 4.000
    25 FQRVpEKKVA 1.000
    8 EAVKeEQKLV 0.600
    7 TEAVkEEQKL 0.400
    43 WFQNfVILDL 0.400
    62 LEWLeITKDL 0.400
    12 EEQKlVQTSL 0.400
    40 SLTWfQNFVI 0.400
    56 WHQNnSLEWL 0.400
    58 QNNSlEWLEI 0.400
    33 VAWKyNNSLT 0.300
    1 HQNVvVTEAV 0.200
    39 NSLTwFQNFV 0.200
    19 TSLHcGFQRV 0.200
    9 AVKEeQKLVQ 0.150
    28 VPEKkVAWKY 0.120
    53 YKEWhQNNSL 0.120
    47 FVILdLYKEW 0.100
    16 LVQTsLHCGF 0.100
    59 NNSLeWLEIT 0.100
    4 VVVTeAVKEE 0.050
    5 VVTEaVKEEQ 0.050
    3 NVVVtEAVKE 0.050
    27 RVPEkKVAWK 0.050
    24 GFQRvPEKKV 0.030
    51 DLYKeWHQNN 0.020
    44 FQNFvILDLY 0.020
    37 YNNSlTWFQN 0.020
    38 NNSLtWFQNF 0.020
    60 NSLEwLEITK 0.010
    13 EQKLvQTSLH 0.010
    20 SLHCgFQRVP 0.010
    18 QTSLhCGFQR 0.010
    23 CGFQrVPEKK 0.010
    22 HCGFqRVPEK 0.010
    17 VQTSlHCGFQ 0.010
    57 HQNNsLEWLE 0.010
    15 KLVQtSLHCG 0.010
    14 QKLVqTSLHC 0.010
    45 QNFViLDLYK 0.010
    48 VILDlYKEWH 0.010
    2 QNVVvTEAVK 0.010
    34 AWKYnNSLTW 0.006
    64 WLEItKDLQD 0.003
    10 VKEEqKLVQT 0.003
    49 ILDLyKEWHQ 0.003
    6 VTEAvKEEQK 0.003
    61 SLEWlEITKD 0.003
    35 WKYNnSLTWF 0.002
    26 QRVPeKKVAW 0.002
    55 EWHQnNSLEW 0.002
    30 EKKVaWKYNN 0.002
    52 LYKEwHQNNS 0.002
    31 KKVAwKYNNS 0.002
    50 LDLYkEWHQN 0.002
    21 LHCGfQRVPE 0.002
    36 KYNNsLTWFQ 0.001
    63 EWLEiTKDLQ 0.001
    65 LEITkDLQDE 0.001
    54 KEWHqNNSLE 0.001
    42 TWFQnFVILD 0.001
    46 NFVIlDLYKE 0.001
    11 KEEQkLVQTS 0.001
    29 PEKKvAWKYN 0.000
  • TABLE XX
    58P1D12v.4-B3501-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    Pos Subsequence Score
    66 ITKDLQDEL 6.000
    38 NSLTWFQNF 5.000
    7 EAVKEEQKL 4.500
    32 VAWKYNNSL 3.000
    12 EQKLVQTSL 3.000
    26 RVPEKKVAW 2.000
    44 QNFVILDLY 2.000
    8 AVKEEQKLV 1.800
    47 VILDLYKEW 1.000
    16 VQTSLHCGF 1.000
    59 NSLEWLEIT 1.000
    43 FQNFVILDL 1.000
    56 HQNNSLEWL 1.000
    58 NNSLEWLEI 0.600
    24 FQRVPEKKV 0.600
    40 LTWFQNFVI 0.400
    62 EWLEITKDL 0.200
    31 KVAWKYNNS 0.200
    39 SLTWFQNFV 0.200
    53 KEWHQNNSL 0.200
    14 KLVQTSLHC 0.200
    19 SLHCGFQRV 0.200
    35 KYNNSLTWF 0.200
    1 QNVVVTEAV 0.200
    50 DLYKEWHQN 0.150
    37 NNSLTWFQN 0.100
    41 TWFQNFVIL 0.100
    51 LYKEWHQNN 0.060
    27 VPEKKVAWK 0.060
    28 PEKKVAWKY 0.060
    34 WKYNNSLTW 0.050
    55 WHQNNSLEW 0.050
    18 TSLHCGFQR 0.050
    29 EKKVAWKYN 0.030
    33 AWKYNNSLT 0.030
    4 VVTEAVKEE 0.020
    30 KKVAWKYNN 0.020
    65 EITKDLQDE 0.015
    25 QRVPEKKVA 0.015
    46 FVILDLYKE 0.015
    3 VVVTEAVKE 0.015
    57 QNNSLEWLE 0.010
    36 YNNSLTWFQ 0.010
    22 CGFQRVPEK 0.010
    17 QTSLHCGFQ 0.010
    11 EEQKLVQTS 0.010
    15 LVQTSLHCG 0.010
    2 NVVVTEAVK 0.010
    21 HCGFQRVPE 0.010
    10 KEEQKLVQT 0.006
    52 YKEWHQNNS 0.003
    63 WLEITKDLQ 0.003
    48 ILDLYKEWH 0.003
    5 VTEAVKEEQ 0.003
    60 SLEWLEITK 0.003
    61 LEWLEITKD 0.002
    54 EWHQNNSLE 0.001
    45 NFVILDLYK 0.001
    23 GFQRVPEKK 0.001
    20 LHCGFQRVP 0.001
    6 TEAVKEEQK 0.001
    42 WFQNFVILD 0.001
    13 QKLVQTSLH 0.001
    49 LDLYKEWHQ 0.001
    64 LEITKDLQD 0.001
    9 VKEEQKLVQ 0.001
  • TABLE XXI
    58P1D12v.4-B3501-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    Pos Subsequence Score
    28 VPEKkVAWKY 12.000
    44 FQNFvILDLY 2.000
    32 KVAWkYNNSL 2.000
    39 NSLTwFQNFV 1.000
    41 LTWFqNFVIL 1.000
    16 LVQTsLHCGF 1.000
    38 NNSLtWFQNF 1.000
    19 TSLHcGFQRV 1.000
    66 EITKdLQDEL 1.000
    8 EAVKeEQKLV 0.900
    58 QNNSlEWLEI 0.600
    47 FVILdLYKEW 0.500
    25 FQRVpEKKVA 0.450
    40 SLTWfQNFVI 0.400
    33 VAWKyNNSLT 0.300
    1 HQNVvVTEAV 0.200
    7 TEAVkEEQKL 0.150
    34 AWKYnNSLTW 0.150
    51 DLYKeWHQNN 0.100
    62 LEWLeITKDL 0.100
    43 WFQNfVILDL 0.100
    56 WHQNnSLEWL 0.100
    59 NNSLeWLEIT 0.100
    60 NSLEwLEITK 0.100
    37 YNNSlTWFQN 0.100
    35 WKYNnSLTWF 0.100
    12 EEQKlVQTSL 0.100
    9 AVKEeQKLVQ 0.060
    52 LYKEwHQNNS 0.060
    55 EWHQnNSLEW 0.050
    26 QRVPeKKVAW 0.050
    27 RVPEkKVAWK 0.040
    30 EKKVaWKYNN 0.030
    13 EQKLvQTSLH 0.030
    53 YKEWhQNNSL 0.030
    48 VILDlYKEWH 0.020
    24 GFQRvPEKKV 0.020
    31 KKVAwKYNNS 0.020
    15 KLVQtSLHCG 0.020
    5 VVTEaVKEEQ 0.020
    3 NVVVtEAVKE 0.015
    50 LDLYkEWHQN 0.015
    57 HQNNsLEWLE 0.010
    23 CGFQrVPEKK 0.010
    22 HCGFqRVPEK 0.010
    20 SLHCgFQRVP 0.010
    18 QTSLhCGFQR 0.010
    45 QNFViLDLYK 0.010
    2 QNVVvTEAVK 0.010
    14 QKLVqTSLHC 0.010
    17 VQTSlHCGFQ 0.010
    4 VVVTeAVKEE 0.010
    11 KEEQkLVQTS 0.006
    10 VKEEqKLVQT 0.006
    61 SLEWlEITKD 0.004
    29 PEKKvAWKYN 0.003
    49 ILDLyKEWHQ 0.003
    6 VTEAvKEEQK 0.003
    64 WLEItKDLQD 0.003
    54 KEWHqNNSLE 0.002
    63 EWLEiTKDLQ 0.002
    36 KYNNsLTWFQ 0.002
    65 LEITkDLQDE 0.002
    46 NFVIlDLYKE 0.002
    42 TWFQnFVILD 0.001
    21 LHCGfQRVPE 0.001
  • TABLE XXII
    58P1D12v.1-A1-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    121 WSDGSNSQY 31
    140 GSEKCVVMY 31
    186 PTAPVEKPY 27
    32 FADFKHPCY 26
    62 ESEGGVLLS 26
    211 AGIIPNLIY 24
    208 VTEAGIIPN 23
    111 TSGACPDLY 21
    155 PGLGGPYLY 20
    125 SNSQYRNWY 19
    133 YTDEPSCGS 19
    94 ISDGDFWIG 18
    153 ANPGLGGPY 18
    172 MKHNYICKY 18
    223 PTIPLLLLI 18
    189 PVEKPYLTN 17
    36 KHPCYKMAY 16
    168 DRCNMKHNY 15
  • TABLE XXIII
    58P1D12v.1-A0201-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    6 SLLLGAALL 28
    212 GIIPNLIYV 27
    213 IIPNLIYVV 26
    217 LIYVVIPTI 26
    220 VVIPTIPLL 24
    224 TIPLLLLIL 24
    79 LIESMLQNL 23
    176 YICKYEPEI 22
    63 SEGGVLLSL 21
    67 VLLSLENEA 21
    227 LLLLILVAF 21
    228 LLLILVAFG 21
    93 GISDGDFWI 20
    221 VIPTIPLLL 20
    59 LACESEGGV 19
    86 NLTKPGTGI 19
    209 TEAGIIPNL 19
    216 NLIYVVIPT 19
    219 YVVIPTIPL 19
    223 PTIPLLLLI 19
    225 IPLLLLILV 19
    226 PLLLLILVA 19
    229 LLILVAFGT 19
    251 KTSPNQSTL 19
    82 SMLQNLTKP 18
    5 VSLLLGAAL 17
    7 LLLGAALLC 17
    8 LLGAALLCG 17
    12 ALLCGHGAF 17
    40 YKMAYFHEL 17
    71 LENEAEQKL 17
    110 QTSGACPDL 17
    222 IPTIPLLLL 17
    13 LLCGHGAFC 16
    61 CESEGGVLL 16
    78 KLIESMLQN 16
    3 RVVSLLLGA 15
    4 VVSLLLGAA 15
    44 YFHELSSRV 15
    51 RVSFQEARL 15
    95 SDGDFWIGL 15
    156 GLGGPYLYQ 15
    187 TAPVEKPYL 15
    194 YLTNQPGDT 15
    205 NVVVTEAGI 15
    11 AALLCGHGA 14
    152 TANPGLGGP 14
    182 PEINPTAPV 14
    230 LILVAFGTC 14
    242 MLHKSKGRT 14
    253 SPNQSTLWI 14
  • TABLE XXIV
    58P1D12v.1-A0203-9-mers
    No Results Found.
  • TABLE XXV
    58P1D12v.1-A3-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    70 SLENEAEQK 28
    12 ALLCGHGAF 23
    78 KLIESMLQN 23
    6 SLLLGAALL 22
    227 LLLLILVAF 22
    258 TLWISKSTR 22
  • TABLE XXIV
    58P1D12v.1-A0203-9-mers
    No Results Found.
  • TABLE XXV
    58P1D12v.1-A3-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    7 LLLGAALLC 20
    23 RVVSGQKVC 20
    24 VVSGQKVCF 20
    226 PLLLLILVA 20
    243 LHKSKGRTK 19
    13 LLCGHGAFC 18
    21 CRRVVSGQK 18
    81 ESMLQNLTK 18
    161 YLYQWNDDR 18
    183 EINPTAPVE 18
    229 LLILVAFGT 18
    231 ILVAFGTCC 18
    3 RVVSLLLGA 17
    29 KVCFADFKH 17
    51 RVSFQEARL 17
    184 INPTAPVEK 17
    189 PVEKPYLTN 17
    217 LIYVVIPTI 17
    232 LVAFGTCCF 17
    102 GLWRNGDGQ 16
    135 DEPSCGSEK 16
    211 AGIIPNLIY 16
    220 VVIPTIPLL 16
    228 LLLILVAFG 16
    8 LLGAALLCG 15
    28 QKVCFADFK 15
    47 ELSSRVSFQ 15
    58 RLACESEGG 15
    153 ANPGLGGPY 15
  • TABLE XXIV
    58P1D12v.1-A0203-9-mers
    No Results Found.
  • TABLE XXV
    58P1D12v.1-A3-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    156 GLGGPYLYQ 15
    213 IIPNLIYVV 15
    230 LILVAFGTC 15
    259 LWISKSTRK 15
    19 AFCRRVVSG 14
    33 ADFKHPCYK 14
    86 NLTKPGTGI 14
    117 DLYQWSDGS 14
    121 WSDGSNSQY 14
    145 VVMYHQPTA 14
    165 WNDDRCNMK 14
    206 VVVTEAGII 14
    207 VVTEAGIIP 14
    212 GIIPNLIYV 14
    216 NLIYVVIPT 14
  • TABLE XXVI
    58P1D12v.1-A26-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    220 VVIPTIPLL 27
    219 YVVIPTIPL 23
    76 EQKLIESML 22
    3 RVVSLLLGA 21
    201 DTHQNVVVT 21
    251 KTSPNQSTL 20
    186 PTAPVEKPY 19
    223 PTIPLLLLI 19
    224 TIPLLLLIL 19
    24 VVSGQKVCF 18
    51 RVSFQEARL 18
    168 DRCNMKHNY 18
    232 LVAFGTCCF 18
    4 VVSLLLGAA 17
    79 LIESMLQNL 17
    91 GTGISDGDF 17
    47 ELSSRVSFQ 16
    64 EGGVLLSLE 16
    74 EAEQKLIES 16
    110 QTSGACPDL 16
    23 RVVSGQKVC 15
    62 ESEGGVLLS 15
    63 SEGGVLLSL 15
    140 GSEKCVVMY 15
    172 MKHNYICKY 15
    183 EINPTAPVE 15
    206 VVVTEAGII 15
    207 VVTEAGIIP 15
    209 TEAGIIPNL 15
    211 AGIIPNLIY 15
    212 GIIPNLIYV 15
    221 VIPTIPLLL 15
    227 LLLLILVAF 15
    66 GVLLSLENE 14
    144 CVVMYHQPT 14
    205 NVVVTEAGI 14
    222 IPTIPLLLL 14
  • TABLE XXVII
    58P1D12v.1-B0702-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    222 IPTIPLLLL 26
    154 NPGLGGPYL 23
    214 IPNLIYVVI 20
    253 SPNQSTLWI 19
    188 APVEKPYLT 18
    37 HPCYKMAYF 17
    198 QPGDTHQNV 17
    225 IPLLLLILV 17
    61 CESEGGVLL 15
    63 SEGGVLLSL 15
    110 QTSGACPDL 15
    185 NPTAPVEKP 15
    219 YVVIPTIPL 15
    1 MSRVVSLLL 14
    51 RVSFQEARL 14
    60 ACESEGGVL 14
    209 TEAGIIPNL 14
    251 KTSPNQSTL 14
    5 VSLLLGAAL 13
    40 YKMAYFHEL 13
    136 EPSCGSEKC 13
    181 EPEINPTAP 13
  • TABLE XXVIII
    58P1D12v.1-B08-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    37 HPCYKMAYF 24
    76 EQKLIESML 23
    243 LHKSKGRTK 20
    6 SLLLGAALL 19
    245 KSKGRTKTS 19
    27 GQKVCFADF 18
    188 APVEKPYLT 18
    1 MSRVVSLLL 17
    47 ELSSRVSFQ 17
    74 EAEQKLIES 17
    154 NPGLGGPYL 17
    222 IPTIPLLLL 17
    169 RCNMKHNYI 16
    247 KGRTKTSPN 16
    32 FADFKHPCY 15
    187 TAPVEKPYL 15
    214 IPNLIYVVI 15
    221 VIPTIPLLL 15
    224 TIPLLLLIL 15
    227 LLLLILVAF 15
    18 GAFCRRVVS 14
    79 LIESMLQNL 14
    253 SPNQSTLWI 14
    5 VSLLLGAAL 13
    12 ALLCGHGAF 13
    56 EARLACESE 13
    61 CESEGGVLL 13
    139 CGSEKCVVM 13
    141 SEKCVVMYH 13
    261 ISKSTRKES 13
