WO1999013111A1 - Reagents and methods useful for detecting diseases of the prostate - Google Patents

Reagents and methods useful for detecting diseases of the prostate Download PDF

Info

Publication number
WO1999013111A1
WO1999013111A1 PCT/US1998/018983 US9818983W WO9913111A1 WO 1999013111 A1 WO1999013111 A1 WO 1999013111A1 US 9818983 W US9818983 W US 9818983W WO 9913111 A1 WO9913111 A1 WO 9913111A1
Authority
WO
WIPO (PCT)
Prior art keywords
sequence
polynucleotide
polypeptide
antigen
test sample
Prior art date
Application number
PCT/US1998/018983
Other languages
French (fr)
Inventor
Patricia A. Billing-Medel
Maurice Cohen
Tracey L. Colpitts
Paula N. Friedman
Julian Gordon
Edward N. Granados
Steven C. Hodges
Michael R. Klass
Jon D. Kratochvil
John C. Russell
Stephen D. Stroupe
Original Assignee
Abbott Laboratories
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Abbott Laboratories filed Critical Abbott Laboratories
Priority to CA002303404A priority Critical patent/CA2303404A1/en
Priority to EP98948162A priority patent/EP1012343A1/en
Publication of WO1999013111A1 publication Critical patent/WO1999013111A1/en

Links

Classifications

    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57434Specifically defined cancers of prostate
    • 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

Definitions

  • This invention relates generally to detecting diseases of the prostate. Furthermore, the invention also relates to reagents and methods for detecting diseases of the prostate. More particularly, the present invention relates to reagents such as polynucleotide sequences and the polypeptide sequences encoded thereby, as well as methods which utilize these sequences.
  • the polynucleotide and polypeptide sequences are useful for detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining predisposition to diseases or conditions of the prostate such as prostate cancer.
  • Prostate cancer is the most common form of cancer occurring in males in the United States, with projections of 184,500 new cases diagnosed and 39,200 related deaths to occur during 1998 (American Cancer Society).
  • Prostate cancer also has shown the largest increase in incidence as compared to other types of cancer, increasing 142% from 1992 to 1996.
  • Procedures used for detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining predisposition to diseases or conditions of the prostate such as prostate cancer are of critical importance to the outcome of the patient. For example, patients diagnosed with localized prostate cancer have greater than a 90% five-year survival rate compared to a rate of 25 to 31 % for patients diagnosed with distant metastasis. (American Cancer Society statistics).
  • a diagnostic procedure for early detection of prostate cancer should, therefore, specifically detect this disease and be capable of detecting the presence of prostate cancer before symptoms appear.
  • Such procedures could include assays based upon the appearance of various disease markers in test samples such as blood, plasma, serum, or urine obtained by minimally invasive procedures which are detectable by immunological methods. These procedures would provide information to aid the physician in managing the patient with disease of the prostate and at low cost to the patient.
  • Markers such as the prostate specific antigen (PSA) exist and are used clinically for screening patients for prostate cancer. Elevated levels of PSA protein in serum can be used as a marker in the early detection of prostate cancer in asymptomatic men.
  • PSA prostate specific antigen
  • PSA normally is secreted by the prostate at high levels into the seminal fluid, but is present in very low levels in the blood of men with normal prostates.
  • BPH benign prostatic hyperplasia
  • adenocarcinoma of the prostate the level of PSA can be markedly elevated in the blood and thus be useful as an indicator of prostate disease.
  • PSA cannot differentiate between BPH and prostate cancer, which reduces its specificity as a marker for prostate cancer.
  • a critical step in managing patients with prostate cancer is the presurgical staging of the cancer to provide prognostic value and criteria for designing optimal therapy. Improved procedures for accurately staging prostate cancer prior to surgery are needed.
  • Prostate cancer management also could be improved by utilizing new markers found in an inappropriate body compartment. Such markers could be mRNA or protein markers expressed by cells originating from the primary prostate tumor but residing in blood, bone marrow or lymph nodes and could be sensitive indicators for metastasis to these distal organs.
  • PSA protein has been detected by immunohistochemical techniques in bone marrow
  • PSA mRNA has been detected by RT-PCR in cells of blood, lymph nodes and bone marrow.
  • New markers which could predict the biologic behavior of early prostate cancers would also be of significant value. Early prostate cancers that threaten or will threaten the life of the patient are more clinically important than those that do not or will not be a threat. G.E. Hanks, supra. A need therefore exists for new markers which can differentiate between the clinically important and unimportant prostate cancers. Such markers would allow the clinician to accurately identify and effectively treat early cancers localized to the prostate which could otherwise metastasize and kill the patient. Further, if one could show that such a marker characteristic of aggressive cancer was absent, the patient could be spared expensive and non-beneficial treatment. It also would be beneficial to find a prostate associated marker which is more sensitive in detecting recurrence of prostate cancer than PSA and which is not affected by androgens.
  • PSA has proven to be the most sensitive marker for detecting recurrent disease.
  • tumor progression occurs without PSA elevation due to hormonal therapy utilized for treating the cancer.
  • the decrease in androgen results in a concomitant decrease in PSA, it does not necessarily reflect a decrease in tumor metastasis.
  • This complication is the result of androgen-stimulated PSA expression.
  • Part of the decline in PSA observed after androgen ablation is due not to tumor cell death but to diminished PSA expression. G.E. Hanks, supra.
  • Such methods would include assaying a test sample for products of a gene which are overexpressed in prostate diseases and conditions such as cancer. Such methods may also include assaying a test sample for products of a gene alteration associated with prostate disease or condition. Such methods may further include assaying a test sample for products of a gene whose distribution among the various tissues and compartments of the body have been altered by a prostate- associated disease or condition such as cancer.
  • Useful reagents include polynucleotide(s), or fragment(s) thereof which may be used in diagnostic methods such as reverse transcriptase-polymerase chain reaction (RT-PCR), PCR, or hybridization assays of mRNA extracted from biopsied tissue, blood or other test samples; polypeptides or proteins which are the translation products of such mRNAs; or antibodies directed against these polypeptides or proteins.
  • RT-PCR reverse transcriptase-polymerase chain reaction
  • PCR reverse transcriptase-polymerase chain reaction
  • This invention relates to uses for a newly discovered polynucleotide (depicted in SEQUENCE ID NO 1 and described by Brash, A. R. et al., (1997) Proc Natl Acad Sci USA 94(12):6148-6152) and polypeptide encoded by this polynucleotide(depicted in SEQUENCE ID NO 6 herein and also described by Brash et al., supra).
  • This polynucleotide represents a second human 15S- lipoxygenase.
  • This polynucleotide and polypeptide will be hereinafter referred to as PS213.
  • the present invention provides a method of detecting a target PS213 polynucleotide in a test sample which comprises contacting the test sample with at least one PS213-specific polynucleotide, wherein the PS213 polynucleotide has at least 50% identity with (i) SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1 , or (iii) complements of (i) or (ii), and detecting the presence of the target PS213 polynucleotides in the test sample which bind to the PS213-specific polynucleotide.
  • the PS213-specific polynucleotide may be attached to a solid phase prior to performing the method.
  • the present invention also provides a method for detecting PS213 mRNA in a test sample, which comprises performing reverse transcription (RT) with at least one primer in order to produce cDNA, amplifying the cDNA so obtained using PS213 oligonucleotides as sense and antisense primers to obtain PS213 amplicon, and detecting the presence of the PS213 amplicon as an indication of the presence of PS213 mRNA in the test sample.
  • the PS213 oligonucleotides utilized in the method have at least 50% identity with (i) SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1, or (iii) complements of (i) or (ii). Amplification can be performed by the polymerase chain reaction.
  • test sample can be reacted with a solid phase prior to performing the method, prior to amplification or prior to detection.
  • This reaction can be a direct or an indirect reaction.
  • detection step can comprise utilizing a detectable label capable of generating a measurable signal.
  • the detectable label can be attached to a solid phase.
  • the present invention further provides a method of detecting a target PS213 polynucleotide in a test sample suspected of containing target PS213 polynucleotides, which comprises (a) contacting the test sample with at least one PS213 oligonucleotide as a sense primer and at least one PS213 oligonucleotide as an anti-sense primer, and amplifying same to obtain a first stage reaction product; (b) contacting the first stage reaction product with at least one other PS213 oligonucleotide to obtain a second stage reaction product, with the proviso that the other PS213 oligonucleotide is located 3' to the PS213 oligonucleotides utilized in step (a) and is complementary to the first stage reaction product; and (c) detecting the second stage reaction product as an indication of the presence of a target PS213 polynucleotide in the test sample.
  • the PS213 oligonucleotides used in the method have at least 50% identity with (i) SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1, or (iii) complements of (i) or (ii).
  • Amplification may be performed by the polymerase chain reaction.
  • the test sample can be reacted either directly or indirectly with a solid phase prior to performing the method, or prior to amplification, or prior to detection.
  • the detection step may also comprise utilizing a detectable label capable of generating a measurable signal; further, the detectable label can be attached to a solid phase.
  • Test kits useful for detecting target PS213 polynucleotides in a test sample comprise a container containing at least one PS213- specific polynucleotide.
  • the polynucleotide has at least 50% identity with (i) SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1, or (iii) complements of (i) or (ii).
  • These test kits further comprise containers with tools useful for collecting test samples (such as, for example, blood, urine, saliva and stool).
  • tools include lancets and absorbent paper or cloth for collecting and stabilizing blood; swabs for collecting and stabilizing saliva; and cups for collecting and stabilizing urine or stool samples.
  • Collection materials such as, papers, cloths, swabs, cups, and the like, may optionally be treated to avoid denaturation or irreversible adsorption of the sample.
  • the collection materials also may be treated with or contain preservatives, stabilizers or antimicrobial agents to help maintain the integrity of the specimens.
  • the present invention also provides a purified polynucleotide or fragment thereof derived from a PS213 gene.
  • the purified polynucleotide encodes a polypeptide having at least 50% identity with a polypeptide having an amino acid sequence selected from the group consisting of SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
  • the purified polynucleotide can be produced by recombinant and/or synthetic techniques.
  • the purified recombinant polynucleotide can be contained within a recombinant vector.
  • the invention further comprises a host cell transfected with the recombinant vector.
  • the present invention further provides a recombinant expression system comprising a nucleic acid sequence that includes an open reading frame derived from PS213.
  • the nucleic acid sequence is operably linked to a control sequence compatible with a desired host.
  • a cell transfected with this recombinant expression system is also provided.
  • the present invention also provides a polypeptide encoded by PS213.
  • the polypeptide can be produced by recombinant technology, provided in purified form, or produced by synthetic techniques.
  • the polypeptide comprises an amino acid sequence which has at least 50% identity with an amino acid sequence selected from the group consisting of SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
  • a specific binding molecule which specifically binds to at least one PS213 epitope.
  • the specific binding molecule can be an antibody.
  • the epitope is derived from an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
  • test kits for determining the presence of PS213 antigen or anti-PS213 antibody in a test sample are also included.
  • the test kits comprise a container containing at least one PS213 polypeptide having at least 50% identity with an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8,
  • the test kit can comprise a container with tools useful for collecting test samples (such as blood, urine, saliva, and stool).
  • tools include lancets and absorbent paper or cloth for collecting and stabilizing blood; swabs for collecting and stabilizing saliva; and cups for collecting and stabilizing urine or stool samples.
  • Collection materials such as papers, cloths, swabs, cups, and the like, may optionally be treated to avoid denaturation or irreversible adsorption of the sample. These collection materials also may be treated with or contain preservatives, stabilizers or antimicrobiol agents to help maintain the integrity of the specimens.
  • the polypeptide can be attached to a solid phase.
  • specific binding molecules such as antibodies or fragments thereof against the PS213 antigen
  • Such antibodies can be polyclonal or monoclonal, or made by molecular biology techniques, and can be labeled with a variety of detectable labels, including but not limited to radioisotopes and paramagnetic metals.
  • antibodies or fragments thereof whether monoclonal, polyclonal, or made by molecular biology techniques, can be used as therapeutic agents for the treatment of diseases characterized by expression of the PS213 antigen.
  • the antibody may be used without derivitization, or it may be derivitized with a cytotoxic agent such as a radioisotope, enzyme, toxin, drug, prodrug, or the like.
  • test kit for determining the presence of PS213 antigen or anti- PS213 antibody in a test sample comprises a container containing a specific binding molecule, such as an antibody, which specifically binds to a PS213 antigen, wherein the PS213 antigen comprises at least one PS213-encoded epitope.
  • the PS213 antigen has at least about 50% identity to a sequence of a PS213- encoded antigen selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
  • These test kits can further comprise containers with tools useful for collecting test samples (such as blood, urine, saliva, and stool).
  • Such tools include lancets and absorbent paper or cloth for collecting and stabilizing blood; swabs for collecting and stabilizing saliva; cups for collecting and stabilizing urine or stool samples.
  • Collection materials such as papers, cloths, swabs, cups and the like, may optionally be treated to avoid denaturation or irreversible adsorption of the sample. These collection materials also may be treated with, or contain, preservatives, stabilizers or antimicrobial agents to help maintain the integrity of the specimens.
  • the antibody can be attached to a solid phase.
  • a method for producing a polypeptide which contains at least one epitope of PS213 comprises incubating host cells transfected with an expression vector.