    265 TRKESGMEV 13
    25 VSGQKVCFA 12
    63 SEGGVLLSL 12
    71 LENEAEQKL 12
    86 NLTKPGTGI 12
    95 SDGDFWIGL 12
    149 HQPTANPGL 12
    177 ICKYEPEIN 12
    210 EAGIIPNLI 12
    217 LIYVVIPTI 12
    220 VVIPTIPLL 12
  • TABLE XXIX
    58P1D12v.1-B1510-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    16 GHGAFCRRV 15
    45 FHELSSRVS 15
    61 CESEGGVLL 15
    202 THQNVVVTE 15
    209 TEAGIIPNL 15
    251 KTSPNQSTL 15
    187 TAPVEKPYL 14
    220 VVIPTIPLL 14
    51 RVSFQEARL 13
    60 ACESEGGVL 13
    110 QTSGACPDL 13
    148 YHQPTANPG 13
    222 IPTIPLLLL 13
    243 LHKSKGRTK 13
    36 KHPCYKMAY 12
    40 YKMAYFHEL 12
    139 CGSEKCVVM 12
    154 NPGLGGPYL 12
    219 YVVIPTIPL 12
    1 MSRVVSLLL 11
    5 VSLLLGAAL 11
    6 SLLLGAALL 11
    24 VVSGQKVCF 11
    63 SEGGVLLSL 11
    71 LENEAEQKL 11
    76 EQKLIESML 11
    79 LIESMLQNL 11
    95 SDGDFWIGL 11
    149 HQPTANPGL 11
    221 VIPTIPLLL 11
    224 TIPLLLLIL 11
    173 KHNYICKYE 10
    235 FGTCCFQML 10
    46 HELSSRVSF 9
    227 LLLLILVAF 9
    12 ALLCGHGAF 8
    27 GQKVCFADF 8
    75 AEQKLIESM 8
    164 QWNDDRCNM 8
    232 LVAFGTCCF 8
  • TABLE XXX
    58P1D12v.1-B2705-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    21 CRRVVSGQK 23
    50 SRVSFQEAR 23
    168 DRCNMKHNY 21
    209 TEAGIIPNL 19
    248 GRTKTSPNQ 19
    22 RRVVSGQKV 18
    43 AYFHELSSR 18
    97 GDFWIGLWR 18
    259 LWISKSTRK 18
    6 SLLLGAALL 17
    51 RVSFQEARL 17
    220 VVIPTIPLL 17
    251 KTSPNQSTL 17
    33 ADFKHPCYK 16
    63 SEGGVLLSL 16
    75 AEQKLIESM 16
    227 LLLLILVAF 16
    5 VSLLLGAAL 15
    12 ALLCGHGAF 15
    46 HELSSRVSF 15
    57 ARLACESEG 15
    91 GTGISDGDF 15
    140 GSEKCVVMY 15
    155 PGLGGPYLY 15
    161 YLYQWNDDR 15
    2 SRVVSLLLG 14
    15 CGHGAFCRR 14
    24 VVSGQKVCF 14
    27 GQKVCFADF 14
    38 PCYKMAYFH 14
    61 CESEGGVLL 14
    81 ESMLQNLTK 14
    121 WSDGSNSQY 14
    154 NPGLGGPYL 14
    211 AGIIPNLIY 14
    217 LIYVVIPTI 14
    222 IPTIPLLLL 14
    224 TIPLLLLIL 14
    236 GTCCFQMLH 14
    241 QMLHKSKGR 14
    258 TLWISKSTR 14
    265 TRKESGMEV 14
    1 MSRVVSLLL 13
    14 LCGHGAFCR 13
    29 KVCFADFKH 13
    60 ACESEGGVL 13
    70 SLENEAEQK 13
    71 LENEAEQKL 13
    76 EQKLIESML 13
    79 LIESMLQNL 13
    104 WRNGDGQTS 13
    110 QTSGACPDL 13
    172 MKHNYICKY 13
    184 INPTAPVEK 13
    187 TAPVEKPYL 13
    219 YVVIPTIPL 13
    255 NQSTLWISK 13
    28 QKVCFADFK 12
    34 DFKHPCYKM 12
    36 KHPCYKMAY 12
    37 HPCYKMAYF 12
    93 GISDGDFWI 12
    135 DEPSCGSEK 12
    139 CGSEKCVVM 12
    149 HQPTANPGL 12
    153 ANPGLGGPY 12
    166 NDDRCNMKH 12
    169 RCNMKHNYI 12
    195 LTNQPGDTH 12
    221 VIPTIPLLL 12
    223 PTIPLLLLI 12
    232 LVAFGTCCF 12
    239 CFQMLHKSK 12
    243 LHKSKGRTK 12
    263 KSTRKESGM 12
  • TABLE XXXI
    58P1D12v.1-B2709-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    22 RRVVSGQKV 23
    265 TRKESGMEV 18
    248 GRTKTSPNQ 16
    51 RVSFQEARL 15
    6 SLLLGAALL 13
    57 ARLACESEG 13
    200 GDTHQNVVV 13
    209 TEAGIIPNL 13
    212 GIIPNLIYV 13
    220 VVIPTIPLL 13
    222 IPTIPLLLL 13
    251 KTSPNQSTL 13
    2 SRVVSLLLG 12
    5 VSLLLGAAL 12
    16 GHGAFCRRV 12
    46 HELSSRVSF 12
    50 SRVSFQEAR 12
    60 ACESEGGVL 12
    79 LIESMLQNL 12
    169 RCNMKHNYI 12
    221 VIPTIPLLL 12
    227 LLLLILVAF 12
    1 MSRVVSLLL 11
    12 ALLCGHGAF 11
    21 CRRVVSGQK 11
    27 GQKVCFADF 11
    61 CESEGGVLL 11
    63 SEGGVLLSL 11
    76 EQKLIESML 11
    91 GTGISDGDF 11
    93 GISDGDFWI 11
    104 WRNGDGQTS 11
    110 QTSGACPDL 11
    149 HQPTANPGL 11
    154 NPGLGGPYL 11
    168 DRCNMKHNY 11
    187 TAPVEKPYL 11
    214 IPNLIYVVI 11
    217 LIYVVIPTI 11
    219 YVVIPTIPL 11
    223 PTIPLLLLI 11
    224 TIPLLLLIL 11
    225 IPLLLLILV 11
    235 FGTCCFQML 11
    263 KSTRKESGM 11
  • TABLE XXXII
    58P1D12v.1-B4402-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    209 TEAGIIPNL 25
    63 SEGGVLLSL 24
    46 HELSSRVSF 23
    61 CESEGGVLL 23
    71 LENEAEQKL 21
    220 VVIPTIPLL 20
    211 AGIIPNLIY 17
    223 PTIPLLLLI 17
    227 LLLLILVAF 17
    251 KTSPNQSTL 17
    12 ALLCGHGAF 16
    75 AEQKLIESM 16
    221 VIPTIPLLL 16
    6 SLLLGAALL 15
    96 DGDFWIGLW 15
    113 GACPDLYQW 15
    153 ANPGLGGPY 15
    157 LGGPYLYQW 15
    172 MKHNYICKY 15
    182 PEINPTAPV 15
    186 PTAPVEKPY 15
    5 VSLLLGAAL 14
    36 KHPCYKMAY 14
    60 ACESEGGVL 14
    76 EQKLIESML 14
    125 SNSQYRNWY 14
    155 PGLGGPYLY 14
    190 VEKPYLTNQ 14
    210 EAGIIPNLI 14
    224 TIPLLLLIL 14
    24 VVSGQKVCF 13
    40 YKMAYFHEL 13
    72 ENEAEQKLI 13
    80 IESMLQNLT 13
    92 TGISDGDFW 13
    95 SDGDFWIGL 13
    121 WSDGSNSQY 13
    180 YEPEINPTA 13
    222 IPTIPLLLL 13
    252 TSPNQSTLW 13
  • TABLE XXXIII
    58P1D12v.1-B5101-9-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    214 IPNLIYVVI 26
    225 IPLLLLILV 24
    59 LACESEGGV 23
    210 EAGIIPNLI 22
    253 SPNQSTLWI 22
    217 LIYVVIPTI 21
    198 QPGDTHQNV 20
    222 IPTIPLLLL 20
    199 PGDTHQNVV 19
    17 HGAFCRRVV 18
    187 TAPVEKPYL 18
    154 NPGLGGPYL 17
    42 MAYFHELSS 16
    185 NPTAPVEKP 16
    192 KPYLTNQPG 16
    18 GAFCRRVVS 15
    159 GPYLYQWND 15
    213 IIPNLIYVV 15
    176 YICKYEPEI 14
    188 APVEKPYLT 14
    233 VAFGTCCFQ 14
    235 FGTCCFQML 14
    44 YFHELSSRV 13
    74 EAEQKLIES 13
    86 NLTKPGTGI 13
    139 CGSEKCVVM 13
    200 GDTHQNVVV 13
    206 VVVTEAGII 13
  • TABLE XXXIV
    58P1D12v.1-A1-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    110 QTSGACPDLY 24
    210 EAGIIPNLIY 22
    62 ESEGGVLLSL 21
    35 FKHPCYKMAY 20
    124 GSNSQYRNWY 20
    152 TANPGLGGPY 20
    94 ISDGDFWIGL 19
    133 YTDEPSCGSE 18
    96 DGDFWIGLWR 17
    139 CGSEKCVVMY 17
    154 NPGLGGPYLY 17
    171 NMKHNYICKY 17
    208 VTEAGIIPNL 17
    72 ENEAEQKLIE 16
    120 QWSDGSNSQY 16
    223 PTIPLLLLIL 16
    31 CFADFKHPCY 15
    70 SLENEAEQKL 15
    87 LTKPGTGISD 15
    121 WSDGSNSQYR 15
    134 TDEPSCGSEK 15
    140 GSEKCVVMYH 15
    167 DDRCNMKHNY 15
    179 KYEPEINPTA 15
    185 NPTAPVEKPY 15
    60 ACESEGGVLL 14
    165 WNDDRCNMKH 13
    220 VVIPTIPLLL 13
    1 MSRVVSLLLG 12
    45 FHELSSRVSF 12
    52 VSFQEARLAC 12
    79 LIESMLQNLT 12
    199 PGDTHQNVVV 12
    236 GTCCFQMLHK 12
    251 KTSPNQSTLW 12
  • TABLE XXXV
    58P1D12v.1-A0201-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    78 KLIESMLQNL 29
    212 GIIPNLIYVV 28
    216 NLIYVVIPTI 26
    221 VIPTIPLLLL 24
    58 RLACESEGGV 23
    70 SLENEAEQKL 23
    224 TIPLLLLILV 22
    7 LLLGAALLCG 21
    219 YVVIPTIPLL 21
    213 IIPNLIYVVI 20
    220 VVIPTIPLLL 20
    223 PTIPLLLLIL 20
    227 LLLLILVAFG 20
    208 VTEAGIIPNL 19
    228 LLLILVAFGT 19
    4 VVSLLLGAAL 18
    59 LACESEGGVL 18
    156 GLGGPYLYQW 18
    211 AGIIPNLIYV 18
    264 STRKESGMEV 18
    8 LLGAALLCGH 17
    12 ALLCGHGAFC 16
    68 LLSLENEAEQ 16
    83 MLQNLTKPGT 16
    102 GLWRNGDGQT 16
    148 YHQPTANPGL 16
    186 PTAPVEKPYL 16
    226 PLLLLILVAF 16
    229 LLILVAFGTC 16
    5 VSLLLGAALL 15
    6 SLLLGAALLC 15
    24 VVSGQKVCFA 15
    62 ESEGGVLLSL 15
    94 ISDGDFWIGL 15
    153 ANPGLGGPYL 15
  • TABLE XXXVI
    58P1D12v.1-A0203-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    3 RVVSLLLGAA 19
    4 VVSLLLGAAL 17
    2 SRVVSLLLGA 10
    10 GAALLCGHGA 10
    24 VVSGQKVCFA 10
    34 DFKHPCYKMA 10
    48 LSSRVSFQEA 10
    51 RVSFQEARLA 10
    66 GVLLSLENEA 10
    105 RNGDGQTSGA 10
    144 CVVMYHQPTA 10
    179 KYEPEINPTA 10
    202 THQNVVVTEA 10
    225 IPLLLLILVA 10
    11 AALLCGHGAF 9
    25 VSGQKVCFAD 9
    35 FKHPCYKMAY 9
    49 SSRVSFQEAR 9
    52 VSFQEARLAC 9
    67 VLLSLENEAE 9
    106 NGDGQTSGAC 9
    145 VVMYHQPTAN 9
    180 YEPEINPTAP 9
    203 HQNVVVTEAG 9
    226 PLLLLILVAF 9
  • TABLE XXXVII
    58P1D12v.1-A3-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    242 MLHKSKGRTK 27
    183 EINPTAPVEK 26
    23 RVVSGQKVCF 22
    194 YLTNQPGDTH 22
    226 PLLLLILVAF 22
    258 TLWISKSTRK 22
    12 ALLCGHGAFC 21
    6 SLLLGAALLC 20
    20 FCRRVVSGQK 20
    220 VVIPTIPLLL 20
    13 LLCGHGAFCR 19
    69 LSLENEAEQK 19
    80 IESMLQNLTK 19
    134 TDEPSCGSEK 19
    213 IIPNLIYVVI 19
    229 LLILVAFGTC 19
    4 VVSLLLGAAL 18
    212 GIIPNLIYVV 18
    231 ILVAFGTCCF 18
    7 LLLGAALLCG 17
    51 RVSFQEARLA 17
    78 KLIESMLQNL 17
    102 GLWRNGDGQT 17
    117 DLYQWSDGSN 17
    216 NLIYVVIPTI 17
    3 RVVSLLLGAA 16
    8 LLGAALLCGH 16
    58 RLACESEGGV 16
    227 LLLLILVAFG 16
    228 LLLILVAFGT 16
    120 QWSDGSNSQY 15
    156 GLGGPYLYQW 15
    27 GQKVCFADFK 14
    68 LLSLENEAEQ 14
    144 CVVMYHQPTA 14
    161 YLYQWNDDRC 14
    164 QWNDDRCNMK 14
    205 NVVVTEAGII 14
    217 LIYVVIPTIP 14
    230 LILVAFGTCC 14
    257 STLWISKSTR 14
  • TABLE XXXVIII
    58P1D12v.1-A26-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    219 YVVIPTIPLL 26
    223 PTIPLLLLIL 26
    62 ESEGGVLLSL 24
    220 VVIPTIPLLL 24
    23 RVVSGQKVCF 23
    3 RVVSLLLGAA 21
    201 DTHQNVVVTE 21
    210 EAGIIPNLIY 21
    4 VVSLLLGAAL 20
    208 VTEAGIIPNL 20
    78 KLIESMLQNL 18
    110 QTSGACPDLY 18
    167 DDRCNMKHNY 18
    207 VVTEAGIIPN 18
    139 CGSEKCVVMY 17
    186 PTAPVEKPYL 17
    221 VIPTIPLLLL 17
    206 VVVTEAGIIP 16
    74 EAEQKLIESM 15
    81 ESMLQNLTKP 15
    144 CVVMYHQPTA 15
    171 NMKHNYICKY 15
    183 EINPTAPVEK 15
    205 NVVVTEAGII 15
    24 VVSGQKVCFA 14
    50 SRVSFQEARL 14
    152 TANPGLGGPY 14
    189 PVEKPYLTNQ 14
    120 QWSDGSNSQY 13
    151 PTANPGLGGP 13
    212 GIIPNLIYVV 13
    218 IYVVIPTIPL 13
    226 PLLLLILVAF 13
    250 TKTSPNQSTL 13
  • TABLE XXXIX
    58P1D12v.1-B0702-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    181 EPEINPTAPV 20
    225 IPLLLLILVA 20
    136 EPSCGSEKCV 19
    222 IPTIPLLLLI 19
    198 QPGDTHQNVV 18
    62 ESEGGVLLSL 15
    153 ANPGLGGPYL 15
    4 VVSLLLGAAL 14
    60 ACESEGGVLL 14
    94 ISDGDFWIGL 14
    188 APVEKPYLTN 14
    214 IPNLIYVVIP 14
    234 AFGTCCFQML 14
    75 AEQKLIESML 13
    150 QPTANPGLGG 13
    186 PTAPVEKPYL 13
    218 IYVVIPTIPL 13
    220 VVIPTIPLLL 13
    223 PTIPLLLLIL 13
    24 VVSGQKVCFA 12
    89 KPGTGISDGD 12
    148 YHQPTANPGL 12
    185 NPTAPVEKPY 12
    192 KPYLTNQPGD 12
    208 VTEAGIIPNL 12
    221 VIPTIPLLLL 12
    5 VSLLLGAALL 11
    37 HPCYKMAYFH 11
    39 CYKMAYFHEL 11
    50 SRVSFQEARL 11
    59 LACESEGGVL 11
    78 KLIESMLQNL 11
    109 GQTSGACPDL 11
    115 CPDLYQWSDG 11
    154 NPGLGGPYLY 11
    219 YVVIPTIPLL 11
    16 GHGAFCRRVV 10
    70 SLENEAEQKL 10
    138 SCGSEKCVVM 10
    159 GPYLYQWNDD 10
    199 PGDTHQNVVV 10
    200 GDTHQNVVVT 10
    213 IIPNLIYVVI 10
    250 TKTSPNQSTL 10
    253 SPNQSTLWIS 10
  • TABLE XL
    58P1D12v.1-B08-10-mers
    No Results Found.
  • TABLE XLI
    58P1D12v.1-B1510-10-mers
    No Results Found.
  • TABLE XLII
    58P1D12v.1-B2705-10-mers
    No Results Found.
  • TABLE XLIII
    58P1D12v.1-B2709-10-mers
    No Results Found.
  • TABLE XLIV
    58P1D12v.1-B4402-10-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    75 AEQKLIESML 23
    209 TEAGIIPNLI 22
    71 LENEAEQKLI 21
    220 VVIPTIPLLL 19
    223 PTIPLLLLIL 18
    11 AALLCGHGAF 17
    95 SDGDFWIGLW 16
    182 PEINPTAPVE 16
    190 VEKPYLTNQP 16
    216 NLIYVVIPTI 16
    226 PLLLLILVAF 16
    251 KTSPNQSTLW 16
    35 FKHPCYKMAY 15
    60 ACESEGGVLL 15
    62 ESEGGVLLSL 15
    78 KLIESMLQNL 15
    153 ANPGLGGPYL 15
    171 NMKHNYICKY 15
    221 VIPTIPLLLL 15
    234 AFGTCCFQML 15
    4 VVSLLLGAAL 14
    5 VSLLLGAALL 14
    61 CESEGGVLLS 14
    63 SEGGVLLSLE 14
    110 QTSGACPDLY 14
    154 NPGLGGPYLY 14
    180 YEPEINPTAP 14
    219 YVVIPTIPLL 14
    45 FHELSSRVSF 13
    70 SLENEAEQKL 13
    73 NEAEQKLIES 13
    80 IESMLQNLTK 13
    94 ISDGDFWIGL 13
    112 SGACPDLYQW 13
    120 QWSDGSNSQY 13
    123 DGSNSQYRNW 13
    139 CGSEKCVVMY 13
    148 YHQPTANPGL 13
    152 TANPGLGGPY 13
    156 GLGGPYLYQW 13
    185 NPTAPVEKPY 13
    186 PTAPVEKPYL 13
    208 VTEAGIIPNL 13
    210 EAGIIPNLIY 13
    26 SGQKVCFADF 12
    36 KHPCYKMAYF 12
    39 CYKMAYFHEL 12
    46 HELSSRVSFQ 12
    124 GSNSQYRNWY 12
    135 DEPSCGSEKC 12
    175 NYICKYEPEI 12
    213 IIPNLIYVVI 12
    218 IYVVIPTIPL 12
    252 TSPNQSTLWI 12
  • TABLE XLV
    58P1D12v.1-B5101-10-mers
    No Results Found.
  • TABLE XLVI
    58P1D12v.1-DRB1 0101-15-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 15 amino acids, and the end position for each
     peptide is the start position plus fourteen.
    2 SRVVSLLLGAALLCG 33
    42 MAYFHELSSRVSFQE 33
    49 SSRVSFQEARLACES 33
    216 NLIYVVIPTIPLLLL 31
    215 PNLIYVVIPTIPLLL 29
    226 PLLLLILVAFGTCCF 29
    101 IGLWRNGDGQTSGAC 28
    144 CVVMYHQPTANPGLG 28
    8 LLGAALLCGHGAFCR 27
    130 RNWYTDEPSCGSEKC 27
    3 RVVSLLLGAALLCGH 26
    84 LQNLTKPGTGISDGD 26
    116 PDLYQWSDGSNSQYR 26
    126 NSQYRNWYTDEPSCG 26
    203 HQNVVVTEAGIIPNL 26
    1 MSRVVSLLLGAALLC 25
    19 AFCRRVVSGQKVCFA 25
    151 PTANPGLGGPYLYQW 25
    204 QNVVVTEAGIIPNLI 25
    222 IPTIPLLLLILVAFG 25
    224 TIPLLLLILVAFGTC 25
    41 KMAYFHELSSRVSFQ 24
    211 AGIIPNLIYVVIPTI 24
    229 LLILVAFGTCCFQML 24
    192 KPYLTNQPGDTHQNV 23
    22 RRVVSGQKVCFADFK 22
    114 ACPDLYQWSDGSNSQ 22
    214 IPNLIYVVIPTIPLL 22
    218 IYVVIPTIPLLLLIL 22
    219 YVVIPTIPLLLLILV 22
    178 CKYEPEINPTAPVEK 21
    32 FADFKHPCYKMAYFH 20
    37 HPCYKMAYFHELSSR 20
    97 GDFWIGLWRNGDGQT 20
    207 VVTEAGIIPNLIYVV 20
    217 LIYVVIPTIPLLLLI 20
    237 TCCFQMLHKSKGRTK 20
    250 TKTSPNQSTLWISKS 20
    83 MLQNLTKPGTGISDG 19
    240 FQMLHKSKGRTKTSP 19
    258 TLWISKSTRKESGME 19
    27 GQKVCFADFKHPCYK 18
  • TABLE XLV
    58P1D12v.1-B5101-10-mers
    No Results Found.
  • TABLE XLVI
    58P1D12v.1-DRB1 0101-15-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 15 amino acids, and the end position for each
    peptide is the start position plus fourteen.
    56 EARLACESEGGVLLS 18
    73 NEAEQKLIESMLQNL 18
    81 ESMLQNLTKPGTGIS 18
    142 EKCVVMYHQPTANPG 18
    145 VVMYHQPTANPGLGG 18
    173 KHNYICKYEPEINPT 18
    177 ICKYEPEINPTAPVE 18
    232 LVAFGTCCFQMLHKS 18
    256 QSTLWISKSTRKESG 18
    257 STLWISKSTRKESGM 18
    4 VVSLLLGAALLCGHG 17
    11 AALLCGHGAFCRRVV 17
    21 CRRVVSGQKVCFADF 17
    57 ARLACESEGGVLLSL 17
    58 RLACESEGGVLLSLE 17
    64 EGGVLLSLENEAEQK 17
    65 GGVLLSLENEAEQKL 17
    68 LLSLENEAEQKLIES 17
    79 LIESMLQNLTKPGTG 17
    129 YRNWYTDEPSCGSEK 17
    174 HNYICKYEPEINPTA 17
    179 KYEPEINPTAPVEKP 17
    202 THQNVVVTEAGIIPN 17
    223 PTIPLLLLILVAFGT 17
    228 LLLILVAFGTCCFQM 17
    242 MLHKSKGRTKTSPNQ 17
    246 SKGRTKTSPNQSTLW 17
    253 SPNQSTLWISKSTRK 17
  • TABLE XLVII
    58P1D12v.1-DRB1 0301-15-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 15 amino acids, and the end position for each
    peptide is the start position plus fourteen.
    21 CRRVVSGQKVCFADF 27
    68 LLSLENEAEQKLIES 26
    224 TIPLLLLILVAFGTC 23
    2 SRVVSLLLGAALLCG 22
    76 EQKLIESMLQNLTKP 22
    218 IYVVIPTIPLLLLIL 22
    64 EGGVLLSLENEAEQK 21
    65 GGVLLSLENEAEQKL 21
    219 YVVIPTIPLLLLILV 21
    77 QKLIESMLQNLTKPG 20
    169 RCNMKHNYICKYEPE 20
    217 LIYVVIPTIPLLLLI 20
    229 LLILVAFGTCCFQML 20
    239 CFQMLHKSKGRTKTS 19
    66 GVLLSLENEAEQKLI 18
    92 TGISDGDFWIGLWRN 18
    142 EKCVVMYHQPTANPG 18
    29 KVCFADFKHPCYKMA 17
    39 CYKMAYFHELSSRVS 17
    43 AYFHELSSRVSFQEA 17
    48 LSSRVSFQEARLACE 17
    56 EARLACESEGGVLLS 17
    98 DFWIGLWRNGDGQTS 17
    102 GLWRNGDGQTSGACP 17
    161 YLYQWNDDRCNMKHN 17
    232 LVAFGTCCFQMLHKS 17
    28 QKVCFADFKHPCYKM 16
    33 ADFKHPCYKMAYFHE 16
    81 ESMLQNLTKPGTGIS 16
    122 SDGSNSQYRNWYTDE 16
    165 WNDDRCNMKHNYICK 16
    184 INPTAPVEKPYLTNQ 16
    208 VTEAGIIPNLIYVVI 16
    237 TCCFQMLHKSKGRTK 16
    248 GRTKTSPNQSTLWIS 16
    258 TLWISKSTRKESGME 16
    4 VVSLLLGAALLCGHG 15
    118 LYQWSDGSNSQYRNW 15
    146 VMYHQPTANPGLGGP 15
    183 EINPTAPVEKPYLTN 15
    225 IPLLLLILVAFGTCC 15
    1 MSRVVSLLLGAALLC 14
    3 RVVSLLLGAALLCGH 14
    73 NEAEQKLIESMLQNL 14
  • TABLE XLVIII
    58P1D12v.1-DRB1 0401-15-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 15 amino acids, and the end position for each
    peptide is the start position plus fourteen.
    41 KMAYFHELSSRVSFQ 28
    42 MAYFHELSSRVSFQE 28
    130 RNWYTDEPSCGSEKC 28
    177 ICKYEPEINPTAPVE 28
    216 NLIYVVIPTIPLLLL 28
    29 KVCFADFKHPCYKMA 22
    37 HPCYKMAYFHELSSR 22
    97 GDFWIGLWRNGDGQT 22
    162 LYQWNDDRCNMKHNY 22
    1 MSRVVSLLLGAALLC 20
    2 SRVVSLLLGAALLCG 20
    39 CYKMAYFHELSSRVS 20
    56 EARLACESEGGVLLS 20
    65 GGVLLSLENEAEQKL 20
    66 GVLLSLENEAEQKLI 20
    68 LLSLENEAEQKLIES 20
    77 QKLIESMLQNLTKPG 20
    81 ESMLQNLTKPGTGIS 20
    84 LQNLTKPGTGISDGD 20
    142 EKCVVMYHQPTANPG 20
    205 NVVVTEAGIIPNLIY 20
    211 AGIIPNLIYVVIPTI 20
    214 IPNLIYVVIPTIPLL 20
    219 YVVIPTIPLLLLILV 20
    222 IPTIPLLLLILVAFG 20
    226 PLLLLILVAFGTCCF 20
    229 LLILVAFGTCCFQML 20
    256 QSTLWISKSTRKESG 20
    258 TLWISKSTRKESGME 20
    14 LCGHGAFCRRVVSGQ 18
    18 GAFCRRVVSGQKVCF 18
    62 ESEGGVLLSLENEAE 18
    69 LSLENEAEQKLIESM 18
    74 EAEQKLIESMLQNLT 18
    78 KLIESMLQNLTKPGT 18
    117 DLYQWSDGSNSQYRN 18
    161 YLYQWNDDRCNMKHN 18
    195 LTNQPGDTHQNVVVT 18
    32 FADFKHPCYKMAYFH 16
    51 RVSFQEARLACESEG 16
    101 IGLWRNGDGQTSGAC 16
    116 PDLYQWSDGSNSQYR 16
    118 LYQWSDGSNSQYRNW 16
    126 NSQYRNWYTDEPSCG 16
    129 YRNWYTDEPSCGSEK 16
    158 GGPYLYQWNDDRCNM 16
    160 PYLYQWNDDRCNMKH 16
    191 EKPYLTNQPGDTHQN 16
    232 LVAFGTCCFQMLHKS 16
    237 TCCFQMLHKSKGRTK 16
    5 VSLLLGAALLCGHGA 14
    6 SLLLGAALLCGHGAF 14
    22 RRVVSGQKVCFADFK 14
    27 GQKVCFADFKHPCYK 14
    49 SSRVSFQEARLACES 14
    64 EGGVLLSLENEAEQK 14
    76 EQKLIESMLQNLTKP 14
    80 IESMLQNLTKPGTGI 14
    98 DFWIGLWRNGDGQTS 14
    100 WIGLWRNGDGQTSGA 14
    115 CPDLYQWSDGSNSQY 14
    143 KCVVMYHQPTANPGL 14
    144 CVVMYHQPTANPGLG 14
    174 HNYICKYEPEINPTA 14
    181 EPEINPTAPVEKPYL 14
    187 TAPVEKPYLTNQPGD 14
    203 HQNVVVTEAGIIPNL 14
    204 QNVVVTEAGIIPNLI 14
    210 EAGIIPNLIYVVIPT 14
    215 PNLIYVVIPTIPLLL 14
    217 LIYVVIPTIPLLLLI 14
    218 IYVVIPTIPLLLLIL 14
    224 TIPLLLLILVAFGTC 14
    225 IPLLLLILVAFGTCC 14
    227 LLLLILVAFGTCCFQ 14
    228 LLLILVAFGTCCFQM 14
    230 LILVAFGTCCFQMLH 14
    240 FQMLHKSKGRTKTSP 14
  • TABLE XLIX
    58P1D12v.1-DRB1 1101-15-mers
    Each peptide is a portion of SEQ ID NO: 3; each
    start position is specified, the length of peptide
    is 15 amino acids, and the end position for each
    peptide is the start position plus fourteen.
    237 TCCFQMLHKSKGRTK 31
    42 MAYFHELSSRVSFQE 24
    97 GDFWIGLWRNGDGQT 24
    81 ESMLQNLTKPGTGIS 20
    171 NMKHNYICKYEPEIN 20
    215 PNLIYVVIPTIPLLL 20
    258 TLWISKSTRKESGME 20
    1 MSRVVSLLLGAALLC 18
    29 KVCFADFKHPCYKMA 18
    130 RNWYTDEPSCGSEKC 18
    225 IPLLLLILVAFGTCC 18
    96 DGDFWIGLWRNGDGQ 17
    216 NLIYVVIPTIPLLLL 17
    32 FADFKHPCYKMAYFH 16
    101 IGLWRNGDGQTSGAC 16
    116 PDLYQWSDGSNSQYR 16
    126 NSQYRNWYTDEPSCG 16
    129 YRNWYTDEPSCGSEK 16
    145 VVMYHQPTANPGLGG 16
    177 ICKYEPEINPTAPVE 16
    239 CFQMLHKSKGRTKTS 16
    15 CGHGAFCRRVVSGQK 15
    38 PCYKMAYFHELSSRV 15
    84 LQNLTKPGTGISDGD 15
    141 SEKCVVMYHQPTANP 15
    201 DTHQNVVVTEAGIIP 15
    236 GTCCFQMLHKSKGRT 15
  • TABLE XXII
    58P1D12v.4-A1-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    9 VKEEQKLVQ 20
    5 VTEAVKEEQ 19
    28 PEKKVAWKY 19
    44 QNFVILDLY 17
    60 SLEWLEITK 17
    63 WLEITKDLQ 13
    27 VPEKKVAWK 12
    48 ILDLYKEWH 12
    52 YKEWHQNNS 12
    10 KEEQKLVQT 10
    40 LTWFQNFVI 10
  • TABLE XXIII
    58P1D12v.4-A0201-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    19 SLHCGFQRV 24
    39 SLTWFQNFV 23
    32 VAWKYNNSL 20
    66 ITKDLQDEL 19
    8 AVKEEQKLV 16
    43 FQNFVILDL 16
    53 KEWHQNNSL 15
    56 HQNNSLEWL 15
    40 LTWFQNFVI 14
    46 FVILDLYKE 14
    47 VILDLYKEW 14
    60 SLEWLEITK 14
    1 QNVVVTEAV 13
    7 EAVKEEQKL 13
    24 FQRVPEKKV 13
    41 TWFQNFVIL 13
    48 ILDLYKEWH 13
    14 KLVQTSLHC 12
    59 NSLEWLEIT 12
    62 EWLEITKDL 12
  • TABLE XXIV
    58P1D12v.4-A0203-9-mers
    No Results Found.
  • TABLE XXV
    58P1D12v.4-A3-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    2 NVVVTEAVK 27