  • This vector comprises a polynucleotide sequence encoding a polypeptide, wherein the polypeptide comprises an amino acid sequence having at least 50% identity with a PS213 amino acid sequence selected from the group consisting of SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
  • a method for detecting PS213 antigen in a test sample suspected of containing PS213 antigen comprises contacting the test sample with a specific binding molecule, such as an antibody or fragment thereof, which specifically binds to at least one epitope of a PS213 antigen, for a time and under conditions sufficient for the formation of binding molecule/antigen complexes; and detecting the presence of such complexes as an indication of the presence of PS213 antigen in the test sample.
  • the specific binding molecule can be attached to a solid phase antibody.
  • the specific binding molecule specifically binds to at least one epitope of a PS213 antigen selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
  • Another method which detects antibodies which specifically bind to PS213 antigen in a test sample suspected of containing these antibodies. The method comprises contacting the test sample with a polypeptide which contains at least one PS213 epitope, wherein the PS213 epitope comprises an amino acid sequence having at least 50% identity with an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments.
  • the method further entails detecting complexes which contain the polypeptide.
  • the polypeptide can be attached to a solid phase. Further, the polypeptide can be a recombinant protein or a synthetic peptide.
  • the present invention provides a cell transfected with a PS213 polynucleotide as described above.
  • a method for producing antibodies to PS213 antigen comprises administering to an individual an isolated immunogenic polypeptide or fragment thereof, wherein the isolated immunogenic polypeptide comprises at least one PS213 epitope.
  • the immunogenic polypeptide is administered in an amount sufficient to produce an immune response.
  • the isolated, immunogenic polypeptide comprises an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
  • Another method for producing antibodies which specifically bind to PS213 antigen comprises administering to an individual a plasmid comprising a nucleic acid sequence which encodes at least one PS213 epitope derived from an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
  • the plasmid is administered in an amount such that the plasmid is taken up by cells in the individual and expressed at levels sufficient to produce an immune response.
  • Figure 1 shows the results of the Western blot performed on a panel of tissue protein extracts probed with antiserum against a PS213 synthetic peptide
  • the present invention provides methods for assaying a test sample for products of a prostate tissue gene designated as PS213, which comprises making cDNA from mRNA in the test sample, and detecting the cDNA as an indication of the presence of prostate tissue gene PS213.
  • the method may include an amplification step, wherein one or more portions of the mRNA from PS213 corresponding to the gene or fragments thereof, is amplified.
  • Methods also are provided for assaying for the translation products of PS213.
  • Test samples which may be assayed by the methods provided herein include tissues, cells, body fluids and secretions.
  • the present invention also provides reagents such as oligonucleotide primers and polypeptides which are useful in performing these methods.
  • nucleic acid sequences disclosed herein are useful as primers for the reverse transcription of RNA or for the amplification of cDNA; or as probes to determine the presence of certain mRNA sequences in test samples. Also disclosed are nucleic acid sequences which permit the production of encoded polypeptide sequences which are useful as standards or reagents in diagnostic immunoassays, as targets for pharmaceutical screening assays and/or as components or as target sites for various therapies. Monoclonal and polyclonal antibodies directed against at least one epitope contained within these polypeptide sequences are useful as delivery agents for therapeutic agents as well as for diagnostic tests and for screening for diseases or conditions associated with PS213, especially prostate cancer.
  • Isolation of sequences of other portions of the gene of interest can be accomplished utilizing probes or PCR primers derived from these nucleic acid sequences. This allows additional probes of the mRNA or cDNA of interest to be established, as well as corresponding encoded polypeptide sequences. These additional molecules are useful in detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining the predisposition to diseases and conditions of the prostate, such as prostate cancer, characterized by PS213, as disclosed herein.
  • similarity means the exact amino acid to amino acid comparison of two or more polypeptides at the appropriate place, where amino acids are identical or possess similar chemical and/or physical properties such as charge or hydrophobicity. "Percent similarity” then can be determined between the compared polypeptide sequences.
  • Techniques for determining nucleic acid and amino acid sequence identity also are well known in the art and include determining the nucleotide sequence of the mRNA for that gene (usually via a cDNA intermediate) and determining the amino acid sequence encoded thereby, and comparing this to a second amino acid sequence.
  • identity refers to an exact nucleotide to nucleotide or amino acid to amino acid correspondence of two polynucleotides or polypeptide sequences, respectively.
  • Two or more polynucleotide sequences can be compared by determining their "percent identity.”
  • Two or more amino acid sequences likewise can be compared by determining their "percent identity.”
  • the percent identity of two sequences, whether nucleic acid or peptide sequences is the number of exact matches between two aligned sequences divided by the length of the shorter sequences and multiplied by 100.
  • An approximate alignment for nucleic acid sequences is provided by the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482-489 (1981).
  • compositions and methods described herein will enable the identification of certain markers as indicative of a prostate tissue disease or condition; the information obtained therefrom will aid in the detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining diseases or conditions associated with PS213, especially prostate cancer.
  • Test methods include, for example, probe assays which utilize the sequence(s) provided herein and which also may utilize nucleic acid amplification methods such as the polymerase chain reaction (PCR), the ligase chain reaction (LCR), and hybridization.
  • PCR polymerase chain reaction
  • LCR ligase chain reaction
  • nucleotide sequences provided herein contain open reading frames from which an immunogenic epitope may be found.
  • This epitope is believed to be unique to the disease state or condition associated with PS213. It also is thought that the polynucleotides or polypeptides and protein encoded by the PS213 gene are useful as a marker. This marker is either elevated in disease such as prostate cancer, altered in disease such as prostate cancer, or present as a normal protein but appearing in an inappropriate body compartment.
  • the uniqueness of the epitope may be determined by (i) its immunological reactivity and specificity with antibodies directed against proteins and polypeptides encoded by the PS213 gene, and (ii) its nonreactivity with any other tissue markers.
  • R A radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • HA hemagglutination
  • FPIA fluorescence polarization immunoassay
  • CLIA chemiluminescent immunoassay
  • a polynucleotide "derived from” or “specific for” a designated sequence refers to a polynucleotide sequence which comprises a contiguous sequence of approximately at least about 6 nucleotides, preferably at least about 8 nucleotides, more preferably at least about 10-12 nucleotides, and even more preferably at least about 15-20 nucleotides corresponding, i.e., identical or complementary to, a region of the designated nucleotide sequence.
  • the sequence may be complementary or identical to a sequence which is unique to a particular polynucleotide sequence as determined by techniques known in the art.
  • Comparisons to sequences in databanks can be used as a method to determine the uniqueness of a designated sequence.
  • Regions from which sequences may be derived include but are not limited to, regions encoding specific epitopes, as well as non-translated and/or non-transcribed regions.
  • the derived polynucleotide will not necessarily be derived physically from the nucleotide sequence of interest under study, but may be generated in any manner, including, but not limited to, chemical synthesis, replication, reverse transcription or transcription, which is based on the information provided by the sequence of bases in the region(s) from which the polynucleotide is derived. As such, it may represent either a sense or an antisense orientation of the original polynucleotide.
  • combinations of regions corresponding to that of the designated sequence may be modified in ways known in the art to be consistent with the intended use.
  • a “fragment" of a specified polynucleotide refers to a polynucleotide sequence which comprises a contiguous sequence of approximately at least about 6 nucleotides, preferably at least about 8 nucleotides, more preferably at least about 10-12 nucleotides, and even more preferably at least about 15-20 nucleotides corresponding, i.e., identical or complementary to, a region of the specified nucleotide sequence.
  • the term "primer” denotes a specific oligonucleotide sequence which is complementary to a target nucleotide sequence and used to hybridize to the target nucleotide sequence.
  • a primer serves as an initiation point for nucleotide polymerization catalyzed by either DNA polymerase, RNA polymerase or reverse transcriptase.
  • probe denotes a defined nucleic acid segment (or nucleotide analog segment, e.g., PNA as defined hereinbelow) which can be used to identify a specific polynucleotide present in samples bearing the complementary sequence.
  • Encoded by refers to a nucleic acid sequence which codes for a polypeptide sequence, wherein the polypeptide sequence or a portion thereof contains an amino acid sequence of at least 3 to 5 amino acids, more preferably at least 8 to 10 amino acids, and even more preferably at least 15 to 20 amino acids from a polypeptide encoded by the nucleic acid sequence. Also encompassed are polypeptide sequences which are immunologically identifiable with a polypeptide encoded by the sequence. Thus, a "polypeptide,” “protein,” or “amino acid” sequence has at least about 50% identity, preferably about 60% identity, more preferably about 75-85% identity, and most preferably about 90-95% or more identity with a PS213 amino acid sequence.
  • the PS213 "polypeptide,” “protein,” or “amino acid” sequence may have at least about 60% similarity, preferably at least about 75% similarity, more preferably about 85% similarity, and most preferably about 95% or more similarity to a polypeptide or amino acid sequence of PS213.
  • This amino acid sequence can be selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
  • a recombinant or encoded polypeptide or protein is not necessarily translated from a designated nucleic acid sequence. It also may be generated in any manner, including chemical synthesis or expression of a recombinant expression system.
  • synthetic peptide as used herein means a polymeric form of amino acids of any length, which may be chemically synthesized by methods well- known to the routineer. These synthetic peptides are useful in various applications.
  • polynucleotide as used herein means a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides. This term refers only to the primary structure of the molecule. Thus, the term includes double- and single-stranded DNA, as well as double- and single-stranded RNA. It also includes modifications, such as methylation or capping and unmodified forms of the polynucleotide.
  • polynucleotide oligomer
  • oligonucleotide and “oligo” are used interchangeably herein.
  • a sequence corresponding to a cDNA means that the sequence contains a polynucleotide sequence that is identical or complementary to a sequence in the designated DNA.
  • the degree (or “percent") of identity or complementarity to the cDNA will be approximately 50% or greater, preferably at least about 70% or greater, and more preferably at least about 90% or greater.
  • the sequence that corresponds to the identified cDNA will be at least about 50 nucleotides in length, preferably at least about 60 nucleotides in length, and more preferably at least about 70 nucleotides in length.
  • the correspondence between the gene or gene fragment of interest and the cDNA can be determined by methods known in the art and include, for example, a direct comparison of the sequenced material with the cDNAs described, or hybridization and digestion with single strand nucleases, followed by size determination of the digested fragments.
  • Polynucleotide refers to a polynucleotide of interest or fragment thereof which is essentially free, e.g., contains less than about 50%, preferably less than about 70%, and more preferably less than about 90%, of the protein with which the polynucleotide is naturally associated.
  • Techniques for purifying polynucleotides of interest include, for example, disruption of the cell containing the polynucleotide with a chaotropic agent and separation of the polynucleotide(s) and proteins by ion-exchange chromatography, affinity chromatography and sedimentation according to density.
  • Polypeptide or “purified protein” means a polypeptide of interest or fragment thereof which is essentially free of, e.g., contains less than about 50%, preferably less than about 70%, and more preferably less than about 90%, cellular components with which the polypeptide of interest is naturally associated. Methods for purifying polypeptides of interest are known in the art.
  • isolated means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring).
  • a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or DNA or polypeptide, which is separated from some or all of the coexisting materials in the natural system, is isolated.
  • Such polynucleotide could be part of a vector and/or such polynucleotide or polypeptide could be part of a composition, and still be isolated in that the vector or composition is not part of its natural environment.
  • Polypeptide and “protein” are used interchangeably herein and indicate at least one molecular chain of amino acids linked through covalent and or non- covalent bonds. The terms do not refer to a specific length of the product. Thus peptides, oligopeptides and proteins are included within the definition of polypeptide. The terms include post-translational modifications of the polypeptide, for example, glycosylations, acetylations, phosphorylations and the like. In addition, protein fragments, analogs, mutated or variant proteins, fusion proteins and the like are included within the meaning of polypeptide.
  • a “fragment" of a specified polypeptide refers to an amino acid sequence which comprises at least about 3-5 amino acids, more preferably at least about 8-10 amino acids, and even more preferably at least about 15-20 amino acids derived from the specified polypeptide.
  • Recombinant host cells refer to cells which can be, or have been, used as recipients for recombinant vector or other transferred DNA, and include the original progeny of the original cell which has been transfected.
  • replicon means any genetic element, such as a plasmid, a chromosome or a virus, that behaves as an autonomous unit of polynucleotide replication within a cell.
  • a “vector” is a replicon in which another polynucleotide segment is attached, such as to bring about the replication and/or expression of the attached segment.
  • control sequence refers to a polynucleotide sequence which is necessary to effect the expression of a coding sequence to which it is ligated. The nature of such control sequences differs depending upon the host organism. In prokaryotes, such control sequences generally include a promoter, a ribosomal binding site and terminators; in eukaryotes, such control sequences generally include promoters, terminators and, in some instances, enhancers.
  • control sequence thus is intended to include at a minimum all components whose presence is necessary for expression, and also may include additional components whose presence is advantageous, for example, leader sequences.
  • operably linked refers to a situation wherein the components described are in a relationship permitting them to function in their intended manner.
  • a control sequence "operably linked" to a coding sequence is ligated in such a manner that expression of the coding sequence is achieved under conditions compatible with the control sequence.
  • open reading frame refers to a region of a polynucleotide sequence which encodes a polypeptide. This region may represent a portion of a coding sequence or a total coding sequence.
  • a "coding sequence” is a polynucleotide sequence which is transcribed into mRNA and translated into a polypeptide when placed under the control of appropriate regulatory sequences. The boundaries of the coding sequence are determined by a translation start codon at the 5' -terminus and a translation stop codon at the 3' -terminus.
  • a coding sequence can include, but is not limited to, mRNA, cDNA and recombinant polynucleotide sequences.