    60 SLEWLEITK 25
    26 RVPEKKVAW 21
    48 ILDLYKEWH 18
    3 VVVTEAVKE 17
    50 DLYKEWHQN 17
    14 KLVQTSLHC 16
    6 TEAVKEEQK 15
    8 AVKEEQKLV 15
    23 GFQRVPEKK 15
    27 VPEKKVAWK 15
    31 KVAWKYNNS 15
    45 NFVILDLYK 14
    46 FVILDLYKE 14
    19 SLHCGFQRV 13
  • TABLE XXVI
    58P1D12v.4-A26-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    7 EAVKEEQKL 23
    62 EWLEITKDL 22
    44 QNFVILDLY 20
    12 EQKLVQTSL 19
    46 FVILDLYKE 19
    65 EITKDLQDE 19
    66 ITKDLQDEL 18
    4 VVTEAVKEE 17
    11 EEQKLVQTS 16
    3 VVVTEAVKE 15
    2 NVVVTEAVK 14
    15 LVQTSLHCG 14
    26 RVPEKKVAW 14
    41 TWFQNFVIL 14
    43 FQNFVILDL 13
    8 AVKEEQKLV 12
    35 KYNNSLTWF 12
    54 EWHQNNSLE 12
    56 HQNNSLEWL 12
  • TABLE XXVII
    58P1D12v.4-B0702-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    27 VPEKKVAWK 13
    41 TWFQNFVIL 13
    7 EAVKEEQKL 12
    12 EQKLVQTSL 12
    43 FQNFVILDL 12
    62 EWLEITKDL 12
    24 FQRVPEKKV 11
    32 VAWKYNNSL 11
    53 KEWHQNNSL 11
    66 ITKDLQDEL 11
    56 HQNNSLEWL 10
    58 NNSLEWLEI 10
    10 KEEQKLVQT 9
    1 QNVVVTEAV 8
    8 AVKEEQKLV 8
    25 QRVPEKKVA 8
    33 AWKYNNSLT 8
    35 KYNNSLTWF 8
    19 SLHCGFQRV 7
    38 NSLTWFQNF 7
    39 SLTWFQNFV 7
    40 LTWFQNFVI 7
  • TABLE XXVIII
    58P1D12v.4-B08-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    12 EQKLVQTSL 23
    66 ITKDLQDEL 22
    27 VPEKKVAWK 18
    7 EAVKEEQKL 16
    32 VAWKYNNSL 16
    62 EWLEITKDL 15
    26 RVPEKKVAW 14
    29 EKKVAWKYN 14
    8 AVKEEQKLV 13
    33 AWKYNNSLT 12
    43 FQNFVILDL 12
    51 LYKEWHQNN 12
    6 TEAVKEEQK 11
    10 KEEQKLVQT 11
    41 TWFQNFVIL 11
    49 LDLYKEWHQ 11
    53 KEWHQNNSL 11
    56 HQNNSLEWL 11
  • TABLE XXIX
    58P1D12v.4-B1510-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    20 LHCGFQRVP 15
    41 TWFQNFVIL 15
    66 ITKDLQDEL 14
    7 EAVKEEQKL 12
    55 WHQNNSLEW 12
    62 EWLEITKDL 12
    12 EQKLVQTSL 11
    32 VAWKYNNSL 11
    43 FQNFVILDL 11
    53 KEWHQNNSL 10
    56 HQNNSLEWL 10
  • TABLE XXX
    58P1D12v.4-B2705-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    22 CGFQRVPEK 18
    12 EQKLVQTSL 17
    7 EAVKEEQKL 16
    23 GFQRVPEKK 16
    35 KYNNSLTWF 16
    41 TWFQNFVIL 16
    25 QRVPEKKVA 15
    27 VPEKKVAWK 15
    28 PEKKVAWKY 15
    32 VAWKYNNSL 15
    43 FQNFVILDL 15
    53 KEWHQNNSL 15
    66 ITKDLQDEL 15
    38 NSLTWFQNF 14
    45 NFVILDLYK 14
    62 EWLEITKDL 14
    2 NVVVTEAVK 13
    18 TSLHCGFQR 13
    44 QNFVILDLY 13
    60 SLEWLEITK 13
    6 TEAVKEEQK 12
    56 HQNNSLEWL 12
    13 QKLVQTSLH 11
    16 VQTSLHCGF 11
    48 ILDLYKEWH 11
    58 NNSLEWLEI 10
    61 LEWLEITKD 9
  • TABLE XXXI
    58P1D12v.4-B2709-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    41 TWFQNFVIL 13
    62 EWLEITKDL 13
    25 QRVPEKKVA 12
    53 KEWHQNNSL 12
    7 EAVKEEQKL 11
    38 NSLTWFQNF 11
    43 FQNFVILDL 11
    66 ITKDLQDEL 11
    12 EQKLVQTSL 10
    16 VQTSLHCGF 10
    32 VAWKYNNSL 10
    35 KYNNSLTWF 10
    56 HQNNSLEWL 10
    1 QNVVVTEAV 9
    8 AVKEEQKLV 9
    19 SLHCGFQRV 9
    39 SLTWFQNFV 9
    58 NNSLEWLEI 9
    24 FQRVPEKKV 8
    40 LTWFQNFVI 8
  • TABLE XXXII
    58P1D12v.4-B4402-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    28 PEKKVAWKY 22
    53 KEWHQNNSL 22
    62 EWLEITKDL 17
    64 LEITKDLQD 16
    41 TWFQNFVIL 15
    44 QNFVILDLY 15
    61 LEWLEITKD 15
    10 KEEQKLVQT 14
    11 EEQKLVQTS 14
    26 RVPEKKVAW 14
    34 WKYNNSLTW 14
    38 NSLTWFQNF 14
    43 FQNFVILDL 14
    47 VILDLYKEW 14
    7 EAVKEEQKL 13
    12 EQKLVQTSL 13
    35 KYNNSLTWF 13
    55 WHQNNSLEW 13
    56 HQNNSLEWL 12
    58 NNSLEWLEI 12
    6 TEAVKEEQK 11
    32 VAWKYNNSL 11
    66 ITKDLQDEL 11
  • TABLE XXXIII
    58P1D12v.4-B5101-9-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 9 amino acids, and the end position for each
    peptide is the start position plus eight.
    32 VAWKYNNSL 22
    7 EAVKEEQKL 19
    40 LTWFQNFVI 17
    27 VPEKKVAWK 15
    24 FQRVPEKKV 12
    58 NNSLEWLEI 12
    8 AVKEEQKLV 11
    43 FQNFVILDL 11
    62 EWLEITKDL 11
  • TABLE XXXIV
    58P1D12v.4-A1-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    28 VPEKKVAWKY 28
    44 FQNFVILDLY 18
    6 VTEAVKEEQK 17
    64 WLEITKDLQD 17
    61 SLEWLEITKD 15
  • TABLE XXXV
    58P1D12v.4-A0201-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    32 KVAWKYNNSL 20
    40 SLTWFQNFVI 19
    41 LTWFQNFVIL 18
    61 SLEWLEITKD 17
    15 KLVQTSLHCG 16
    66 EITKDLQDEL 16
    43 WFQNFVILDL 15
    7 TEAVKEEQKL 14
    53 YKEWHQNNSL 14
    56 WHQNNSLEWL 14
    39 NSLTWFQNFV 13
    1 HQNVVVTEAV 12
    10 VKEEQKLVQT 12
    19 TSLHCGFQRV 12
    20 SLHCGFQRVP 12
    24 GFQRVPEKKV 12
    27 RVPEKKVAWK 12
    48 VILDLYKEWH 12
    49 ILDLYKEWHQ 12
    58 QNNSLEWLEI 12
    62 LEWLEITKDL 12
    4 VVVTEAVKEE 10
    8 EAVKEEQKLV 10
    33 VAWKYNNSLT 10
    51 DLYKEWHQNN 10
    64 WLEITKDLQD 10
  • TABLE XXXVI
    58P1D12v.4-A0203-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    25 FQRVPEKKVA 10
    1 HQNVVVTEAV 9
    26 QRVPEKKVAW 9
    2 QNVVVTEAVK 8
    27 RVPEKKVAWK 8
  • TABLE XXXVII
    58P1D12v.4-A3-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    27 RVPEKKVAWK 29
    9 AVKEEQKLVQ 22
    2 QNVVVTEAVK 19
    3 NVVVTEAVKE 17
    60 NSLEWLEITK 17
    64 WLEITKDLQD 17
    32 KVAWKYNNSL 16
    48 VILDLYKEWH 16
    6 VTEAVKEEQK 15
    20 SLHCGFQRVP 15
    40 SLTWFQNFVI 15
    45 QNFVILDLYK 15
    16 LVQTSLHCGF 14
    47 FVILDLYKEW 14
    51 DLYKEWHQNN 14
  • TABLE XXXVIII
    58P1D12v.4-A26-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    66 EITKDLQDEL 23
    12 EEQKLVQTSL 19
    32 KVAWKYNNSL 19
    4 VVVTEAVKEE 18
    16 LVQTSLHCGF 18
    27 RVPEKKVAWK 16
    41 LTWFQNFVIL 16
    3 NVVVTEAVKE 15
    8 EAVKEEQKLV 14
    44 FQNFVILDLY 14
    47 FVILDLYKEW 14
    5 VVTEAVKEEQ 13
    9 AVKEEQKLVQ 13
    43 WFQNFVILDL 13
    35 WKYNNSLTWF 12
    56 WHQNNSLEWL 12
    63 EWLEITKDLQ 12
  • TABLE XXXIX
    58P1D12v.4-B0702-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    12 EEQKLVQTSL 13
    41 LTWFQNFVIL 13
    32 KVAWKYNNSL 12
    43 WFQNFVILDL 12
    66 EITKDLQDEL 12
    7 TEAVKEEQKL 11
    28 VPEKKVAWKY 11
    62 LEWLEITKDL 11
    53 YKEWHQNNSL 10
    56 WHQNNSLEWL 10
    25 FQRVPEKKVA 9
    38 NNSLTWFQNF 9
    1 HQNVVVTEAV 8
    8 EAVKEEQKLV 8
    10 VKEEQKLVQT 8
    58 QNNSLEWLEI 8
    59 NNSLEWLEIT 8
    19 TSLHCGFQRV 7
    33 VAWKYNNSLT 7
    35 WKYNNSLTWF 7
    39 NSLTWFQNFV 7
    40 SLTWFQNFVI 7
  • TABLE XL
    58P1D12v.4-B08-10-mers
    No Results Found.
  • TABLE XLI
    58P1D12v.4-B1510-10-mers
    No Results Found.
  • TABLE XLII
    58P1D12v.4-B2705-10-mers
    No Results Found.
  • TABLE XLIII
    58P1D12v.4-B2709-10-mers
    No Results Found.
  • TABLE XLIV
    58P1D12v.4-B4402-10-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 10 amino acids, and the end position for each
    peptide is the start position plus nine.
    62 LEWLEITKDL 24
    12 EEQKLVQTSL 22
    7 TEAVKEEQKL 21
    26 QRVPEKKVAW 16
    47 FVILDLYKEW 16
    34 AWKYNNSLTW 15
    43 WFQNFVILDL 15
    65 LEITKDLQDE 15
    38 NNSLTWFQNF 14
    11 KEEQKLVQTS 13
    55 EWHQNNSLEW 13
    28 VPEKKVAWKY 12
    29 PEKKVAWKYN 12
    32 KVAWKYNNSL 12
    35 WKYNNSLTWF 12
    41 LTWFQNFVIL 12
    44 FQNFVILDLY 12
    54 KEWHQNNSLE 12
    56 WHQNNSLEWL 12
    66 EITKDLQDEL 12
  • TABLE XLV
    58P1D12v.4-B5101-10-mers
    No Results Found.
  • TABLE XLVI
    58P1D12v.4-DRB1 0101-15-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide 
    is 15 amino acids, and the end position for each
    peptide is the start position plus fourteen.
    27 HCGFQRVPEKKVAWK 26
    49 FQNFVILDLYKEWHQ 23
    4 DTHQNVVVTEAVKEE 21
    6 HQNVVVTEAVKEEQK 19
    15 VKEEQKLVQTSLHCG 19
    55 LDLYKEWHQNNSLEW 19
    64 NNSLEWLEITKDLQD 19
    19 QKLVQTSLHCGFQRV 18
    46 LTWFQNFVILDLYKE 18
    61 WHQNNSLEWLEITKD 18
    23 QTSLHCGFQRVPEKK 17
    37 KVAWKYNNSLTWFQN 17
    39 AWKYNNSLTWFQNFV 17
    43 NNSLTWFQNFVILDL 17
    35 EKKVAWKYNNSLTWF 16
    51 NFVILDLYKEWHQNN 16
    58 YKEWHQNNSLEWLEI 16
    65 NSLEWLEITKDLQDE 16
    66 SLEWLEITKDLQDEL 16
    3 GDTHQNVVVTEAVKE 15
    44 NSLTWFQNFVILDLY 15
    16 KEEQKLVQTSLHCGF 14
    11 VTEAVKEEQKLVQTS 13
  • TABLE XLVII
    58P1D12v.4-DRB1 0301-15-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 15 amino acids, and the end position for each
    peptide is the start position plus fourteen.
    35 EKKVAWKYNNSLTWF 25
    50 QNFVILDLYKEWHQN 23
    12 TEAVKEEQKLVQTSL 19
    19 QKLVQTSLHCGFQRV 19
    8 NVVVTEAVKEEQKLV 18
    51 NFVILDLYKEWHQNN 18
    52 FVILDLYKEWHQNNS 18
    10 VVTEAVKEEQKLVQT 17
    43 NNSLTWFQNFVILDL 17
    46 LTWFQNFVILDLYKE 17
    23 QTSLHCGFQRVPEKK 16
    27 HCGFQRVPEKKVAWK 16
    56 DLYKEWHQNNSLEWL 15
    6 HQNVVVTEAVKEEQK 14
    15 VKEEQKLVQTSLHCG 14
    30 FQRVPEKKVAWKYNN 13
  • TABLE XLVIII
    58P1D12v.4-DRB1 0401-15-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 15 amino acids, and the end position for each
    peptide is the start position plus fourteen.
    43 NNSLTWFQNFVILDL 26
    54 ILDLYKEWHQNNSLE 26
    27 HCGFQRVPEKKVAWK 22
    39 AWKYNNSLTWFQNFV 22
    46 LTWFQNFVILDLYKE 22
    55 LDLYKEWHQNNSLEW 22
    66 SLEWLEITKDLQDEL 22
    8 NVVVTEAVKEEQKLV 20
    12 TEAVKEEQKLVQTSL 20
    4 DTHQNVVVTEAVKEE 18
    11 VTEAVKEEQKLVQTS 18
    15 VKEEQKLVQTSLHCG 18
    65 NSLEWLEITKDLQDE 18
    37 KVAWKYNNSLTWFQN 16
    49 FQNFVILDLYKEWHQ 16
    58 YKEWHQNNSLEWLEI 16
    35 EKKVAWKYNNSLTWF 15
    7 QNVVVTEAVKEEQKL 14
    19 QKLVQTSLHCGFQRV 14
    23 QTSLHCGFQRVPEKK 14
    50 QNFVILDLYKEWHQN 14
    51 NFVILDLYKEWHQNN 14
    52 FVILDLYKEWHQNNS 14
    64 NNSLEWLEITKDLQD 14
  • TABLE XLIX
    58P1D12v.4-DRB1 1101-15-mers
    Each peptide is a portion of SEQ ID NO: 9; each
    start position is specified, the length of peptide
    is 15 amino acids, and the end position for each
    peptide is the start position plus fourteen.
    66 SLEWLEITKDLQDEL 25
    51 NFVILDLYKEWHQNN 20
    8 NVVVTEAVKEEQKLV 18
    27 HCGFQRVPEKKVAWK 17
    49 FQNFVILDLYKEWHQ 17
    28 CGFQRVPEKKVAWKY 16