  • immunologically identifiable with/as refers to the presence of epitope(s) and polypeptide(s) which also are present in and are unique to the designated polypeptide(s). Immunological identity may be determined by antibody binding and/or competition in binding. These techniques are known to the routineer and also are described herein. The uniqueness of an epitope also can be determined by computer searches of known data banks, such as GenBank, for the polynucleotide sequence which encodes the epitope and by amino acid sequence comparisons with other known proteins.
  • epitope means an antigenic determinant of a polypeptide or protein.
  • an epitope can comprise three amino acids in a spatial conformation which is unique to the epitope.
  • an epitope consists of at least five such amino acids and more usually, it consists of at least eight to ten amino acids.
  • Methods of examining spatial conformation include, for example, x-ray crystallography and two-dimensional nuclear magnetic resonance.
  • a “conformational epitope” is an epitope that is comprised of a specific juxtaposition of amino acids in an immunologically recognizable structure, such amino acids being present on the same polypeptide in a contiguous or noncontiguous order or present on different polypeptides.
  • a polypeptide is "immunologically reactive" with an antibody when it binds to an antibody due to antibody recognition of a specific epitope contained within the polypeptide. Immunological reactivity may be determined by antibody binding, more particularly, by the kinetics of antibody binding, and/or by competition in binding using as competitor(s) a known polypeptide(s) containing an epitope against which the antibody is directed. The methods for determining whether a polypeptide is immunologically reactive with an antibody are known in the art.
  • the term "immunogenic polypeptide containing an epitope of interest” means naturally occurring polypeptides of interest or fragments thereof, as well as polypeptides prepared by other means, for example, by chemical synthesis or the expression of the polypeptide in a recombinant organism.
  • transfection refers to the introduction of an exogenous polynucleotide into a prokaryotic or eucaryotic host cell, irrespective of the method used for the introduction.
  • the term “transfection” refers to both stable and transient introduction of the polynucleotide, and encompasses direct uptake of polynucleotides, transformation, transduction, and f-mating.
  • the exogenous polynucleotide may be maintained as a non-integrated replicon, for example, a plasmid, or alternatively, may be integrated into the host genome.
  • Treatment refers to prophylaxis and/or therapy.
  • the term "individual” as used herein refers to vertebrates, particularly members of the mammalian species and includes, but is not limited to, domestic animals, sports animals, primates and humans; more particularly, the term refers to humans.
  • sense strand or "plus strand” (or “+) as used herein denotes a nucleic acid that contains the sequence that encodes the polypeptide.
  • antisense strand or "minus strand” (or “-) denotes a nucleic acid that contains a sequence that is complementary to that of the "plus” strand.
  • test sample refers to a component of an individual's body which is the source of the analyte (such as antibodies of interest or antigens of interest). These components are well known in the art.
  • a test sample is typically anything suspected of containing a target sequence.
  • Test samples can be prepared using methodologies well known in the art such as by obtaining a specimen from an individual and, if necessary, disrupting any cells contained thereby to release target nucleic acids.
  • test samples include biological samples which can be tested by the methods of the present invention described herein and include human and animal body fluids such as whole blood, serum, plasma, cerebrospinal fluid, sputum, bronchial washing, bronchial aspirates, urine, lymph fluids, and various external secretions of the respiratory, intestinal and genitourinary tracts, tears, saliva, milk, white blood cells, myelomas and the like; biological fluids such as cell culture supernatants; tissue specimens which may be fixed; and cell specimens which may be fixed.
  • human and animal body fluids such as whole blood, serum, plasma, cerebrospinal fluid, sputum, bronchial washing, bronchial aspirates, urine, lymph fluids, and various external secretions of the respiratory, intestinal and genitourinary tracts, tears, saliva, milk, white blood cells, myelomas and the like
  • biological fluids such as cell culture supernatants
  • tissue specimens which may be fixed and cell specimens which may be fixed.
  • PNA polypeptide nucleic acid analog
  • MA morpholino analog
  • PNA probes used in assays in place of, for example, the DNA probes of the present invention offer advantages not achievable when DNA probes are used. These advantages include manufacturability, large scale labeling, reproducibility, stability, insensitivity to changes in ionic strength and resistance to enzymatic degradation which is present in methods utilizing DNA or RNA. These PNAs can be labeled with ("attached to") such signal generating compounds as fluorescein, radionucleotides, chemiluminescent compounds and the like. PNAs or other nucleic acid analogs such as MAs thus can be used in assay methods in place of DNA or RNA. Although assays are described herein utilizing DNA probes, it is within the scope of the routineer that PNAs or MAs can be substituted for RNA or DNA with appropriate changes if and as needed in assay reagents.
  • analyte is the substance to be detected which may be present in the test sample.
  • the analyte can be any substance for which there exists a naturally occurring specific binding member (such as an antibody), or for which a specific binding member can be prepared.
  • an analyte is a substance that can bind to one or more specific binding members in an assay.
  • “Analyte” also includes any antigenic substances, haptens, antibodies and combinations thereof.
  • the analyte can be detected by means of naturally occurring specific binding partners (pairs) such as the use of intrinsic factor protein as a member of a specific binding pair for the determination of Vitamin B12, the use of folate-binding protein to determine folic acid, or the use of a lectin as a member of a specific binding pair for the determination of a carbohydrate.
  • the analyte can include a protein, a polypeptide, an amino acid, a nucleotide target and the like.
  • the analyte can be soluble in a body fluid such as blood, blood plasma or serum, urine or the like.
  • the analyte can be in a tissue, either on a cell surface or within a cell.
  • the analyte can be on or in a cell dispersed in a body fluid such as blood, urine, breast aspirate, or obtained as a biopsy sample.
  • disease of the prostate refers to any disease or condition of the prostate including, but not limited to, benign prostatic hyperplasia (BPH), prostatitis, prostatic intraepithelial neoplasia (PIN) and cancer.
  • Prostate cancer refers to any malignant disease of the prostate including, but not limited to, adenocarcinoma, small cell undifferentiated carcinoma and mucinous (colloid) cancer.
  • An "Expressed Sequence Tag” or “EST” refers to the partial sequence of a cDNA insert which has been made by reverse transcription of mRNA extracted from a tissue followed by insertion into a vector.
  • a “transcript image” refers to a table or list giving the quantitative distribution of ESTs in a library and represents the genes active in the tissue from which the library was made.
  • the present invention provides assays which utilize specific binding members.
  • a "specific binding member,” as used herein, is a member of a specific binding pair. That is, two different molecules where one of the molecules, through chemical or physical means, specifically binds to the second molecule. Therefore, in addition to antigen and antibody specific binding pairs of common immunoassays, other specific binding pairs can include biotin and avidin, carbohydrates and lectins, complementary nucleotide sequences, effector and receptor molecules, cofactors and enzymes, enzyme inhibitors, and enzymes and the like. Furthermore, specific binding pairs can include members that are analogs of the original specific binding members, for example, an analyte-analog.
  • Immunoreactive specific binding members include antigens, antigen fragments, antibodies and antibody fragments, both monoclonal and polyclonal and complexes thereof, including those formed by recombinant DNA molecules.
  • Specific binding members include “specific binding molecules.”
  • a “specific binding molecule” intends any specific binding member, particularly an immunoreactive specific binding member.
  • the term “specific binding molecule” encompasses antibody molecules (obtained from both polyclonal and monoclonal preparations), as well as, the following: hybrid (chimeric) antibody molecules (see, for example, Winter, et al, Nature 349:293-299 (1991), and U.S. Patent No. 4,816,567); F(ab') 2 and F(ab) fragments; Fv molecules [non-covalent heterodimers, see, for example, Inbar, et al., Proc. Natl. Acad. Sci.
  • hapten refers to a partial antigen or non-protein binding member which is capable of binding to an antibody, but which is not capable of eliciting antibody formation unless coupled to a carrier protein.
  • a “capture reagent,” as used herein, refers to an unlabeled specific binding member which is specific either for the analyte as in a sandwich assay, for the indicator reagent or analyte as in a competitive assay, or for an ancillary specific binding member, which itself is specific for the analyte, as in an indirect assay.
  • the capture reagent can be directly or indirectly bound to a solid phase material before the performance of the assay or during the performance of the assay, thereby enabling the separation of immobilized complexes from the test sample.
  • the “indicator reagent” comprises a “signal-generating compound” ("label”) which is capable of generating and generates a measurable signal detectable by external means, conjugated (“attached") to a specific binding member.
  • label a “signal-generating compound”
  • the indicator reagent also can be a member of any specific binding pair, including either hapten-anti-hapten systems such as biotin or anti-biotin, avidin or biotin, a carbohydrate or a lectin, a complementary nucleotide sequence, an effector or a receptor molecule, an enzyme cofactor and an enzyme, an enzyme inhibitor or an enzyme, and the like.
  • An immunoreactive specific binding member can be an antibody, an antigen, or an antibody/antigen complex that is capable of binding either to the polypeptide of interest as in a sandwich assay, to the capture reagent as in a competitive assay, or to the ancillary specific binding member as in an indirect assay.
  • reporter molecule comprises a signal generating compound as described hereinabove conjugated to a specific binding member of a specific binding pair, such as carbazole or adamantane.
  • labels include chromagens, catalysts such as enzymes, luminescent compounds such as fluorescein and rhodamine, chemiluminescent compounds such as dioxetanes, acridiniums, phenanthridiniums and luminol, radioactive elements and direct visual labels.
  • luminescent compounds such as fluorescein and rhodamine
  • chemiluminescent compounds such as dioxetanes, acridiniums, phenanthridiniums and luminol
  • radioactive elements include direct visual labels.
  • enzymes include alkaline phosphatase, horseradish peroxidase, beta-galactosidase and the like.
  • the selection of a particular label is not critical, but it must be capable of producing a signal either by itself or in conjunction with one or more additional substances.
  • Solid phases are known to those in the art and include the walls of wells of a reaction tray, test tubes, polystyrene beads, magnetic or non-magnetic beads, nitrocellulose strips, membranes, microparticles such as latex particles, sheep (or other animal) red blood cells and Duracytes ® (red blood cells “fixed” by pyruvic aldehyde and formaldehyde, available from Abbott Laboratories, Abbott Park, IL) and others.
  • the “solid phase” is not critical and can be selected by one skilled in the art.
  • solid phases include ionic, hydrophobic, covalent interactions and the like.
  • a "solid phase,” as used herein, refers to any material which is insoluble, or can be made insoluble by a subsequent reaction.
  • the solid phase can be chosen for its intrinsic ability to attract and immobilize the capture reagent. Alternatively, the solid phase can retain an additional receptor which has the ability to attract and immobilize the capture reagent.
  • the additional receptor can include a charged substance that is oppositely charged with respect to the capture reagent itself or to a charged substance conjugated to the capture reagent.
  • the receptor molecule can be any specific binding member which is immobilized upon (attached to) the solid phase and which has the ability to immobilize the capture reagent through a specific binding reaction. The receptor molecule enables the indirect binding of the capture reagent to a solid phase material before the performance of the assay or during the performance of the assay.
  • the solid phase thus can be a plastic, derivatized plastic, magnetic or non-magnetic metal, glass or silicon surface of a test tube, microtiter well, sheet, bead, microparticle, chip, sheep (or other suitable animal's) red blood cells, Duracytes ® and other configurations known to those of ordinary skill in the art.
  • the solid phase also can comprise any suitable porous material with sufficient porosity to allow access by detection antibodies and a suitable surface affinity to bind antigens.
  • Microporous structures generally are preferred, but materials with a gel structure in the hydrated state may be used as well.
  • Such useful solid supports include, but are not limited to, nitrocellulose and nylon. It is contemplated that such porous solid supports described herein preferably are in the form of sheets of thickness from about 0.01 to 0.5 mm, preferably about 0.1 mm.
  • the pore size may vary within wide limits and preferably is from about 0.025 to 15 microns, especially from about 0.15 to 15 microns.
  • Such supports may be activated by chemical processes which cause covalent linkage of the antigen or antibody to the support.
  • the irreversible binding of the antigen or antibody is obtained, however, in general, by adsorption on the porous material by poorly understood hydrophobic forces.
  • Other suitable solid supports are known in the art.
  • the present invention provides reagents such as polynucleotide sequences derived from a prostate tissue of interest and designated as PS213, polypeptides encoded thereby and antibodies specific for these polypeptides.
  • the present invention also provides reagents such as oligonucleotide fragments derived from the disclosed polynucleotides and nucleic acid sequences complementary to these polynucleotides.
  • the polynucleotides, polypeptides, or antibodies of the present invention may be used to provide information leading to the detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating of, or determining the predisposition to, diseases and conditions of the prostate, such as prostate cancer.
  • the sequences disclosed herein represent unique polynucleotides which can be used in assays or for producing a specific profile of gene transcription activity. Such assays are disclosed in European Patent Number 0373203B1 and International Publication No. WO 95/1 1995.
  • PS213-derived polynucleotides can be used in the methods described herein for the detection of normal or altered gene expression. Such methods may employ PS213 polynucleotides or oligonucleotides, fragments or derivatives thereof, or nucleic acid sequences complementary thereto.
  • polynucleotides disclosed herein, their complementary sequences, or fragments of either, can be used in assays to detect, amplify or quantify genes, nucleic acids, cDNAs or mRNAs relating to prostate tissue disease and conditions associated therewith. They also can be used to identify an entire or partial coding region of a PS213 polypeptide. They further can be provided in individual containers in the form of a kit for assays, or provided as individual compositions. If provided in a kit for assays, other suitable reagents such as buffers, conjugates and the like may be included.
  • the polynucleotide may be in the form of RNA or DNA.
  • Polynucleotides in the form of DNA, cDNA, genomic DNA, nucleic acid analogs and synthetic DNA are within the scope of the present invention.
  • the DNA may be double- stranded or single-stranded, and if single stranded, may be the coding (sense) strand or non-coding (anti-sense) strand.
  • the coding sequence which encodes the polypeptide may be identical to the coding sequence provided herein or may be a different coding sequence which coding sequence, as a result of the redundancy or degeneracy of the genetic code, encodes the same polypeptide as the DNA provided herein.
  • This polynucleotide may include only the coding sequence for the polypeptide, or the coding sequence for the polypeptide and an additional coding sequence such as a leader or secretory sequence or a proprotein sequence, or the coding sequence for the polypeptide (and optionally an additional coding sequence) and non-coding sequence, such as a non-coding sequence 5' and/or 3' of the coding sequence for the polypeptide.
  • the invention includes variant polynucleotides containing modifications such as polynucleotide deletions, substitutions or additions; and any polypeptide modification resulting from the variant polynucleotide sequence.
  • a polynucleotide of the present invention also may have a coding sequence which is a naturally occurring allelic variant of the coding sequence provided herein.
  • the coding sequence for the polypeptide may be fused in the same reading frame to a polynucleotide sequence which aids in expression and secretion of a polypeptide from a host cell, for example, a leader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell.
  • the polypeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cell to form the polypeptide.
  • the polynucleotides may also encode for a proprotein which is the protein plus additional 5' amino acid residues.
  • a protein having a prosequence is a proprotein and may, in some cases, be an inactive form of the protein. Once the prosequence is cleaved, an active protein remains.
  • the polynucleotide of the present invention may encode for a protein, or for a protein having a prosequence, or for a protein having both a presequence (leader sequence) and a prosequence.
  • the polynucleotides of the present invention may also have the coding sequence fused in frame to a marker sequence which allows for purification of the polypeptide of the present invention.
  • the marker sequence may be a hexa-histidine tag supplied by a pQE-9 vector to provide for purification of the polypeptide fused to the marker in the case of a bacterial host, or, for example, the marker sequence may be a hemagglutinin (HA) tag when a mammalian host, e.g. a COS-7 cell line, is used.
  • the HA tag corresponds to an epitope derived from the influenza hemagglutinin protein. See, for example, I. Wilson et al., Cell 37:767 (1984).
  • polynucleotides will be considered to hybridize to the sequences provided herein if there is at least 50%, preferably at least 70%, and more preferably at least 90% identity between the polynucleotide and the sequence.
  • the degree of sequence identity between two nucleic acid molecules greatly affects the efficiency and strength of hybridization events between such molecules.
  • a partially identical nucleic acid sequence is one that will at least partially inhibit a completely identical sequence from hybridizing to a target molecule.
  • Inhibition of hybridization of the completely identical sequence can be assessed using hybridization assays that are well known in the art (e.g., Southern blot, Northern blot, solution hybridization, in situ hybridization, or the like, see Sambrook, et al., Molecular Cloning: A Laboratory Manual. Second Edition, (1989) Cold Spring Harbor, N.Y.).
  • Such assays can be conducted using varying degrees of selectivity, for example, using conditions varying from low to high stringency.
  • a nucleic acid probe is chosen that is complementary to a target nucleic acid sequence, and then by selection of appropriate conditions the probe and the target sequence "selectively hybridize," or bind, to each other to form a hybrid molecule.
  • a nucleic acid molecule is capable of hybridizing selectively to a target sequence under moderately stringent hybridization conditions.
  • moderately stringent hybridization conditions allow detection of a target nucleic acid sequence of at least 14 nucleotides in length having at least approximately 70% sequence identity with the sequence of the selected nucleic acid probe.
  • selective hybridization is performed under stringent hybridization conditions.
  • Stringent hybridization conditions allow detection of target nucleic acid sequences of at least 14 nucleotides in length having a sequence identity of greater than 90% with the sequence of the selected nucleic acid probe.
  • Hybridization conditions useful for probe/target hybridization where the probe and target have a specific degree of sequence identity can be determined as is known in the art (see, for example, Nucleic Acid Hybridization: A Practical Approach, editors B.D. Hames and S.J.
  • Hybrid molecules can be formed, for example, on a solid support, in solution, and in tissue sections. The formation of hybrids can be monitored by inclusion of a reporter molecule, typically, in the probe.
  • reporter molecules, or detectable elements include, but are not limited to, radioactive elements, fluorescent markers, and molecules to which an enzyme-conjugated ligand can bind.
  • stringency conditions for hybridization it is well known in the art that numerous equivalent conditions can be employed to establish a particular stringency by varying, for example, the following factors: the length and nature of probe and target sequences, base composition of the various sequences, concentrations of salts and other hybridization solution components, the presence or absence of blocking agents in the hybridization solutions (e.g., formamide, dextran sulfate, and polyethylene glycol), hybridization reaction temperature and time parameters, as well as, varying wash conditions.
  • blocking agents in the hybridization solutions e.g., formamide, dextran sulfate, and polyethylene glycol
  • hybridization reaction temperature and time parameters as well as, varying wash conditions.
  • the selection of a particular set of hybridization conditions is well within the skill of the routineer in the art (see, for example, Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition, (1989) Cold Spring Harbor, N.Y.).
  • the present invention also provides an antibody produced by using a purified PS213 polypeptide of which at least a portion of the polypeptide is encoded by a PS213 polynucleotide selected from the polynucleotides provided herein.
  • These antibodies may be used in the methods provided herein for the detection of PS213 antigen in test samples.
  • the presence of PS213 antigen in the test samples is indicative of the presence of a prostate disease or condition.
  • the antibody also may be used for therapeutic purposes, for example, in neutralizing the activity of PS213 polypeptide in conditions associated with altered or abnormal expression.
  • the present invention further relates to a PS213 polypeptide which has the amino acid sequence as provided herein, as well as fragments, analogs and derivatives of such polypeptide.
  • the polypeptide for use with the present invention may be a recombinant polypeptide, a natural purified polypeptide or a synthetic polypeptide.
  • the fragment, derivative or analog of the PS213 polypeptide may be one in which one or more of the amino acid residues is substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code; or it may be one in which one or more of the amino acid residues includes a substituent group; or it may be one in which the polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol); or it may be one in which the additional amino acids are fused to the polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification of the polypeptide or a proprotein sequence.
  • Such fragments, derivatives and analogs are within the scope of the present invention.
  • the polypeptides and polynucleotides of the present invention are provided preferably in an isolated form and preferably purified.
  • a polypeptide of the present invention may have an amino acid sequence that is identical to that of the naturally occurring polypeptide or that is different by minor variations due to one or more amino acid substitutions.
  • the variation may be a "conservative change" typically in the range of about 1 to 5 amino acids, wherein the substituted amino acid has similar structural or chemical properties, e.g., replacement of leucine with isoleucine or threonine with serine.
  • variations may include nonconservative changes, e.g., replacement of a glycine with a tryptophan.
  • Similar minor variations may also include amino acid deletions or insertions, or both.
  • Guidance in determining which and how many amino acid residues may be substituted, inserted or deleted without changing biological or immunological activity may be found using computer programs well known in the art, for example, DNASTAR software (DNASTAR Inc., Madison WI).
  • Probes constructed according to the polynucleotide sequences of the present invention can be used in various assay methods to provide various types of analysis.
  • such probes can be used in fluorescent in situ hybridization (FISH) technology to perform chromosomal analysis, and used to identify cancer- specific structural alterations in the chromosomes, such as deletions or translocations that are visible from chromosome spreads or detectable using PCR- generated and/or allele specific oligonucleotides probes, allele specific amplification or by direct sequencing.
  • FISH fluorescent in situ hybridization
  • Probes also can be labeled with radioisotopes, directly- or indirectly- detectable haptens, or fluorescent molecules, and utilized for in situ hybridization studies to evaluate the mRNA expression of the gene comprising the polynucleotide in tissue specimens or cells.
  • This invention also provides teachings as to the production of the polynucleotides and polypeptides provided herein. Probe Assays
  • the sequences provided herein may be used to produce probes which can be used in assays for the detection of nucleic acids in test samples.
  • the probes may be designed from conserved nucleotide regions of the polynucleotides of interest or from non-conserved nucleotide regions of the polynucleotide of interest. The design of such probes for optimization in assays is within the skill of the routineer. Generally, nucleic acid probes are developed from non-conserved or unique regions when maximum specificity is desired, and nucleic acid probes are developed from conserved regions when assaying for nucleotide regions that are closely related to, for example, different members of a multi-gene family or in related species like mouse and man.
  • PCR polymerase chain reaction
  • target a desired nucleic acid sequence contained in a nucleic acid or mixture thereof.
  • a pair of primers are employed in excess to hybridize to the complementary strands of the target nucleic acid.
  • the primers are each extended by a polymerase using the target nucleic acid as a template.
  • the extension products become target sequences themselves, following dissociation from the original target strand.
  • New primers then are hybridized and extended by a polymerase, and the cycle is repeated to geometrically increase the number of target sequence molecules.
  • PCR is disclosed in U.S. Patents 4,683,195 and 4,683,202.
  • LCR Ligase Chain Reaction
  • probe pairs are used which include two primary (first and second) and two secondary (third and fourth) probes, all of which are employed in molar excess to target.
  • the first probe hybridizes to a first segment of the target strand
  • the second probe hybridizes to a second segment of the target strand, the first and second segments being contiguous so that the primary probes abut one another in 5' phosphate-3' hydroxyl relationship, and so that a ligase can covalently fuse or ligate the two probes into a fused product.
  • a third (secondary) probe can hybridize to a portion of the first probe and a fourth (secondary) probe can hybridize to a portion of the second probe in a similar abutting fashion.
  • the secondary probes also will hybridize to the target complement in the first instance.
  • the third and fourth probes which can be ligated to form a complementary, secondary ligated product. It is important to realize that the ligated products are functionally equivalent to either the target or its complement. By repeated cycles of hybridization and ligation, amplification of the target sequence is achieved. This technique is described more completely in EP-A- 320 308 to K. Backman published June 16, 1989 and EP-A-439 182 to K. Backman et al., published July 31 , 1991.
  • RT-PCR polymerase chain reaction
  • RT-AGLCR asymmetric gap ligase chain reaction
  • Other known amplification methods which can be utilized herein include but are not limited to the so-called "NASBA” or "3SR” technique described by J.C. Guatelli et al., Proc. Natl. Acad. Sci.
  • Detection of PS213 may be accomplished using any suitable detection method, including those detection methods which are currently well known in the art, as well as detection strategies which may evolve later. See, for example, Caskey et al, U.S. Patent No. 5,582,989, Gelfand et al., U.S. Patent No. 5,210,015. Examples of such detection methods include target amplification methods as well as signal amplification technologies. An example of presently known detection methods would include the nucleic acid amplification technologies referred to as PCR, LCR, NASBA, SDA, RCR and TMA. See, for example, Caskey et al., U.S. Patent No. 5,582,989, Gelfand et al., U.S. Patent No.
  • Detection may also be accomplished using signal amplification such as that disclosed in Snitman et al., U.S. Patent No. 5,273,882. While the amplification of target or signal is preferred at present, it is contemplated and within the scope of the present invention that ultrasensitive detection methods which do not require amplification can be utilized herein. Detection, both amplified and non-amplified, may be performed using a variety of heterogeneous and homogeneous detection formats. Examples of heterogeneous detection formats are disclosed in Snitman et al., U.S. Patent No. 5,273,882, Albarella et al in EP-841 14441.9, Urdea et al., U.S. Patent No.
  • the present invention generally comprises the steps of contacting a test sample suspected of containing a target polynucleotide sequence with amplification reaction reagents comprising an amplification primer, and a detection probe that can hybridize with an internal region of the amplicon sequences.
  • Probes and primers employed according to the method provided herein are labeled with capture and detection labels, wherein probes are labeled with one type of label and primers are labeled with another type of label. Additionally, the primers and probes are selected such that the probe sequence has a lower melt temperature than the primer sequences.
  • the amplification reagents, detection reagents and test sample are placed under amplification conditions whereby, in the presence of target sequence, copies of the target sequence (an amplicon) are produced.
  • the amplicon is double stranded because primers are provided to amplify a target sequence and its complementary strand.
  • the double stranded amplicon then is thermally denatured to produce single stranded amplicon members.
  • the mixture is cooled to allow the formation of complexes between the probes and single stranded amplicon members.
  • the probe sequences preferentially bind the single stranded amplicon members.
  • the probe sequences generally are selected to be shorter than the primer sequences and therefore have a lower melt temperature than the primers. Accordingly, the melt temperature of the amplicon produced by the primers should also have a higher melt temperature than the probes.
  • the probes are found to preferentially bind the single stranded amplicon members. Moreover, this preference of probe/single stranded amplicon binding exists even when the primer sequences are added in excess of the probes.
  • the probe/single stranded amplicon member hybrids are formed, they are detected.
  • Standard heterogeneous assay formats are suitable for detecting die hybrids using the detection labels and capture labels present on the primers and probes.
  • the hybrids can be bound to a solid phase reagent by virtue of the capture label and detected by virtue of the detection label.
  • the detection label is directly detectable
  • the presence of the hybrids on the solid phase can be detected by causing the label to produce a detectable signal, if necessary, and detecting the signal.
  • the captured hybrids can be contacted with a conjugate, which generally comprises a binding member attached to a directly detectable label.
  • the conjugate becomes bound to the complexes and the conjugate's presence on the complexes can be detected with the directly detectable label.