    55 LDLYKEWHQNNSLEW 16
    12 TEAVKEEQKLVQTSL 15
    3 GDTHQNVVVTEAVKE 14
    19 QKLVQTSLHCGFQRV 14
    23 QTSLHCGFQRVPEKK 14
    54 ILDLYKEWHQNNSLE 14
    64 NNSLEWLEITKDLQD 13
  • TABLE L
    Protein Characteristics of 58P1D12
    Bioinformatic
    58P1D12 var.1 Program URL on World Wide Web Outcome
    ORF ORF finder 380-1201 by (includes stop
    codon)
    Protein length 273aa
    Transmembrane TM Pred .ch.embnet.org/ 2 TM helices (aa1-aa21, aa220-aa243),
    region N-terminus intracellular
    HMMTop .enzim.hu/hmmtop/ 2 TM helices, (aa4-25, 217-238),
    C-terminus extracellular
    Sosui .genome.ad.jp/SOSui/ 2 TM helices (1-21aa, 214-246aa)
    N terminus extracellular
    TMHMM .cbs.dtu.dk/services/TMHMM 1TM helix (218-240aa),
    N-terminus extracellular
    Signal Peptide Signal P .cbs.dtu.dk/services/SignalP/ Yes
    PI pI/MW tool .expasy.ch/tools/ pI 6.38
    Molecular weight pI/MW tool .expasy.ch/tools/ 30.4 kDa
    Localization PSORT psort.nibb.ac.jp/ Endoplasmic reticulum, plasma
    membrane
    PSORT II psort.nibb.ac.jp/ 44.4%: endoplasmic reticulum
    33.3%: Golgi
    22.2%: plasma membrane
    Motifs Pfam .sanger.ac.uk/Pfam/ C-type lectin
    Prints .biochem.ucl.ac.uk/ Mannose receptor, C-type lectin
    Blocks .blocks.fhcrc.org/ C-type lectin, pancreatitis
    associated protein
  • TABLE LI
    Exon boundaries of transcript 58P1D12 v.1
    Exon Start End Length
    1 1 458 458
    2 459 768 310
    3 769 926 158
    4 927 1013 87
    5 1014 1116 103
    6 1117 2533 1417
  • TABLE LII(a)
    Nucleotide sequence of transcript variant 58P1D12 v.2 (SEQ ID NO: 60)
    ctgtggtgtt tttcccccgc tcctctggct gccttcctga tggatctctg tggtcccagg 60
    caggaatggc ctgcttgggg acccagcgag ctcccaaggc ctttcctgct gcttcctcta 120
    tccctgtgtt ttgcttggct ctctaaattg actcagctcc aggacatcag gaccccaggt 180
    tctctggtct tgggactctg agacttgcac caggaatcct gcccaggctc tcaggccttt 240
    ggactcagac tgagctactt cactggcttt cctggttctc cagcttgaag atggcagatc 300
    gtgggacttc tcagcctcca taattgagtg agccaattcc ctggccaaaa ggtgtgtttt 360
    gctgacttca agcatccctg ctacaaaatg gcctacttcc atgaactgtc cagccgagtg 420
    agctttcagg aggcacgcct ggcttgtgag agtgagggag gagtcctcct cagccttgag 480
    aatgaagcag aacagaagtt aatagagagc atgttgcaaa acctgacaaa acccgggaca 540
    gggatttctg atggtgattt ctggataggg ctttggagga atggagatgg gcaaacatct 600
    ggtgcctgcc cagatctcta ccagtggtct gatggaagca attcccagta ccgaaactgg 660
    tacacagatg aaccttcctg cggaagtgaa aagtgtgttg tgatgtatca ccaaccaact 720
    gccaatcctg gccttggggg tccctacctt taccagtgga atgatgacag gtgtaacatg 780
    aagcacaatt atatttgcaa gtatgaacca gagattaatc caacagcccc tgtagaaaag 840
    ccttatctta caaatcaacc aggagacacc catcagaatg tggttgttac tgaagcaggt 900
    ataattccca atctaattta tgttgttata ccaacaatac ccctgctctt actgatactg 960
    gttgcttttg gaacctgttg tttccagatg ctgcataaaa gtaaaggaag aacaaaaact 1020
    agtccaaacc agtctacact gtggatttca aagagtacca gaaaagaaag tggcatggaa 1080
    gtataataac tcattgactt ggttccagaa ttttgtaatt ctggatctgt ataaggaatg 1140
    gcatcagaac aatagcttgg aatggcttga aatcacaaag gatctgcaag atgaactgta 1200
    agctccccct tgaggcaaat attaaagtaa tttttatatg tctattattt catttaaaga 1260
    atatgctgtg ctaataatgg agtgagacat gcttattttg ctaaaggatg cacccaaact 1320
    tcaaacttca agcaaatgaa atggacaatg cagataaagt tgttatcaac acgtcgggag 1380
    tatgtgtgtt agaagcaatt ccttttattt ctttcacctt tcataagttg ttatctagtc 1440
    aatgtaatgt atattgtatt gaaatttaca gtgtgcaaaa gtattttacc tttgcataag 1500
    tgtttgataa aaatgaactg ttctaatatt tatttttatg gcatctcatt tttcaataca 1560
    tgctcttttg attaaagaaa cttattactg ttgtcaactg aattcacaca cacacaaata 1620
    tagtaccata gaaaaagttt gttttctcga aataattcat ctttcagctt ctctgctttt 1680
    ggtcaatgtc taggaaatct cttcagaaat aagaagctat ttcattaagt gtgatataaa 1740
    cctcctcaaa cattttactt agaggcaagg attgtctaat ttcaattgtg caagacatgt 1800
    gccttataat tatttttagc ttaaaattaa acagattttg taataatgta actttgttaa 1860
    taggtgcata aacactaatg cagtcaattt gaacaaaaga agtgacatac acaatataaa 1920
    tcatatgtct tcacacgttg cctatataat gagaagcagc tctctgaggg ttctgaaatc 1980
    aatgtggtcc ctctcttgcc cactaaacaa agatggttgt tcggggtttg ggattgacac 2040
    tggaggcaga tagttgcaaa gttagtctaa ggtttcccta gctgtattta gcctctgact 2100
    atattagtat acaaagaggt catgtggttg agaccaggtg aatagtcact atcagtgtgg 2160
    agacaagcac agcacacaga cattttagga aggaaaggaa ctacgaaatc gtgtgaaaat 2220
    gggttggaac ccatcagtga tcgcatattc attgatgagg gtttgcttga gatagaaaat 2280
    ggtggctcct ttctgtctta tctcctagtt tcttcaatgc ttacgccttg ttcttctcaa 2340
    gagaaagttg taactctctg gtcttcatat gtccctgtgc tccttttaac caaataaaga 2400
    gttcttgttt ctgaagaa 2418
  • TABLE LIII(a)
    Nucleotide sequence alignment of 58P1D12 v.1 (SEQ ID NO: 61) and 58P1D12 v.2 (SEQ ID
    NO: 62)
    Score = 3992 bits (2076), Expect =0.0Identities = 2076/2076 (100%) Strand = Plus/Plus
    Figure US20110008393A1-20110113-C00001
    Figure US20110008393A1-20110113-C00002
    Figure US20110008393A1-20110113-C00003
    Figure US20110008393A1-20110113-C00004
  • TABLE LIV(a)
    Peptide sequences of protein coded by 58P1D12 v.2 (SEQ ID NO: 63)
    MAYFHELSSR VSFQEARLAC ESEGGVLLSL ENEAEQKLIE SMLQNLTKPG TGISDGDFWI 60
    GLWRNGDGQT SGACPDLYQW SDGSNSQYRN WYTDEPSCGS EKCVVMYHQP TANPGLGGPY 120
    LYQWNDDRCN MKHNYICKYE PEINPTAPVE KPYLTNQPGD THQNVVVTEA GIIPNLIYVV 180
    IPTIPLLLLI LVAFGTCCFQ MLHKSKGRTK TSPNQSTLWI SKSTRKESGM EV 232
  • TABLE LV(a)
    Amino acid sequence alignment of 58P1D12 v.1 (SEQ ID NO: 64)
    and 58P1D12 v.2 (SEQ ID NO: 65)
    Score = 493 bits (1269), Expect = e-138 Identities = 232/232 (100%),
    Positives = 232/232 (100%)
    V.1 42 MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI 101
    MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI
    V.2 1 MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI 60
    V.1 102 GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY 161
    GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY
    V.2 61 GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY 120
    V.1 162 LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEAGIIPNLIYVV 221
    LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEAGIIPNLIYVV
    V.2 121 LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEAGIIPNLIYVV 180
    V.1 222 IPTIPLLLLILVAFGTCCFQMLHKSKGRTKTSPNQSTLWISKSTRKESGMEV 273
    IPTIPLLLLILVAFGTCCFQMLHKSKGRTKTSPNQSTLWISKSTRKESGMEV
    V.2 181 IPTIPLLLLILVAFGTCCFQMLHKSKGRTKTSPNQSTLWISKSTRKESGMEV 232
  • TABLE LII(b)
    Nucleotide sequence of transcript variant 58P1D12 v.3 (SEQ ID NO: 66)
    gcttgaagat ggcagatcgt gggacttctc agcctccata attgagtgag ccaattccct 60
    gaatacaaca agaagatggc catctgtaga ccaggaggtg gtccctcccc agaaactgga 120
    tgggccagca cgttgattct gagcttctag cctccagaac tgccaaaagg tgtgttttgc 180
    tgacttcaag catccctgct acaaaatggc ctacttccat gaactgtcca gccgagtgag 240
    ctttcaggag gcacgcctgg cttgtgagag tgagggagga gtcctcctca gccttgagaa 300
    tgaagcagaa cagaagttaa tagagagcat gttgcaaaac ctgacaaaac ccgggacagg 360
    gatttctgat ggtgatttct ggatagggct ttggaggaat ggagatgggc aaacatctgg 420
    tgcctgccca gatctctacc agtggtctga tggaagcaat tcccagtacc gaaactggta 480
    cacagatgaa ccttcctgcg gaagtgaaaa gtgtgttgtg atgtatcacc aaccaactgc 540
    caatcctggc cttgggggtc cctaccttta ccagtggaat gatgacaggt gtaacatgaa 600
    gcacaattat atttgcaagt atgaaccaga gattaatcca acagcccctg tagaaaagcc 660
    ttatcttaca aatcaaccag gagacaccca tcagaatgtg gttgttactg aagcaggtat 720
    aattcccaat ctaatttatg ttgttatacc aacaataccc ctgctcttac tgatactggt 780
    tgcttttgga acctgttgtt tccagatgct gcataaaagt aaaggaagaa caaaaactag 840
    tccaaaccag tctacactgt ggatttcaaa gagtaccaga aaagaaagtg gcatggaagt 900
    ataataactc attgacttgg ttccagaatt ttgtaattct ggatctgtat aaggaatggc 960
    atcagaacaa tagcttggaa tggcttgaaa tcacaaagga tctgcaagat gaactgtaag 1020
    ctcccccttg aggcaaatat taaagtaatt tttatatgtc tattatttca tttaaagaat 1080
    atgctgtgct aataatggag tgagacatgc ttattttgct aaaggatgca cccaaacttc 1140
    aaacttcaag caaatgaaat ggacaatgca gataaagttg ttatcaacac gtcgggagta 1200
    tgtgtgttag aagcaattcc ttttatttct ttcacctttc ataagttgtt atctagtcaa 1260
    tgtaatgtat attgtattga aatttacagt gtgcaaaagt attttacctt tgcataagtg 1320
    tttgataaaa atgaactgtt ctaatattta tttttatggc atctcatttt tcaatacatg 1380
    ctcttttgat taaagaaact tattactgtt gtcaactgaa ttcacacaca cacaaatata 1440
    gtaccataga aaaagtttgt tttctcgaaa taattcatct ttcagcttct ctgcttttgg 1500
    tcaatgtcta ggaaatctct tcagaaataa gaagctattt cattaagtgt gatataaacc 1560
    tcctcaaaca ttttacttag aggcaaggat tgtctaattt caattgtgca agacatgtgc 1620
    cttataatta tttttagctt aaaattaaac agattttgta ataatgtaac tttgttaata 1680
    ggtgcataaa cactaatgca gtcaatttga acaaaagaag tgacatacac aatataaatc 1740
    atatgtcttc acacgttgcc tatataatga gaagcagctc tctgagggtt ctgaaatcaa 1800
    tgtggtccct ctcttgccca ctaaacaaag atggttgttc ggggtttggg attgacactg 1860
    gaggcagata gttgcaaagt tagtctaagg tttccctagc tgtatttagc ctctgactat 1920
    attagtatac aaagaggtca tgtggttgag accaggtgaa tagtcactat cagtgtggag 1980
    acaagcacag cacacagaca ttttaggaag gaaaggaact acgaaatcgt gtgaaaatgg 2040
    gttggaaccc atcagtgatc gcatattcat tgatgagggt ttgcttgaga tagaaaatgg 2100
    tggctccttt ctgtcttatc tcctagtttc ttcaatgctt acgccttgtt cttctcaaga 2160
    gaaagttgta actctctggt cttcatatgt ccctgtgctc cttttaacca aataaagagt 2220
    tcttgtttct gaagaa 2236
  • TABLE LIII(b)
    Nucleotide sequence alignment of 58P1D12 v.1 (SEQ ID NO: 67) and 58P1D12 v.3 (SEQ ID
    NO: 68)
    Score = 3990 bits (2075), Expect = 0.0Identities = 2075/2075 (100%) Strand = Plus/Plus
    Figure US20110008393A1-20110113-C00005
    Figure US20110008393A1-20110113-C00006
    Figure US20110008393A1-20110113-C00007
    Figure US20110008393A1-20110113-C00008
  • TABLE LIV(b)
    Peptide sequences of protein coded by 58P1D12 v.3 (SEQ ID NO: 69)
    MAYFHELSSR VSFQEARLAC ESEGGVLLSL ENEAEQKLIE SMLQNLTKPG TGISDGDFWI 60
    GLWRNGDGQT SGACPDLYQW SDGSNSQYRN WYTDEPSCGS EKCVVMYHQP TANPGLGGPY 120
    LYQWNDDRCN MKHNYICKYE PEINPTAPVE KPYLTNQPGD THQNVVVTEA GIIPNLIYVV 180