  • the presence of the hybrids on the solid phase reagent can be determined.
  • wash steps may be employed to wash away unhybridized amplicon or probe as well as unbound conjugate.
  • the heterogeneous assays can be conveniently performed using a solid phase support that carries an array of nucleic acid molecules.
  • Such arrays are useful for high-throughput and/or multiplexed assay formats.
  • Various methods for forming such arrays from pre-formed nucleic acid molecules, or methods for generating the array using in situ synthesis techniques, are generally known in the art. [See, for example, Dattagupta, et al., EP Publication No. 0 234, 726A3; Southern, U.S. Patent No. 5,700,637; Pirrung, et al., U.S. Patent No. 5,143,854; PCT International Publication No.
  • the target sequence is described as single stranded, it also is contemplated to include the case where the target sequence is actually double stranded but is merely separated from its complement prior to hybridization with the amplification primer sequences. In the case where PCR is employed in this method, the ends of the target sequences are usually known. In cases where LCR or a modification thereof is employed in the preferred method, the entire target sequence is usually known.
  • the target sequence is a nucleic acid sequence such as, for example, RNA or DNA.
  • Amplification reactions typically employ primers to repeatedly generate copies of a target nucleic acid sequence, which target sequence is usually a small region of a much larger nucleic acid sequence.
  • Primers are themselves nucleic acid sequences that are complementary to regions of a target sequence. Under amplification conditions, these primers hybridize or bind to the complementary regions of the target sequence. Copies of the target sequence typically are generated by the process of primer extension and/or ligation which utilizes enzymes with polymerase or ligase activity, separately or in combination, to add nucleotides to the hybridized primers and/or ligate adjacent probe pairs.
  • the nucleotides that are added to the primers or probes, as monomers or preformed oligomers, are also complementary to the target sequence. Once the primers or probes have been sufficiently extended and/or ligated, they are separated from the target sequence, for example, by heating the reaction mixture to a "melt temperature" which is one in which complementary nucleic acid strands dissociate. Thus, a sequence complementary to the target sequence is formed.
  • a new amplification cycle then can take place to further amplify the number of target sequences by separating any double stranded sequences, allowing primers or probes to hybridize to their respective targets, extending and/or Hgating the hybridized primers or probes and re-separating.
  • the complementary sequences that are generated by amplification cycles can serve as templates for primer extension or filling the gap of two probes to further amplify the number of target sequences.
  • a reaction mixture is cycled between 20 and 100 times, more typically, a reaction mixture is cycled between 25 and 50 times.
  • the numbers of cycles can be determined by the routineer. In this manner, multiple copies of the target sequence and its complementary sequence are produced.
  • primers initiate amplification of the target sequence when it is present under amplification conditions.
  • two primers which are complementary to a portion of a target strand and its complement are employed in PCR.
  • four probes two of which are complementary to a target sequence and two of which are similarly complementary to the target's complement, are generally employed.
  • a nucleic acid amplification reaction mixture may also comprise other reagents which are well known and include but are not limited to: enzyme cofactors such as manganese; magnesium; salts; nicotinamide adenine dinucleotide (NAD); and deoxynucleotide triphosphates (dNTPs) such as, for example, deoxyadenine triphosphate, deoxyguanine triphosphate, deoxycytosine triphosphate and deoxythymine triphosphate.
  • enzyme cofactors such as manganese; magnesium; salts; nicotinamide adenine dinucleotide (NAD); and deoxynucleotide triphosphates (dNTPs) such as, for example, deoxyadenine triphosphate, deoxyguanine triphosphate, deoxycytosine triphosphate and deoxythymine triphosphate.
  • Detection probes are generally nucleic acid sequences or uncharged nucleic acid analogs such as, for example, peptide nucleic acids which are disclosed in International Publication No. WO 92/20702; morpholino analogs which are described in U.S. Patents Nos 5,185,444, 5,034,506 and 5,142,047; and the like.
  • the probe is employed to capture or detect the amplicon generated by the amplification reaction.
  • the probe is not involved in amplification of the target sequence and therefore may have to be rendered "non- extendible" in that additional dNTPs cannot be added to the probe.
  • analogs usually are non-extendible and nucleic acid probes can be rendered non-extendible by modifying the 3' end of the probe such that the hydroxyl group is no longer capable of participating in elongation.
  • the 3' end of the probe can be functionalized with the capture or detection label to thereby consume or otherwise block the hydroxyl group.
  • the 3' hydroxyl group simply can be cleaved, replaced or modified.
  • U.S. Patent Application Serial No. 07/049,061 filed April 19, 1993 describes modifications which can be used to render a probe non-extendible.
  • primers to probes are not important.
  • either the probes or primers can be added to the reaction mixture in excess whereby the concentration of one would be greater than the concentration of the other.
  • primers and probes can be employed in equivalent concentrations.
  • the primers are added to the reaction mixture in excess of the probes.
  • primer to probe ratios of, for example, 5: 1 and 20:1, are preferred.
  • the probe sequences are selected such that they have a lower melt temperature than the primer sequences. Hence, the primer sequences are generally longer than the probe sequences.
  • the primer sequences are in the range of between 20 and 50 nucleotides long, more typically in the range of between 20 and 30 nucleotides long.
  • the typical probe is in the range of between 10 and 25 nucleotides long.
  • a primary amine can be attached to a 3' oligo terminus using 3'-Amine- ON CPGTM (Clontech, Palo Alto, CA).
  • a primary amine can be attached to a 5' oligo terminus using Aminomodifier II ® (Clontech).
  • the amines can be reacted to various haptens using conventional activation and linking chemistries.
  • copending applications U.S. Serial Nos. 625,566, filed December 11, 1990 and 630,908, filed December 20, 1990 teach methods for labeling probes at their 5' and 3' termini, respectively.
  • WO 92/10505 published 25 June 1992
  • WO 92/11388 published 9 July 1992
  • a label-phosphoramidite reagent is prepared and used to add the label to the oligonucleotide during its synthesis. See, for example, N.T. Thuong et al., Tet. Letters 29(46):5905-5908 (1988); or J.S.
  • probes are labeled at their 3' and 5' ends.
  • a capture label is attached to the primers or probes and can be a specific binding member which forms a binding pair with the solid phase reagent's specific binding member.
  • the primer or probe itself may serve as the capture label.
  • a solid phase reagent's binding member is a nucleic acid sequence
  • it may be selected such that it binds a complementary portion of the primer or probe to thereby immobilize the primer or probe to the solid phase.
  • the probe itself serves as the binding member
  • the probe will contain a sequence or "tail" that is not complementary to the single stranded amplicon members.
  • the primer itself serves as the capture label
  • at least a portion of the primer will be free to hybridize with a nucleic acid on a solid phase because the probe is selected such that it is not fully complementary to the primer sequence.
  • probe/single stranded amplicon member complexes can be detected using techniques commonly employed to perform heterogeneous immunoassays.
  • detection is performed according to the protocols used by the commercially available Abbott LCx ® instrumentation (Abbott Laboratories, Abbott Park, IL).
  • the primers and probes disclosed herein are useful in typical PCR assays, wherein the test sample is contacted with a pair of primers, amplification is performed, the hybridization probe is added, and detection is performed.
  • Another method provided by the present invention comprises contacting a test sample with a plurality of polynucleotides, wherein at least one polynucleotide is a PS213 molecule as described herein, hybridizing the test sample with the plurality of polynucleotides and detecting hybridization complexes.
  • Hybridization complexes are identified and quantitated to compile a profile which is indicative of prostate tissue disease, such as prostate cancer.
  • Expressed RNA sequences may further be detected by reverse transcription and amplification of the DNA product by procedures well-known in the art, including polymerase chain reaction (PCR). Drug Screening and Gene Therapy.
  • the present invention also encompasses the use of gene therapy methods for the introduction of anti-sense PS213 derived molecules, such as polynucleotides or oligonucleotides of the present invention, into patients with conditions associated with abnormal expression of polynucleotides related to a prostate tissue disease or condition especially prostate cancer.
  • anti-sense PS213 derived molecules such as polynucleotides or oligonucleotides of the present invention
  • these molecules including antisense RNA and DNA fragments and ribozymes, are designed to inhibit the translation of PS213 mRNA, and may be used therapeutically in the treatment of conditions associated with altered or abnormal expression of PS213 polynucleotide.
  • the oligonucleotides described above can be delivered to cells by procedures known in the art such that the anti-sense RNA or DNA may be expressed in vivo to inhibit production of a PS213 polypeptide in the manner described above.
  • Antisense constructs to a PS213 polynucleotide therefore, reverse the action of PS213 transcripts and may be used for treating prostate tissue disease conditions, such as prostate cancer. These antisense constructs may also be used to treat tumor metastases.
  • the present invention also provides a method of screening a plurality of compounds for specific binding to PS213 polypeptide(s), or any fragment thereof, to identify at least one compound which specifically binds the PS213 polypeptide.
  • a method comprises the steps of providing at least one compound; combining the PS213 polypeptide with each compound under suitable conditions for a time sufficient to allow binding; and detecting the PS213 polypeptide binding to each compound.
  • the polypeptide or peptide fragment employed in such a test may either be free in solution, affixed to a solid support, borne on a cell surface or located intracellularly.
  • One method of screening utilizes eukaryotic or prokaryotic host cells which are stably transfected with recombinant nucleic acids which can express the polypeptide or peptide fragment.
  • a drug, compound, or any other agent may be screened against such transfected cells in competitive binding assays. For example, the formation of complexes between a polypeptide and the agent being tested can be measured in either viable or fixed cells.
  • the present invention thus provides methods of screening for drugs, compounds, or any other agent which can be used to treat diseases associated with PS213. These methods comprise contacting the agent with a polypeptide or fragment thereof and assaying for either the presence of a complex between the agent and the polypeptide, or for the presence of a complex between the polypeptide and the cell. In competitive binding assays, the polypeptide typically is labeled. After suitable incubation, free (or uncomplexed) polypeptide or fragment thereof is separated from that present in bound form, and the amount of free or uncomplexed label is used as a measure of the ability of the particular agent to bind to the polypeptide or to interfere with the polypeptide/cell complex.
  • the present invention also encompasses the use of competitive screening assays in which neutralizing antibodies capable of binding polypeptide specifically compete with a test agent for binding to the polypeptide or fragment thereof.
  • the antibodies can be used to detect the presence of any polypeptide in the test sample which shares one or more antigenic determinants with a PS213 polypeptide as provided herein.
  • Another technique for screening provides high throughput screening for compounds having suitable binding affinity to at least one polypeptide of PS213 disclosed herein. Briefly, large numbers of different small peptide test compounds are synthesized on a solid phase, such as plastic pins or some other surface. The peptide test compounds are reacted with polypeptide and washed. Polypeptide thus bound to the solid phase is detected by methods well-known in the art. Purified polypeptide can also be coated directly onto plates for use in the screening techniques described herein. In addition, non-neutralizing antibodies can be used to capture the polypeptide and immobilize it on the solid support. See, for example, EP 84/03564, published on September 13, 1984.
  • the goal of rational drug design is to produce structural analogs of biologically active polypeptides of interest or of the small molecules including agonists, antagonists, or inhibitors with which they interact.
  • Such structural analogs can be used to design drugs which are more active or stable forms of the polypeptide or which enhance or interfere with the function of a polypeptide in vivo.
  • J. Hodgson. Bio/Technology 9: 19-21 (19911.
  • the three-dimensional structure of a polypeptide, or of a polypeptide-inhibitor complex is determined by x-ray crystallography, by computer modeling or, most typically, by a combination of the two approaches.
  • Both the shape and charges of the polypeptide must be ascertained to elucidate the structure and to determine active site(s) of the molecule. Less often, useful information regarding the structure of a polypeptide may be gained by modeling based on the structure of homologous proteins. In both cases, relevant structural information is used to design analogous polypeptide-like molecules or to identify efficient inhibitors.
  • Useful examples of rational drug design may include molecules which have improved activity or stability as shown by S. Braxton et al., Biochemistry 31:7796-7801 (1992), or which act as inhibitors, agonists, or antagonists of native peptides as shown by S.B.P. Athauda et al., J Biochem. (Tokvol 1 13 (6):742-746 (1993).
  • anti-ids anti-idiotypic antibodies
  • the binding site of the anti-id is an analog of the original receptor.
  • the anti-id then can be used to identify and isolate peptides from banks of chemically or biologically produced peptides.
  • the isolated peptides then can act as the pharmacophore (that is, a prototype pharmaceutical drug).
  • a sufficient amount of a recombinant polypeptide of the present invention may be made available to perform analytical studies such as X-ray crystallography.
  • knowledge of the polypeptide amino acid sequence which is derivable from the nucleic acid sequence provided herein will provide guidance to those employing computer modeling techniques in place of, or in addition to, x-ray crystallography.
  • Antibodies specific to a PS213 polypeptide e.g., anti-PS213 antibodies
  • the antibodies may be used in therapy, for example, to treat prostate tissue diseases including prostate cancer and its metastases.
  • Such antibodies can detect the presence or absence of a PS213 polypeptide in a test sample and, therefore, are useful as diagnostic markers for the diagnosis of a prostate tissue disease or condition especially prostate cancer.
  • Such antibodies may also function as a diagnostic marker for prostate tissue disease conditions, such as prostate cancer.
  • the present invention also is directed to antagonists and inhibitors of the PS213 polypeptides.
  • the antagonists and inhibitors are those which inhibit or eliminate the function of the polypeptide.
  • an antagonist may bind to a polypeptide of the present invention and inhibit or eliminate its function.