    IPTIPLLLLI LVAFGTCCFQ MLHKSKGRTK TSPNQSTLWI SKSTRKESGM EV 232
  • TABLE LV(b)
    Amino acid sequence alignment of 58P1D12 v.1 (SEQ ID NO: 70)
    and 58P1D12 v.3 (SEQ ID NO: 71)
    Score = 493 bits (1269), Expect = e-138 Identities = 232/232 (100%),
    Positives = 232/232 (100%)
    V.1 42 MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI 101
    MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI
    V.3 1 MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI 60
    V.1 102 GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY 161
    GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY
    V.3 61 GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY 120
    V.1 162 LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEAGIIPNLIYVV 221
    LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEAGIIPNLIYVV
    V.3 121 LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEAGIIPNLIYVV 180
    V.1 222 IPTIPLLLLILVAFGTCCFQMLHKSKGRTKTSPNQSTLWISKSTRKESGMEV 273
    IPTIPLLLLILVAFGTCCFQMLHKSKGRTKTSPNQSTLWISKSTRKESGMEV
    V.3 181 IPTIPLLLLILVAFGTCCFQMLHKSKGRTKTSPNQSTLWISKSTRKESGMEV 232
  • TABLE LII(c)
    Nucleotide sequence of transcript variant 58P1D12 v.4 (SEQ ID NO: 72)
    gcttgaagat ggcagatcgt gggacttctc agcctccata attgagtgag ccaattccct 60
    gaatacaaca agaagatggc catctgtaga ccaggaggtg gtccctcccc agaaactgga 120
    tgggccagca cgttgattct gagcttctag cctccagaac tgccaaaagg tgtgttttgc 180
    tgacttcaag catccctgct acaaaatggc ctacttccat gaactgtcca gccgagtgag 240
    ctttcaggag gcacgcctgg cttgtgagag tgagggagga gtcctcctca gccttgagaa 300
    tgaagcagaa cagaagttaa tagagagcat gttgcaaaac ctgacaaaac ccgggacagg 360
    gatttctgat ggtgatttct ggatagggct ttggaggaat ggagatgggc aaacatctgg 420
    tgcctgccca gatctctacc agtggtctga tggaagcaat tcccagtacc gaaactggta 480
    cacagatgaa ccttcctgcg gaagtgaaaa gtgtgttgtg atgtatcacc aaccaactgc 540
    caatcctggc cttgggggtc cctaccttta ccagtggaat gatgacaggt gtaacatgaa 600
    gcacaattat atttgcaagt atgaaccaga gattaatcca acagcccctg tagaaaagcc 660
    ttatcttaca aatcaaccag gagacaccca tcagaatgtg gttgttactg aagcagtaaa 720
    ggaagaacaa aaactagtcc aaaccagtct acactgtgga tttcaaagag taccagaaaa 780
    gaaagtggca tggaagtata ataactcatt gacttggttc cagaattttg taattctgga 840
    tctgtataag gaatggcatc agaacaatag cttggaatgg cttgaaatca caaaggatct 900
    gcaagatgaa ctgtaagctc ccccttgagg caaatattaa agtaattttt atatgtctat 960
    tatttcattt aaagaatatg ctgtgctaat aatggagtga gacatgctta ttttgctaaa 1020
    ggatgcaccc aaacttcaaa cttcaagcaa atgaaatgga caatgcagat aaagttgtta 1080
    tcaacacgtc gggagtatgt gtgttagaag caattccttt tatttctttc acctttcata 1140
    agttgttatc tagtcaatgt aatgtatatt gtattgaaat ttacagtgtg caaaagtatt 1200
    ttacctttgc ataagtgttt gataaaaatg aactgttcta atatttattt ttatggcatc 1260
    tcatttttca atacatgctc ttttgattaa agaaacttat tactgttgtc aactgaattc 1320
    acacacacac aaatatagta ccatagaaaa agtttgtttt ctcgaaataa ttcatctttc 1380
    agcttctctg cttttggtca atgtctagga aatctcttca gaaataagaa gctatttcat 1440
    taagtgtgat ataaacctcc tcaaacattt tacttagagg caaggattgt ctaatttcaa 1500
    ttgtgcaaga catgtgcctt ataattattt ttagcttaaa attaaacaga ttttgtaata 1560
    atgtaacttt gttaataggt gcataaacac taatgcagtc aatttgaaca aaagaagtga 1620
    catacacaat ataaatcata tgtcttcaca cgttgcctat ataatgagaa gcagctctct 1680
    gagggttctg aaatcaatgt ggtccctctc ttgcccacta aacaaagatg gttgttcggg 1740
    gtttgggatt gacactggag gcagatagtt gcaaagttag tctaaggttt ccctagctgt 1800
    atttagcctc tgactatatt agtatacaaa gaggtcatgt ggttgagacc aggtgaatag 1860
    tcactatcag tgtggagaca agcacagcac acagacattt taggaaggaa aggaactacg 1920
    aaatcgtgtg aaaatgggtt ggaacccatc agtgatcgca tattcattga tgagggtttg 1980
    cttgagatag aaaatggtgg ctcctttctg tcttatctcc tagtttcttc aatgcttacg 2040
    ccttgttctt ctcaagagaa agttgtaact ctctggtctt catatgtccc tgtgctcctt 2100
    ttaaccaaat aaagagttct tgtttctgaa gaa 2133
  • TABLE LIII(c)
    Nucleotide sequence alignment of 58P1D12 v.1 (SEQ ID NO: 73) and 58P1D12 v.4 (SEQ ID
    NO: 74)
    Score = 1067 bits (555), Expect = 0.0Identities =555/555 (100%) Strand = Plus/Plus
    Figure US20110008393A1-20110113-C00009
    Score = 2728 bits (1419) , Expect = 0.0Identities = 1419/1419 (100%)
    Strand = Plus/Plus
    Figure US20110008393A1-20110113-C00010
    Figure US20110008393A1-20110113-C00011
    Figure US20110008393A1-20110113-C00012
    Figure US20110008393A1-20110113-C00013
  • TABLE LIV(c)
    Peptide sequences of protein coded by 58P1D12 v.4 (SEQ ID NO: 75)
    MAYFHELSSR VSFQEARLAC ESEGGVLLSL ENEAEQKLIE SMLQNLTKPG TGISDGDFWI 60
    GLWRNGDGQT SGACPDLYQW SDGSNSQYRN WYTDEPSCGS EKCVVMYHQP TANPGLGGPY 120
    LYQWNDDRCN MKHNYICKYE PEINPTAPVE KPYLTNQPGD THQNVVVTEA VKEEQKLVQT 180
    SLHCGFQRVP EKKVAWKYNN SLTWFQNFVI LDLYKEWHQN NSLEWLEITK DLQDEL 236
  • TABLE LV(c)
    Amino acid sequence alignment of 58P1D12 v.1 (SEQ ID NO: 76)
    and 58P1D12 v.4 (SEQ ID NO: 77)
    Score = 370 bits (950), Expect = e-101 Identities = 170/170 (100%),
    Positives = 170/170 (100%)
    V.1 42 MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI 101
    MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI
    V.4 1 MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI 60
    V.1 102 GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY 161
    GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY
    V.4 61 GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY 120
    V.1 162 LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEA 211
    LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEA
    V.4 121 LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEA 170
  • TABLE LII(d)
    Nucleotide sequence of transcript variant 58P1D12 v.5 (SEQ ID NO: 78)
    gcttgaagat ggcagatcgt gggacttctc agcctccata attgagtgag ccaattccct 60
    ggccaaaagg tgtgttttgc tgacttcaag catccctgct acaaaatggc ctacttccat 120
    gaactgtcca gccgagtgag ctttcaggag gcacgcctgg cttgtgagag tgagggagga 180
    gtcctcctca gccttgagaa tgaagcagaa cagaagttaa tagagagcat gttgcaaaac 240
    ctgacaaaac ccgggacagg gatttctgat ggtgatttct ggatagggct ttggaggaat 300
    ggagatgggc aaacatctgg tgcctgccca gatctctacc agtggtctga tggaagcaat 360
    tcccagtacc gaaactggta cacagatgaa ccttcctgcg gaagtgaaaa gtgtgttgtg 420
    atgtatcacc aaccaactgc caatcctggc cttgggggtc cctaccttta ccagtggaat 480
    gatgacaggt gtaacatgaa gcacaattat atttgcaagt atgaaccaga gattaatcca 540
    acagcccctg tagaaaagcc ttatcttaca aatcaaccag gagacaccca tcagaatgtg 600
    gttgttactg aagcagtaaa ggaagaacaa aaactagtcc aaaccagtct acactgtgga 660
    tttcaaagag taccagaaaa gaaagtggca tggaagtata ataactcatt gacttggttc 720
    cagaattttg taattctgga tctgtataag gaatggcatc agaacaatag cttggaatgg 780
    cttgaaatca caaaggatct gcaagatgaa ctgtaagctc ccccttgagg caaatattaa 840
    agtaattttt atatgtctat tatttcattt aaagaatatg ctgtgctaat aatggagtga 900
    gacatgctta ttttgctaaa ggatgcaccc aaacttcaaa cttcaagcaa atgaaatgga 960
    caatgcagat aaagttgtta tcaacacgtc gggagtatgt gtgttagaag caattccttt 1020
    tatttctttc acctttcata agttgttatc tagtcaatgt aatgtatatt gtattgaaat 1080
    ttacagtgtg caaaagtatt ttacctttgc ataagtgttt gataaaaatg aactgttcta 1140
    atatttattt ttatggcatc tcatttttca atacatgctc ttttgattaa agaaacttat 1200
    tactgttgtc aactgaattc acacacacac aaatatagta ccatagaaaa agtttgtttt 1260
    ctcgaaataa ttcatctttc agcttctctg cttttggtca atgtctagga aatctcttca 1320
    gaaataagaa gctatttcat taagtgtgat ataaacctcc tcaaacattt tacttagagg 1380
    caaggattgt ctaatttcaa ttgtgcaaga catgtgcctt ataattattt ttagcttaaa 1440
    attaaacaga ttttgtaata atgtaacttt gttaataggt gcataaacac taatgcagtc 1500
    aatttgaaca aaagaagtga catacacaat ataaatcata tgtcttcaca cgttgcctat 1560
    ataatgagaa gcagctctct gagggttctg aaatcaatgt ggtccctctc ttgcccacta 1620
    aacaaagatg gttgttcggg gtttgggatt gacactggag gcagatagtt gcaaagttag 1680
    tctaaggttt ccctagctgt atttagcctc tgactatatt agtatacaaa gaggtcatgt 1740
    ggttgagacc aggtgaatag tcactatcag tgtggagaca agcacagcac acagacattt 1800
    taggaaggaa aggaactacg aaatcgtgtg aaaatgggtt ggaacccatc agtgatcgca 1860
    tattcattga tgagggtttg cttgagatag aaaatggtgg ctcctttctg tcttatctcc 1920
    tagtttcttc aatgcttacg ccttgttctt ctcaagagaa agttgtaact ctctggtctt 1980
    catatgtccc tgtgctcctt ttaaccaaat aaagagttct tgtttctgaa gaa 2033
  • TABLE LIII(d)
    Nucleotide sequence alignment of 58P1D12 v.1 (SEQ ID NO: 79) and 58P1D12 v.5 (SEQ ID
    NO: 80)
    Score = 1069 bits (556), Expect = 0.0Identities = 556/556 (100%) Strand = Plus/Plus
    Figure US20110008393A1-20110113-C00014
    Score = 2728 bits (1419), Expect = 0.0Identities = 1419/1419 (100%)
    Strand = Plus/Plus
    Figure US20110008393A1-20110113-C00015
    Figure US20110008393A1-20110113-C00016
    Figure US20110008393A1-20110113-C00017
    Figure US20110008393A1-20110113-C00018
  • TABLE LIV(d)
    Peptide sequences of protein coded by 58P1D12 v.5 (SEQ ID NO: 81)
    MAYFHELSSR VSFQEARLAC ESEGGVLLSL ENEAEQKLIE SMLQNLTKPG TGISDGDFWI 60
    GLWRNGDGQT SGACPDLYQW SDGSNSQYRN WYTDEPSCGS EKCVVMYHQP TANPGLGGPY 120
    LYQWNDDRCN MKHNYICKYE PEINPTAPVE KPYLTNQPGD THQNVVVTEA VKEEQKLVQT 180
    SLHCGFQRVP EKKVAWKYNN SLTWFQNFVI LDLYKEWHQN NSLEWLEITK DLQDEL 236
  • TABLE LV(d)
    Amino acid sequence alignment of 58P1D12 v.1 (SEQ ID NO: 82)
    and 58P1D12 v.5 (SEQ ID NO: 83)
    Score = 370 bits (950), Expect = e-101 Identities = 170/170 (100%),
    Positives = 170/170 (100%)
    V.1 42 MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI 101
    MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI
    V.5 1 MAYFHELSSRVSFQEARLACESEGGVLLSLENEAEQKLIESMLQNLTKPGTGISDGDFWI 60
    V.1 102 GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY 161
    GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY
    V.5 61 GLWRNGDGQTSGACPDLYQWSDGSNSQYRNWYTDEPSCGSEKCVVMYHQPTANPGLGGPY 120
    V.1 162 LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEA 211
    LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEA
    V.5 121 LYQWNDDRCNMKHNYICKYEPEINPTAPVEKPYLTNQPGDTHQNVVVTEA 170

Claims (34)

1. An isolated transcript variant of a 58P1D12 gene, wherein the transcript variant is transcribed from the 58P1D12 gene and encodes a protein comprising at least one amino acid substitution, addition or deletion relative to SEQ ID NO:12.
2. The transcript variant of claim 1, wherein the transcript variant comprises a nucleotide sequence selected from the group consisting of SEQ ID NO:4 or SEQ ID NO:8.
3. The transcript variant of claim 1, wherein the transcript variant encodes a protein comprising the amino acid sequence selected from the group consisting of SEQ ID NO:13, or SEQ ID NO:14.
4. A recombinant expression vector comprising a transcript variant, wherein the transcript variant is transcribed from the 58P1D12 gene and encodes a protein comprising at least one amino acid substitution, addition or deletion relative to SEQ ID NO:12.
5. The recombinant expression vector of claim 4, wherein the transcript variant comprises a nucleotide sequence selected from the group consisting of SEQ ID NO:4 or SEQ ID NO:8.
6. The recombinant expression vector of claim 4, further comprising a nucleotide sequence encoding a selection marker.
7. The recombinant expression vector of claim 4, further comprising at least one suitable regulatory sequence operably linked to the transcript variant.
8. A host cell comprising a recombinant expression vector comprising a transcript variant, wherein the transcript variant is transcribed from the 58P1D12 gene and encodes a protein comprising at least one amino acid substitution, addition or deletion relative to SEQ ID NO:12.
9. The host cell of clam 8, wherein the transcript variant comprises a nucleotide sequence selected from the group consisting of SEQ ID NO:4 or SEQ ID NO:8.
10. The host cell of claim 8, wherein the host cell is a prokaryotic cell.
11. The host cell of claim 10, wherein the prokaryotic cell is E. coli.
12. The host cell of claim 8, wherein the host cell is a eucaryotic cell.
13. The host cell of claim 12, wherein the host cell is selected from the group consisting of a yeast cell, a plant cell and an animal cell.
14. The host cell of claim 13, wherein the animal cell is a mammalian cell.
15. The host cell of claim 14, wherein the mammalian cell is selected from the group consisting of COS cells, CHO cells, 293 cells, and 293T cells.
16. An isolated protein encoded by a transcript variant of a 58P1D12 gene, wherein the transcript variant is transcribed from the 58P1D12 gene and encodes a protein comprising at least one amino acid substitution, addition or deletion relative to SEQ ID NO:12.
17. The protein of claim 16, wherein the protein is encoded by a transcript comprising the nucleic acid sequence selected from the group consisting of SEQ ID NO:4, or SEQ ID NO:8.
18. The protein of claim 16, wherein the protein comprises the amino acid sequence selected from the group consisting of SEQ ID NO:13, or SEQ ID NO:14.
19. An isolated antibody or fragment thereof that specifically binds to the protein of claim 16.
20. The antibody or fragment thereof of claim 19, wherein the antibody is monoclonal.
21. The antibody or fragment thereof of claim 19, wherein the antibody is a human antibody, a humanized antibody or a chimeric antibody.
22. The antibody or fragment thereof of claim 19, wherein the antibody or fragment thereof is labeled with an agent.
23. A method of inducing a mammalian immune response directed to a protein encoded by a transcript variant of a 58P1D12 gene, wherein the transcript variant is transcribed from the 58P1D12 gene and encodes a protein comprising at least one amino acid substitution, addition or deletion relative to SEQ ID NO:12, comprising:
exposing cells of the immune system of a mammal to an immunogenic portion of
the protein comprising at least one amino acid substitution, addition or deletion relative to SEQ ID NO:12; or
the transcript variant, or a combination of both, whereby the mammalian immune response directed to the protein is induced.
24. The method of claim 23, wherein the immunogenic portion comprises at least one T cell or at least one B cell epitope.
25. The method of claim 24, whereby the immunogenic portion induces a B cell to generate antibodies that specifically bind to the protein.
26. The method of claim 24, whereby the immunogenic portion activates a cytotoxic T cell (CTL) that kills an autologous cell that expresses the protein.
27. The method of claim 24, wherein the T cell is a helper T cell (HTL), whereby the immunogenic portion activates an HTL that secretes cytokines that facilitate the cytotoxic activity of a CTL or the antibody producing activity of a B cell.
28. The method of claim 23, wherein the immune response comprises production of an antibody that binds specifically to the protein.
29. The method of claim 23, wherein the protein is encoded by a transcript variant comprising the nucleic acid sequence selected from the group consisting of SEQ ID NO:4, or SEQ ID NO:8.
30. The method of claim 23, wherein the protein comprises the amino acid sequence selected from the group consisting of SEQ ID NO:13, or SEQ ID NO:14.
31. An immunogenic composition comprising an immunogenic portion of:
a protein encoded by a transcript variant of a 58P1D12 gene, wherein the transcript variant is transcribed from the 58P1D12 gene and encodes a protein comprising at least one amino acid substitution, addition or deletion relative to SEQ ID NO:12; and/or
transcript variant that encodes the protein.
32. The immunogenic composition of claim 31, wherein the protein is encoded by a transcript variant comprising the nucleic acid sequence selected from the group consisting of SEQ ID NO:4, or SEQ ID NO:8.
33. The immunogenic composition of claim 31, wherein the protein comprises the amino acid sequence selected from the group consisting of SEQ ID NO:13, or SEQ ID NO:14.
34. The immunogenic composition of claim 31, further comprising a pharmaceutically acceptable carrier.
US12/836,449 1999-08-12 2010-07-14 Nucleic acids and corresponding proteins entitled 58p1d12 useful in treatment and detection of cancer Abandoned US20110008393A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/836,449 US20110008393A1 (en) 1999-08-12 2010-07-14 Nucleic acids and corresponding proteins entitled 58p1d12 useful in treatment and detection of cancer