  • the antagonist for example, could be an antibody against the polypeptide which eliminates the activity of a PS213 polypeptide by binding a PS213 polypeptide, or in some cases the antagonist may be an oligonucleotide.
  • small molecule inhibitors include, but are not limited to, small peptides or peptide-like molecules.
  • the antagonists and inhibitors may be employed as a composition with a pharmaceutically acceptable carrier including, but not limited to. saline, buffered saline, dextrose, water, glycerol. ethanol and combinations thereof.
  • a pharmaceutically acceptable carrier including, but not limited to. saline, buffered saline, dextrose, water, glycerol. ethanol and combinations thereof.
  • Administration of PS213 polypeptide inhibitors is preferably systemic.
  • the present invention also provides an antibody which inhibits the action of such a polypeptide.
  • Antisense technology can be used to reduce gene expression through triple- helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA.
  • the 5' coding portion of the polynucleotide sequence which encodes for the polypeptide of the present invention, is used to design an antisense RNA oligonucleotide of from 10 to 40 base pairs in length.
  • a DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription, thereby preventing transcription and the production of the PS213 polypeptide.
  • triple helix see. for example, Lee et al., Nuc. Acids Res.
  • RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of a mRNA molecule into the PS213 polypeptide.
  • antisense see, for example, Okano, J. Neurochem. 56:560 (1991): and Oligodeoxynucleotides as Antisense Inhibitors of Gene
  • Antisense oligonucleotides act with greater efficacy when modified to contain artificial internucleotide linkages which render the molecule resistant to nucleolytic cleavage.
  • artificial internucleotide linkages include, but are not limited to, methylphosphonate, phosphorothiolate and phosphoroamydate internucleotide linkages.
  • the present invention provides host cells and expression vectors comprising PS213 polynucleotides of the present invention and methods for the production of the polypeptide(s) they encode. Such methods comprise culturing the host cells under conditions suitable for the expression of the PS213 polynucleotide and recovering the PS213 polypeptide from the cell culture.
  • the present invention also provides vectors which include PS213 polynucleotides of the present invention, host cells which are genetically engineered with vectors of the present invention and the production of polypeptides of the present invention by recombinant techniques.
  • Host cells are genetically engineered (transfected, transduced or transformed) with the vectors of this invention which may be cloning vectors or expression vectors.
  • the vector may be in the form of a plasmid, a viral particle, a phage, etc.
  • the engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transfected cells, or amplifying PS213 gene(s).
  • the culture conditions such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
  • the polynucleotides of the present invention may be employed for producing a polypeptide by recombinant techniques.
  • the polynucleotide sequence may be included in any one of a variety of expression vehicles, in particular, vectors or plasmids for expressing a polypeptide.
  • vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus and pseudorabies.
  • any other plasmid or vector may be used so long as it is replicable and viable in the host.
  • the appropriate DNA sequence may be inserted into the vector by a variety of procedures. In general, the DNA sequence is inserted into appropriate restriction endonuclease sites by procedures known in the art. Such procedures and others are deemed to be within the scope of those skilled in the art.
  • the DNA sequence in the expression vector is operatively linked to an appropriate expression control sequence(s) (promoter) to direct mRNA synthesis.
  • promoters include, but are not limited to, the LTR or the SV40 promoter, the I coji lac or tip, the phage lambda P sub L promoter and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses.
  • the expression vector also contains a ribosome binding site for translation initiation and a transcription terminator.
  • the vector may also include appropriate sequences for amplifying expression.
  • the expression vectors preferably contain a gene to provide a phenotypic trait for selection of transfected host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in FL coli.
  • the vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transfect an appropriate host to permit the host to express the protein.
  • appropriate hosts there may be mentioned: bacterial cells, such as IL coli. Salmonella typhimurium Streptomyces sp.: fungal cells, such as yeast; insect cells, such as Drosophila and Sf9; animal cells, such as CHO, COS or Bowes melanoma; plant cells, etc.
  • bacterial cells such as IL coli.
  • Salmonella typhimurium Streptomyces sp. fungal cells, such as yeast
  • insect cells such as Drosophila and Sf9
  • animal cells such as CHO, COS or Bowes melanoma
  • plant cells etc.
  • the selection of an appropriate host is deemed to be within the scope of those skilled in the art from the teachings provided herein.
  • the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above.
  • the constructs comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a forward or reverse orientation.
  • the construct further comprises regulatory sequences including, for example, a promoter, operably linked to the sequence.
  • suitable vectors and promoters are known to those of skill in the art and are commercially available. The following vectors are provided by way of example.
  • Bacterial pINCY (Incyte Pharmaceuticals Inc., Palo Alto, CA), pSPORTl (Life Technologies, Gaithersburg, MD), pQE70, pQE60, pQE-9 (Qiagen) pBs, phagescript, psiX174, pBluescript SK, pBsKS, pNH8a, pNHl ⁇ a, pNH18a, pNH46a (Stratagene); pTrc99A, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia); Eukaryotic: pWLneo, pSV2cat, pOG44, pXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia). However, any other plasmid or vector may be used as long as it is replicable and viable in the host.
  • Plasmid pINCY is generally identical to the plasmid pSPORTl (available from Life Technologies, Gaithersburg, MD) with the exception that it has two modifications in the polylinker (multiple cloning site). These modifications are (1) it lacks a Hindffl restriction site and (2) its EcoRI restriction site lies at a different location.
  • pINCY is created from pSPORTl by cleaving pSPORTl with both Hindi ⁇ and EcoRI and replacing the excised fragment of the polylinker with synthetic DNA fragments (SEQUENCE ID NO 2 and SEQUENCE ID NO 3). This replacement may be made in any manner known to those of ordinary skill in the art.
  • the two nucleotide sequences may be generated synthetically with 5' terminal phosphates, mixed together, and then ligated under standard conditions for performing staggered end ligations into the pSPORTl plasmid cut with Hindi ⁇ and EcoRI.
  • Suitable host cells such as EL coH DH5 ⁇ cells
  • Plasmid DNA then is prepared from individual clones and subjected to restriction enzyme analysis or DNA sequencing in order to confirm the presence of insert sequences in the proper orientation.
  • Other cloning strategies known to the ordinary artisan also may be employed.
  • Promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers.
  • Two appropriate vectors are pKK232-8 and pCM7.
  • Particular named bacterial promoters include lad, lacZ, T3, SP6, T7, gpt, lambda P sub R, P sub L and tip.
  • Eukaryotic promoters include cytomegalovirus (CMV) immediate early, herpes simplex virus (HSV) thymidine kinase, early and late SV40, LTRs from retroviruses and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art.
  • the present invention provides host cells containing the above-described construct.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, or electroporation [L. Davis et al, Basic Methods in Molecular Biology. 2nd edition, Appleton and Lang, Paramount Publishing, East Norwalk, CT (1994)].
  • constructs in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence.
  • the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
  • Recombinant proteins can be expressed in mammalian cells, yeast, bacteria, or other cells, under the control of appropriate promoters.
  • Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention.
  • Appropriate cloning and expression vectors- for use with prokaryotic and eukaryotic hosts are described by Sambrook et al., Molecular Cloning: A Laboratory Manual. Second Edition, (Cold Spring Harbor, NY, 1989).
  • Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription. Examples include the SV40 enhancer on the late side of the replication origin (bp 100 to 270), a cytomegalovirus early promoter enhancer, a polyoma enhancer on the late side of the replication origin and adenovirus enhancers.
  • recombinant expression vectors will include origins of replication and selectable markers permitting transfection of the host cell, e.g., the ampicillin resistance gene of EL coH and S cerevisiae TRP 1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence.
  • promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), alpha factor, acid phosphatase, or heat shock proteins, among others.
  • the heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium.
  • the heterologous sequence can encode a fusion protein including an N- terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
  • Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter.
  • the vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desirable, provide amplification within the host.
  • Suitable prokaryotic hosts for transfection include EL coH, Bacillus subtilis. Salmonella typhimurium and various species within the genera Pseudomonas. Strep tomyces and Staphylococcus. although others may also be employed as a routine matter of choice.
  • Useful expression vectors for bacterial use comprise a selectable marker and bacterial origin of replication derived from plasmids comprising genetic elements of the well-known cloning vector pBR322 (ATCC 37017).
  • Other vectors include but are not limited to PKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEM1 (Promega Biotec, Madison, WI). These pBR322 "backbone" sections are combined with an appropriate promoter and the structural sequence to be expressed.
  • the selected promoter is derepressed by appropriate means (e.g., temperature shift or chemical induction), and cells are cultured for an additional period.
  • Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.
  • Microbial cells employed in expression of proteins can be disrupted by any convenient method including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents. Such methods are well-known to the ordinary artisan.
  • mammalian cell culture systems can also be employed to express recombinant protein.
  • mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts described by Gluzman, Cell 23: 175 (1981), and other cell lines capable of expressing a compatible vector, such as the C127, HEK-293, 3T3, CHO, HeLa and BHK cell lines.
  • Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer and also any necessary ribosome binding sites, polyadenylation sites, splice donor and acceptor sites, transcriptional termination sequences and 5' flanking nontranscribed sequences.
  • DNA sequences derived from the SV40 viral genome for example, SV40 origin, early promoter, enhancer, splice, and polyadenylation sites may be used to provide the required nontranscribed genetic elements.
  • useful vectors include pRc/CMV and pcDNA3 (available from Invitrogen, San Diego, CA).
  • PS213 polypeptides are recovered and purified from recombinant cell cultures by known methods including affinity chromatography, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, hydroxyapatite chromatography or lectin chromatography. It is preferred to have low concentrations (approximately 0.1-5 mM) of calcium ion present during purification [Price, et al., J. Biol. Chem. 244:917 (1969)]. Protein refolding steps can be used, as necessary, in completing configuration of the polypeptide. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.
  • HPLC high performance liquid chromatography
  • polypeptides of the present invention may be naturally purified products expressed from a high expressing cell line, or a product of chemical synthetic procedures, or produced by recombinant techniques from a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and mammalian cells in culture).
  • a prokaryotic or eukaryotic host for example, by bacterial, yeast, higher plant, insect and mammalian cells in culture.
  • the polypeptides of the present invention may be glycosylated with mammalian or other eukaryotic carbohydrates or may be non- glycosylated.
  • the polypeptides of the invention may also include an initial methionine amino acid residue.
  • the starting plasmids can be constructed from available plasmids in accord with published, known procedures.
  • mRNA is isolated from prostate tissue and used to generate the cDNA library.
  • Prostate tissue is obtained from patients by surgical resection and is classified as tumor or non-tumor tissue by a pathologist.
  • the cDNA inserts from random isolates of the prostate tissue libraries are sequenced in part, analyzed in detail as set forth in the Examples.
  • the reading frame of the nucleotide sequence can be ascertained by several types of analyses. First, reading frames contained within the coding sequence can be analyzed for the presence of start codon ATG and stop codons TGA, TAA or TAG. Typically, one reading frame will continue throughout the major portion of a cDNA sequence while other reading frames tend to contain numerous stop codons. In such cases, reading frame determination is straightforward. In other more difficult cases, further analysis is required.
  • Coding DNA for particular organisms tends to contain certain nucleotides within certain triplet periodicities, such as a significant preference for pyrimidines in the third codon position.
  • Coding DNA for particular organisms (bacteria, plants and animals) tends to contain certain nucleotides within certain triplet periodicities, such as a significant preference for pyrimidines in the third codon position.
  • These preferences have been incorporated into widely available software which can be used to determine coding potential (and frame) of a given stretch of DNA.
  • the algorithm-derived information combined with start stop codon information can be used to determine proper frame with a high degree of certainty. This, in turn, readily permits cloning of the sequence in the correct reading frame into appropriate expression vectors.
  • vectors of interest include cloning vectors, such as plasmids, cosmids, phage derivatives, phagemids, as well as sequencing, replication and expression vectors, and the like.
  • vectors contain an origin of replication functional in at least one organism, convenient restriction endonuclease digestion sites and selectable markers appropriate for particular host cells.
  • the vectors can be transferred by a variety of means known to those of skill in the art into suitable host cells which then produce the desired DNA, RNA or polypeptides.
  • nucleotide sequences provided herein have been prepared by current, state-of-the-art, automated methods and, as such, may contain unidentified nucleotides. These will not present a problem to those skilled in the art who wish to practice the invention.
  • Several methods employing standard recombinant techniques, described in J. Sambrook (supra) or periodic updates thereof, may be used to complete the missing sequence information.
  • the same techniques used for obtaining a full length sequence, as described herein, may be used to obtain nucleotide sequences.
  • Expression of a particular cDNA may be accomplished by subcloning the cDNA into an appropriate expression vector and transfecting this vector into an appropriate expression host.
  • the cloning vector used for the generation of the prostate tissue cDNA library can be used for transcribing mRNA of a particular cDNA and contains a promoter for beta-galactosidase, an amino-terminal met and the subsequent seven amino acid residues of beta-galactosidase. Immediately following these eight residues is an engineered bacteriophage promoter useful for artificial priming and transcription, as well as a number of unique restriction sites, including EcoRI, for cloning.
  • the vector can be transfected into an appropriate host strain of EL coli.
  • IPTG isopropylthiogalactoside
  • the cDNA can be shuttled into other vectors known to be useful for expression of protein in specific hosts.
  • Oligonucleotide primers containing cloning sites and segments of DNA sufficient to hybridize to stretches at both ends of the target cDNA can be synthesized chemically by standard methods. These primers can then be used to amplify the desired gene segments by PCR.
  • the resulting new gene segments can be digested with appropriate restriction enzymes under standard conditions and isolated by gel electrophoresis. Alternately, similar gene segments can be produced by digestion of the cDNA with appropriate restriction enzymes and filling in the missing gene segments with chemically synthesized oligonucleotides. Segments of the coding sequence from more than one gene can be ligated together and cloned in appropriate vectors to optimize expression of recombinant sequence.
  • Suitable expression hosts for such chimeric molecules include, but are not limited to, mammalian cells, such as Chinese Hamster Ovary (CHO) and human embryonic kidney (HEK) 293 cells, insect cells, such as Sf9 cells, yeast cells, such as Saccharomyces cerevisiae and bacteria, such as IL coli.
  • a useful expression vector may also include an origin of replication to allow propagation in bacteria and a selectable marker such as the beta-lactamase antibiotic resistance gene to allow selection in bacteria.
  • the vectors may include a second selectable marker, such as the neomycin phosphotransferase gene, to allow selection in transfected eukaryotic host cells.
  • Vectors for use in eukaryotic expression hosts may require the addition of 3' poly A tail if the sequence of interest lacks poly A.
  • the vector may contain promoters or enhancers which increase gene expression.
  • promoters are host specific and include, but are not limited to, MMTV, SV40, or metallothionine promoters for CHO cells; tip, lac, tac or T7 promoters for bacterial hosts; or alpha factor, alcohol oxidase or PGH promoters for yeast.
  • Adenoviral vectors with or without transcription enhancers such as the Rous sarcoma virus (RSV) enhancer, may be used to drive protein expression in mammalian cell lines. Once homogeneous cultures of recombinant cells are obtained, large quantities of recombinantly produced protein can be recovered from the conditioned medium and analyzed using chromatographic methods well known in the art.
  • RSV Rous sarcoma virus
  • An alternative method for the production of large amounts of secreted protein involves the transfection of mammalian embryos and the recovery of the recombinant protein from milk produced by transgenic cows, goats, sheep, etc. Polypeptides and closely related molecules may be expressed recombinantly in such a way as to facilitate protein purification.
  • One approach involves expression of a chimeric protein which includes one or more additional polypeptide domains not naturally present on human polypeptides.
  • Such purification-facilitating domains include, but are not limited to, metal-chelating peptides such as histidine-tryptophan domains that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp, Seattle, WA).
  • metal-chelating peptides such as histidine-tryptophan domains that allow purification on immobilized metals
  • protein A domains that allow purification on immobilized immunoglobulin
  • the domain utilized in the FLAGS extension/affinity purification system Immunex Corp, Seattle, WA.
  • the inclusion of a cleavable linker sequence such as Factor XA or enterokinase from Invitrogen (San Diego, CA) between the polypeptide sequence and the purification domain may be useful for recovering the polypeptide.
  • PS213 polypeptides including fragments, derivatives, and analogs thereof, or cells expressing such polypeptides, can be utilized in a variety of assays, many of which are described herein, for the detection of antibodies to prostate tissue. They also can be used as immunogens to produce antibodies. These antibodies can be, for example, polyclonal or monoclonal antibodies, chimeric, single chain and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments.
  • antibodies generated against a polypeptide comprising a sequence of the present invention can be obtained by direct injection of the polypeptide into an animal or by administering the polypeptide to an animal such as a mouse, rabbit, goat or human. A mouse, rabbit or goat is preferred.
  • the polypeptide is selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9,
  • SEQUENCE ID NO 10 SEQUENCE ID NO 10, and fragments thereof.
  • the antibody so obtained then will bind the polypeptide itself. In this manner, even a sequence encoding only a fragment of the polypeptide can be used to generate antibodies that bind the native polypeptide. Such antibodies then can be used to isolate the polypeptide from test samples such as tissue suspected of containing that polypeptide.
  • test samples such as tissue suspected of containing that polypeptide.
  • any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique as described by Kohler and Milstein, Nature 256:495-497 (1975), the trioma technique, the human B-cell hybridoma technique as described by Kozbor et al., Immun.
  • Various assay formats may utilize the antibodies of the present invention, including "sandwich” immunoassays and probe assays.
  • the antibodies of the present invention, or fragments thereof can be employed in various assay systems to determine the presence, if any, of PS213 antigen in a test sample.
  • a polyclonal or monoclonal antibody or fragment thereof, or a combination of these antibodies which has been coated on a solid phase, is contacted with a test sample, to form a first mixture. This first mixture is incubated for a time and under conditions sufficient to form antigen/antibody complexes.
  • an indicator reagent comprising a monoclonal or a polyclonal antibody or a fragment thereof, or a combination of these antibodies, to which a signal generating compound has been attached, is contacted with the antigen/antibody complexes to form a second mixture.
  • This second mixture then is incubated for a time and under conditions sufficient to form antibody /antigen/antibody complexes.
  • the presence of PS213 antigen in the test sample and captured on the solid phase, if any, is determined by detecting the measurable signal generated by the signal generating compound.
  • the amount of PS213 antigen present in the test sample is proportional to the signal generated.
  • a mixture is formed by contacting: (1) a polyclonal antibody, monoclonal antibody, or fragment thereof, which specifically binds to PS213 antigen, or a combination of such antibodies bound to a solid support; (2) the test sample; and (3) an indicator reagent comprising a monoclonal antibody, polyclonal antibody, or fragment thereof, which specifically binds to a different PS213 antigen (or a combination of these antibodies) to which a signal generating compound is attached.
  • This mixture is incubated for a time and under conditions sufficient to form antibody/antigen/antibody complexes.
  • the presence, if any, of PS213 antigen present in the test sample and captured on the solid phase is determined by detecting the measurable signal generated by the signal generating compound.
  • the amount of PS213 antigen present in the test sample is proportional to the signal generated.
  • one or a combination of at least two monoclonal antibodies of the invention can be employed as a competitive probe for the detection of antibodies to PS213 antigen.
  • PS213 polypeptides such as the recombinant antigens disclosed herein, either alone or in combination, are coated on a solid phase.
  • a test sample suspected of containing antibody to PS213 antigen then is incubated with an indicator reagent comprising a signal generating compound and at least one monoclonal antibody of the invention for a time and under conditions sufficient to form antigen/antibody complexes of either the test sample and indicator reagent bound to the solid phase or the indicator reagent bound to the solid phase.
  • each of the monoclonal or polyclonal antibodies of the present invention can be employed in the detection of PS213 antigens in tissue sections, as well as in cells, by immunohistochemical analysis.
  • the tissue sections can be cut from either frozen or chemically fixed samples of tissue.
  • the antigens are to be detected in cells, the cells can be isolated from blood, urine, breast aspirates, or other bodily fluids.
  • the cells may be obtained by biopsy, either surgical or by needle.
  • the cells can be isolated by centrifugation or magnetic attraction after labeling with magnetic particles or ferrofluids so as to enrich a particular fraction of cells for staining with the antibodies of the present invention.
  • these monoclonal antibodies can be bound to matrices similar to CNBr-activated Sepharose and used for the affinity purification of specific PS213 polypeptides from cell cultures or biological tissues such as to purify recombinant and native PS213 proteins.
  • the monoclonal antibodies of the invention also can be used for the generation of chimeric antibodies for therapeutic use, or other similar applications.
  • the monoclonal antibodies or fragments thereof can be provided individually to detect PS213 antigens.
  • Combinations of the monoclonal antibodies (and fragments thereof) provided herein also may be used together as components in a mixture or "cocktail" of at least one PS213 antibody of the invention, along with antibodies which specifically bind to other PS213 regions, each antibody having different binding specificities.
  • this cocktail can include the monoclonal antibodies of the invention which are directed to PS213 polypeptides disclosed herein and other monoclonal antibodies specific to other antigenic determinants of PS213 antigens or other related proteins.
  • the polyclonal antibody or fragment thereof which can be used in the assay formats should specifically bind to a PS213 polypeptide or other PS213 polypeptides additionally used in the assay.
  • the polyclonal antibody used preferably is of mammalian origin such as, human, goat, rabbit or sheep polyclonal antibody which binds PS213 polypeptide. Most preferably, the polyclonal antibody is of rabbit origin.
  • the polyclonal antibodies used in the assays can be used either alone or as a cocktail of polyclonal antibodies.
  • the cocktails used in the assay formats are comprised of either monoclonal antibodies or polyclonal antibodies having different binding specificity to PS213 polypeptides, they are useful for the detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining the predisposition to, diseases and conditions of the prostate, such as prostate cancer.
  • PS213 antigen may be detectable in assays by use of a recombinant antigen as well as by use of a synthetic peptide or purified peptide, which peptide comprises an amino acid sequence of PS213.
  • amino acid sequence of such a polypeptide is selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID -NO 10, and fragments thereof. It also is within the scope of the present invention that different synthetic, recombinant or purified peptides, identifying different epitopes of PS213, can be used in combination in an assay for the detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining the predisposition to diseases and conditions of the prostate, such as prostate cancer.
  • all of these peptides can be coated onto one solid phase; or each separate peptide may be coated onto separate solid phases, such as microparticles, and then combined to form a mixture of peptides which can be later used in assays.
  • multiple peptides which define epitopes from different antigens may be used for the detection, diagnosis, staging, monitoring, prognosis, prevention or treatment of, or determining the predisposition to. diseases and conditions of the prostate, such as prostate cancer.
  • Peptides coated on solid phases or labeled with detectable labels are then allowed to compete with those present in a patient sample (if any) for a limited amount of antibody.
  • a reduction in binding of the synthetic, recombinant, or purified peptides to the antibody (or antibodies) is an indication of the presence of PS213 antigen in the patient sample.
  • the presence of PS213 antigen indicates the presence of prostate tissue disease, especially prostate cancer, in the patient. Variations of assay formats are known to those of ordinary skill in the art and many are discussed herein below.
  • the presence of anti-PS213 antibody and/or PS213 antigen can be detected in a simultaneous assay, as follows.
  • a test sample is simultaneously contacted with a capture reagent of a first analyte, wherein said capture reagent comprises a first binding member specific for a first analyte attached to a solid phase and a capture reagent for a second analyte, wherein said capture reagent comprises a first binding member for a second analyte attached to a second solid phase, to thereby form a mixture.
  • This mixture is incubated for a time and under conditions sufficient to form capture reagent/first analyte and capture reagent/second analyte complexes.
  • Such so-formed complexes then are contacted with an indicator reagent comprising a member of a binding pair specific for the first analyte labeled with a signal generating compound and an indicator reagent comprising a member of a binding pair specific for the second analyte labeled with a signal generating compound to form a second mixture.
  • This second mixture is incubated for a time and under conditions sufficient to form capture reagent first analyte/indicator reagent complexes and capture reagent/second analyte/indicator reagent complexes.
  • the presence of one or more analytes is determined by detecting a signal generated in connection with the complexes formed on either or both solid phases as an indication of the presence of one or more analytes in the test sample.
  • recombinant antigens derived from the expression systems disclosed herein may be utilized, as well as monoclonal antibodies produced from the proteins derived from the expression systems as disclosed herein.
  • PS213 antigen can be the first analyte.
  • the polypeptides disclosed herein may be utilized to detect the presence of antibody against PS213 antigen in test samples.
  • a test sample is incubated with a solid phase to which at least one polypeptide such as a recombinant protein or synthetic peptide has been attached.
  • the polypeptide is selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9,
  • SEQUENCE ID NO 10 SEQUENCE ID NO 10, and fragments thereof. These are reacted for a time and under conditions sufficient to form antigen/antibody complexes. Following incubation, the antigen antibody complex is detected. Indicator reagents may be used to facilitate detection, depending upon the assay system chosen. In another assay format, a test sample is contacted with a solid phase to which a recombinant protein produced as described herein is attached, and also is contacted with a monoclonal or polyclonal antibody specific for the protein, which preferably has been labeled with an indicator reagent.
  • the solid phase After incubation for a time and under conditions sufficient for antibody/antigen complexes to form, the solid phase is separated from the free phase, and the label is detected in either the solid or free phase as an indication of the presence of antibody against PS213 antigen.
  • Other assay formats utilizing the recombinant antigens disclosed herein are contemplated. These include contacting a test sample with a solid phase to which at least one antigen from a first source has been attached, incubating the solid phase and test sample for a time and under conditions sufficient to form antigen/antibody complexes, and then contacting the solid phase with a labeled antigen, which antigen is derived from a second source different from the first source.
  • a recombinant protein derived from a first source such as IL coH is used as a capture antigen on a solid phase
  • a test sample is added to the so-prepared solid phase, and following standard incubation and washing steps as deemed or required, a recombinant protein derived from a different source (i.e., non-E. coli) is utilized as a part of an indicator reagent which subsequently is detected.
  • a recombinant antigen on a solid phase and synthetic peptide in the indicator phase also are possible.
  • any assay format which utilizes an antigen specific for PS213 produced or derived from a first source as the capture antigen and an antigen specific for PS213 from a different second source is contemplated.
  • various combinations of recombinant antigens, as well as the use of synthetic peptides. purified proteins and the like, are within the scope of this invention. Assays such as this and others are described in U.S. Patent No. 5,254,458. which enjoys common ownership.
  • ion capture procedures for immobilizing an immobilizable reaction complex with a negatively charged polymer can be employed according to the present invention to effect a fast solution-phase immunochemical reaction.
  • An immobilizable immune complex is separated from the rest of the reaction mixture by ionic interactions between the negatively charged poly-anion/immune complex and the previously treated, positively charged porous matrix and detected by using various signal generating systems previously described, including those described in chemiluminescent signal measurements as described in EPO Publication No. 0 273,115.