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US14893599P 1999-08-12 1999-08-12
US09/638,203 US6602501B1 (en) 1999-08-12 2000-08-11 C-type lectin transmembrane antigen expressed in human prostate cancer and uses thereof
US10/460,512 US7153940B2 (en) 1999-08-12 2003-06-11 C-type lectin transmembrane antigen expressed in human prostate cancer and uses thereof
US10/919,654 US7842458B2 (en) 1999-08-12 2004-08-16 Nucleic acids and corresponding proteins entitled 58P1D12 useful in treatment and detection of cancer
US12/836,449 US20110008393A1 (en) 1999-08-12 2010-07-14 Nucleic acids and corresponding proteins entitled 58p1d12 useful in treatment and detection of cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/919,654 Continuation US7842458B2 (en) 1999-08-12 2004-08-16 Nucleic acids and corresponding proteins entitled 58P1D12 useful in treatment and detection of cancer

Publications (1)

Publication Number Publication Date
US20110008393A1 true US20110008393A1 (en) 2011-01-13

Family

ID=46302559

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/919,654 Expired - Fee Related US7842458B2 (en) 1999-08-12 2004-08-16 Nucleic acids and corresponding proteins entitled 58P1D12 useful in treatment and detection of cancer
US12/836,449 Abandoned US20110008393A1 (en) 1999-08-12 2010-07-14 Nucleic acids and corresponding proteins entitled 58p1d12 useful in treatment and detection of cancer

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/919,654 Expired - Fee Related US7842458B2 (en) 1999-08-12 2004-08-16 Nucleic acids and corresponding proteins entitled 58P1D12 useful in treatment and detection of cancer

Country Status (1)

Country Link
US (2) US7842458B2 (en)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10857181B2 (en) 2015-04-21 2020-12-08 Enlivex Therapeutics Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
US11000548B2 (en) 2015-02-18 2021-05-11 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11304976B2 (en) 2015-02-18 2022-04-19 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11318163B2 (en) 2015-02-18 2022-05-03 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11497767B2 (en) 2015-02-18 2022-11-15 Enlivex Therapeutics R&D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11512289B2 (en) 2015-02-18 2022-11-29 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11596652B2 (en) 2015-02-18 2023-03-07 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use in treating sepsis
US11730761B2 (en) 2016-02-18 2023-08-22 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2786763A1 (en) * 2007-07-27 2014-10-08 The United States of America as Represented by The Secretary of the Navy Capsule composition for use as immunogen against Campylobacter jejuni
ES2601460T3 (en) 2011-12-30 2017-02-15 Abbott Molecular Inc. Materials and methods for the diagnosis of bladder cancer and its recurrence control
HUE065174T2 (en) * 2017-02-12 2024-05-28 Biontech Us Inc Hla-based methods and compositions and uses thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5798266A (en) * 1996-08-27 1998-08-25 K-Quay Enterprises, Llc Methods and kits for obtaining and assaying mammary fluid samples for breast diseases, including cancer
US5879898A (en) * 1992-11-20 1999-03-09 Isis Innovation Limited Antibodies specific for peptide corresponding to CD44 exon 6, and use of these antibodies for diagnosis of tumors
US20020168717A1 (en) * 1995-06-06 2002-11-14 Soppet Daniel R. Human prostate specific G-protein receptor HPRAJ70

Family Cites Families (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5681923A (en) 1995-10-06 1997-10-28 Platt; David Tumor derived carbohydrate binding protein
US6117977A (en) 1996-04-24 2000-09-12 Genentech, Inc. Type C lectins
US7435793B2 (en) * 1998-05-15 2008-10-14 Genentech, Inc. Peptides that induce chondrocyte redifferentiation
US20030004311A1 (en) * 1997-06-18 2003-01-02 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20020137890A1 (en) * 1997-03-31 2002-09-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030022239A1 (en) * 1997-06-18 2003-01-30 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US7285625B2 (en) * 1997-06-18 2007-10-23 Genentech, Inc. PRO536 polypeptides
US20030166109A1 (en) * 1997-09-18 2003-09-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030166104A1 (en) * 1997-09-18 2003-09-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US7160985B2 (en) 1997-10-29 2007-01-09 Genentech, Inc. Pro180 polypeptide
US7125959B2 (en) * 1998-05-15 2006-10-24 Genentech, Inc. PRO4405 polypeptides
US20030050465A1 (en) * 1998-08-10 2003-03-13 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030009013A1 (en) 1998-12-30 2003-01-09 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
PT1609863E (en) 1999-03-23 2009-01-02 Genentech Inc Secreted and transmembrane polypeptides and nucleic acids encoding the same
EP1074617A3 (en) * 1999-07-29 2004-04-21 Research Association for Biotechnology Primers for synthesising full-length cDNA and their use
EP1200590B1 (en) 1999-08-12 2009-01-07 Agensys, Inc. C-type lectin transmembrane antigen expressed in human prostate cancer and uses thereof
US7300758B2 (en) * 1999-12-09 2007-11-27 Genetech, Inc. Nucleic acid underexpressed in esophageal tumor
US7250495B2 (en) 2001-06-20 2007-07-31 Genentech, Inc. PRO20044 polypeptides
US7202335B2 (en) * 2001-12-06 2007-04-10 Genentech, Inc. PRO300 polypeptides
JP2004187620A (en) 2002-12-13 2004-07-08 Sumitomo Pharmaceut Co Ltd Disorder marker for kidney disease and utilization thereof
US20090068098A1 (en) * 2007-08-20 2009-03-12 Kanner Steven B Antibodies and related molecules that bind to 58p1d12 proteins

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5879898A (en) * 1992-11-20 1999-03-09 Isis Innovation Limited Antibodies specific for peptide corresponding to CD44 exon 6, and use of these antibodies for diagnosis of tumors
US20020168717A1 (en) * 1995-06-06 2002-11-14 Soppet Daniel R. Human prostate specific G-protein receptor HPRAJ70
US5798266A (en) * 1996-08-27 1998-08-25 K-Quay Enterprises, Llc Methods and kits for obtaining and assaying mammary fluid samples for breast diseases, including cancer

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11000548B2 (en) 2015-02-18 2021-05-11 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11304976B2 (en) 2015-02-18 2022-04-19 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11318163B2 (en) 2015-02-18 2022-05-03 Enlivex Therapeutics Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11497767B2 (en) 2015-02-18 2022-11-15 Enlivex Therapeutics R&D Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11512289B2 (en) 2015-02-18 2022-11-29 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment
US11596652B2 (en) 2015-02-18 2023-03-07 Enlivex Therapeutics R&D Ltd Early apoptotic cells for use in treating sepsis
US11717539B2 (en) 2015-02-18 2023-08-08 Enlivex Therapeutics RDO Ltd. Combination immune therapy and cytokine control therapy for cancer treatment
US10857181B2 (en) 2015-04-21 2020-12-08 Enlivex Therapeutics Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
US11883429B2 (en) 2015-04-21 2024-01-30 Enlivex Therapeutics Rdo Ltd Therapeutic pooled blood apoptotic cell preparations and uses thereof
US11730761B2 (en) 2016-02-18 2023-08-22 Enlivex Therapeutics Rdo Ltd Combination immune therapy and cytokine control therapy for cancer treatment

Also Published As

Publication number Publication date
US7842458B2 (en) 2010-11-30
US20050136435A1 (en) 2005-06-23

Similar Documents

Publication Publication Date Title
US20190092874A1 (en) Nucleic acid and corresponding protein entitled 161p2f10b useful in treatment and detection of cancer
US8426571B2 (en) Nucleic acids and corresponding proteins entitled 202P5A5 useful in treatment and detection of cancer
US7622564B2 (en) Prostate stem cell antigen (PSCA) variants and subsequences thereof
US7968090B2 (en) Nucleic acids and corresponding proteins entitled 191P4D12(b) useful in treatment and detection of cancer
US8524872B2 (en) Nucleic acids and corresponding proteins entitled 158P3D2 useful in treatment and detection of cancer
CA2481503A1 (en) Nucleic acid and corresponding protein entitled 98p4b6 useful in treatment and detection of cancer
AU2002361610A1 (en) Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
AU2010200938B2 (en) Nucleic acids and corresponding proteins entitled 58P1D12 useful in treatment and detection of cancer
US20110008393A1 (en) Nucleic acids and corresponding proteins entitled 58p1d12 useful in treatment and detection of cancer
CA2522994C (en) Nucleic acids and corresponding proteins entitled 109p1d4 useful in treatment and detection of cancer
EP2302041A1 (en) Nucleic acid and corresponding protein entitled 161P2F10B useful in treatment and detection of cancer
CA2496566A1 (en) Nucleic acid and corresponding protein entitled 98p4b6 useful in treatment and detection of cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: AGENSYS, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KANNER, STEVEN B.;JAKOBOVITS, AYA;CHALLITA-EID, PIA M.;AND OTHERS;SIGNING DATES FROM 20040811 TO 20040817;REEL/FRAME:032479/0361

STCB Information on status: application discontinuation

Free format text: ABANDONMENT FOR FAILURE TO CORRECT DRAWINGS/OATH/NONPUB REQUEST