  • the methods of the present invention can be adapted for use in systems which utilize microparticle technology including automated and semi- automated systems wherein the solid phase comprises a microparticle (magnetic or non-magnetic).
  • Such systems include those described in, for example, published EPO applications Nos. EP 0 425 633 and EP 0 424 634, respectively.
  • SPM scanning probe microscopy
  • the capture phase for example, at least one of the monoclonal antibodies of the invention
  • a scanning probe microscope is utilized to detect antigen/antibody complexes which may be present on the surface of the solid phase.
  • the use of scanning tunneling microscopy eliminates the need for labels which normally must be utilized in many immunoassay systems to detect antigen/antibody complexes.
  • SPM to monitor specific binding reactions can occur in many ways.
  • one member of a specific binding partner is attached to a surface suitable for scanning.
  • the attachment of the analyte specific substance may be by adsorption to a test piece which comprises a solid phase of a plastic or metal surface, following methods known to those of ordinary skill in the art.
  • covalent attachment of a specific binding partner may be by adsorption to a test piece which comprises a solid phase of a plastic or metal surface, following methods known to those of ordinary skill in the art.
  • test piece which test piece comprises a solid phase of derivatized plastic, metal, silicon, or glass
  • Covalent attachment methods are known to those skilled in the art and include a variety of means to irreversibly link specific binding partners to the test piece. If the test piece is silicon or glass, the surface must be activated prior to attaching the specific binding partner. Also, polyelectrolyte interactions may be used to immobilize a specific binding partner on a surface of a test piece by using techniques and chemistries. The preferred method of attachment is by covalent means. Following attachment of a specific binding member, the surface may be further treated with materials such as serum, proteins, or other blocking agents to minimize non-specific binding. The surface also may be scanned either at the site of manufacture or point of use to verify its suitability for assay purposes. The scanning process is not anticipated to alter the specific binding properties of the test piece.
  • the reagents such as antibodies, proteins and peptides of the present invention can be utilized in non-solid phase assay systems. These assay systems are known to those skilled in the art, and are considered to be within the scope of the present invention. It is contemplated that the reagent employed for the assay can be provided in the form of a test kit with one or more containers such as vials or bottles, with each container containing a separate reagent such as a probe, primer, monoclonal antibody or a cocktail of monoclonal antibodies, or a polypeptide (e.g. recombinantly, synthetically produced or purified) employed in the assay.
  • a test kit with one or more containers such as vials or bottles, with each container containing a separate reagent such as a probe, primer, monoclonal antibody or a cocktail of monoclonal antibodies, or a polypeptide (e.g. recombinantly, synthetically produced or purified) employed in the assay.
  • the polypeptide is selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
  • Other components such as buffers, controls and the like, known to those of ordinary skill in art, may be included in such test kits. It also is contemplated to provide test kits which have means for collecting test samples comprising accessible body fluids, e.g., blood, urine, saliva and stool.
  • Such tools useful for collection (“collection materials”) include lancets and absorbent paper or cloth for collecting and stabilizing blood; swabs for collecting and stabilizing saliva; cups for collecting and stabilizing urine or stool samples.
  • Collection materials, papers, cloths, swabs, cups and the like may optionally be treated to avoid denaturation or irreversible adsorption of the sample.
  • the collection materials also may be treated with or contain preservatives, stabilizers or antimicrobial agents to help maintain the integrity of the specimens.
  • Test kits designed for the collection, stabilization and preservation of test specimens obtained by surgery or needle biopsy are also useful. It is contemplated that all kits may be configured in two components which can be provided separately; one component for collection and transport of the specimen and the other component for the analysis of the specimen.
  • the collection component for example, can be provided to the open market user while the components for analysis can be provided to others such as laboratory personnel for determination of the presence, absence or amount of analyte.
  • kits for the collection, stabilization and preservation of test specimens may be configured for use by untrained personnel and may be available in the open market for use at home with subsequent transportation to a laboratory for analysis of the test sample. In Vivo Antibody Use.
  • Antibodies of the present invention can be used in vivo: that is, they can be injected into patients suspected of having diseases of the prostate for diagnostic or therapeutic uses.
  • the use of antibodies for in vivo diagnosis is well known in the art.
  • Sumerdon et al, Nucl. Med. Biol 17:247-254 (1990) have described an optimized antibody-chelator for the radioimmunoscintographic imaging of carcinoembryonic antigen (CEA) expressing tumors using Indium- 111 as the label.
  • CEA carcinoembryonic antigen
  • paramagnetic ions such as Gadolinium (III) or Manganese (II) can be used.
  • Localization of the label within the prostate or external to the prostate may allow determination of spread of the disease.
  • the amount of label within the prostate may allow determination of the presence or absence of cancer of the prostate.
  • injection of an antibody directed against PS213 antigen may have therapeutic benefit.
  • the antibody may exert its effect without the use of attached agents by binding to PS213 antigen expressed on or in the tissue or organ.
  • the antibody may be conjugated to cytotoxic agents such as drugs, toxins, or radionuclides to enhance its therapeutic effect.
  • EST's expressed sequence tags
  • LEFSEQTM database available from Incyte Pharmaceuticals, Palo Alto, CA
  • a transcript image is a listing of the number of EST's for each of the represented genes in a given tissue library. EST's sharing regions of mutual sequence overlap are classified into clusters. A cluster is assigned a clone number from a representative 5' EST.
  • a cluster of interest can be extended by comparing its consensus sequence with sequences of other EST's which did not meet the criteria for automated clustering.
  • the alignment of all available clusters and single EST's represent a contig from which a consensus sequence is derived.
  • the transcript images then were evaluated to identify EST sequences that were representative primarily of the prostrate tissue libraries. These target clones then were ranked according to their abundance (occurrence) in the target libraries and their absence from background libraries. Higher abundance clones with low background occurrence were given higher study priority.
  • EST's corresponding to the sequence of PS213 (or fragments thereof) were found in 30.9% (13 of 42) of prostrate tissue libraries.
  • Overlapping clones were identified for further study. These represented the minimum number of clones that were needed to form the contig and from which a consensus sequence was derived.
  • the nucleotide sequences of the clones were entered in the SequencherTM Program (available from Gene Codes Corporation, Ann Arbor, MI. in order to generate a nucleotide alignment (contig map) and then generate their consensus sequence. Following this, a three-frame translation was performed on the consensus sequence. The first forward frame was found to have an open reading frame which was compared with published sequences using software and techniques known to those skilled in the art. The nucleotide sequence was found to be the same as that deposited with GenBank under Accession No.
  • Example 2 Sequencing of PS213 EST-Specific Clones DNA sequences for clones which comprise the most upstream and downstream EST's of the PS213 gene contig are determined using dideoxy termination sequencing with dye terminators following known methods [F. Sanger et al., Proc. Natl. Acad. Sci. USA 74:5463 (1977)].
  • vectors such as pSPORTl (Life Technologies, Gaithersburg, MD) and pINCY (available from Incyte Pharmaceuticals, Inc., Palo Alto, CA) contain universal priming sites just adjacent to the 3' and 5' ligation junctions of the inserts, the inserts are sequenced in both directions using universal primers, SEQUENCE ED NO 4 and SEQUENCE ID NO 5 ( New England Biolabs,
  • sequencing reactions are run on a polyacrylamide denaturing gel, and the sequences are determined by an Applied Biosystems 377 Sequencer (available from Applied Biosystems. Foster City, CA) or other sequencing apparatus.
  • RNA Extraction from Tissue Total RNA is isolated from prostrate tissues and from non-prostrate tissues. Various methods are utilized, including but not limited to the lithium chloride/urea technique, known in the art and described by Kato et al., (J. Virol. 61:2182-2191, 1987), and TRIzolTM (Gibco-BRL, Grand Island, NY).
  • tissue is placed in a sterile conical tube on ice and 10-15 volumes of 3 M LiCl, 6 M urea, 5 mM EDTA, 0.1 M ⁇ -mercaptoethanol, 50 mM Tris-HCl
  • tissue is homogenized with a Polytron ® homogenizer (Brinkman Instruments, Inc., Westbury, NY) for 30-50 sec on ice.
  • the solution is transferred to a 15 ml plastic centrifuge tube and placed overnight at -20°C.
  • the tube is centrifuged for 90 min at 9,000 x g at 0-4°C and the supernatant is immediately decanted.
  • Ten ml of 3 M LiCl are added and the tube is vortexed for 5 sec. The tube is centrifuged for 45 min at 1 1,000 x g at 0-4°C.
  • the tube is vortexed for 1-3 sec and centrifuged for 20 min at 3,000 x g at 10°C.
  • the PCI extraction is repeated and followed by two similar extractions with chloroform isoamyl alcohol (Cl).
  • the final aqueous solution is transferred to a prechilled 15 ml Corex glass tube containing 6 ml of absolute ethanol, the tube is covered with parafilm, and placed at -20°C overnight.
  • the tube is centrifuged for 30 min at 10,000 x g at 0-4°C and the ethanol supernatant is decanted immediately.
  • the RNA pellet is washed four times with 10 ml of 75% ice-cold ethanol and the final pellet is air dried for 15 min at room temperature.
  • RNA samples are suspended in 0.5 ml of 10 mM TE (pH 7.6, 1 mM EDTA) and its concentration is determined spectrophotometrically. RNA samples are aliquoted and stored at -70°C as ethanol precipitates. The quality of the RNA is determined by agarose gel electrophoresis (see
  • Example 5 Northern Blot Analysis and staining with 0.5 ⁇ g/ml ethidium bromide for one hour. RNA samples that do not contain intact rRNAs are excluded from the study.
  • RNA reagent 1 ml of Ultraspec RNA reagent is added to 120 mg of pulverized tissue in a 2.0 ml polypropylene microfuge tube, homogenized with a Polytron ® homogenizer (Brinkman Instruments, Inc., Westbury, NY) for 50 sec and placed on ice for 5 min. Then, 0.2 ml of chloroform is added to each sample, followed by vortexing for 15 sec. The sample is placed on ice for another 5 min, followed by centrifugation at 12,000 x g for 15 min at 4°C. The upper layer is collected and transferred to another RNase-free 2.0 ml microfuge tube.
  • RNA pellet is dried in a Speedvac (Savant, Farmingdale, NY) for 5 min and reconstituted in RNase-free water.
  • RNA Extraction from Blood Mononuclear Cells Mononuclear cells are isolated from blood samples from patients by centrifugation using Ficoll-Hypaque as follows. A 10 ml volume of whole blood is mixed with an equal volume of
  • RPMI Medium (Gibco-BRL, Grand Island, NY). This mixture is then underlayed with 10 ml of Ficoll-Hypaque (Pharmacia, Piscataway, NJ) and centrifuged for 30 minutes at 200 x g. The buffy coat containing the mononuclear cells is removed, diluted to 50 ml with Dulbecco's PBS (Gibco-BRL, Grand Island, NY) and the mixture centrifuged for 10 minutes at 200 x g. After two washes, the resulting pellet is resuspended in Dulbecco's PBS to a final volume of 1 ml. RNA is prepared from the isolated mononuclear cells as described by N.
  • the pelleted mononuclear cells are brought to a final volume of 1 ml and then are resuspended in 250 ⁇ L of PBS and mixed with 2.5 ml of 3M LiCl, 6M urea, 5mM EDTA, 0.1M 2- mercaptoethanol, 50mM Tris-HCl (pH 7.5). The resulting mixture is homogenized and incubated at -20°C overnight. The homogenate is centrifuged at 8,000 RPM in a Beckman J2-21M rotor for 90 minutes at 0-4°C.
  • the pellet is resuspended in 10 ml of 3M LiCl by vortexing and then centrifuged at 10,000 RPM in a Beckman J2-21M rotor centrifuge for 45 minutes at 0-4°C. The resuspending and pelleting steps then are repeated.
  • the pellet is resuspended in 2 ml of 1 mM EDTA, 0.5% SDS, 10 mM Tris (pH 7.5) and 400 ⁇ g Proteinase K with vortexing and then it is incubated at 37°C for 30 minutes with shaking. One tenth volume of 3M NaCl then is added and the mixture is vortexed.
  • RNA is precipitated by the addition of 6 ml of absolute ethanol followed by overnight incubation at -20°C. After the precipitated RNA is collected by centrifugation, the pellet is washed 4 times in 75% ethanol. The pelleted RNA is then dissolved in solution containing ImM EDTA, lOmM Tris-HCl (pH 7.5). Non-prostrate tissues are used as negative controls.
  • the mRNA can be further purified from total RNA by using commercially available kits such as oligo dT cellulose spin columns (RediColTM from Pharmacia, Uppsala, Sweden) for the isolation of poly-adenylated RNA.
  • Total RNA or mRNA can be dissolved in lysis buffer (5M guanidine thiocyanate, 0.1M EDTA, pH 7.0) for analysis in the ribonuclease protection assay.
  • RNA Extraction from polysomes Tissue is minced in saline at 4°C and mixed with 2.5 volumes of 0.8 M sucrose in a TK 150 M (150 mM KCI, 5 mM MgCl 2 , 50 mM Tris-HCl, pH 7.4) solution containing 6 mM 2-mercaptoethanol.
  • the tissue is homogenized in a Teflon-glass Potter homogenizer with five strokes at 100-200 rpm followed by six strokes in a Dounce homogenizer, as described by B. Mechler, Methods in Enzymology 152:241-248 (1987). The homogenate then is centrifuged at 12,000 x g for 15 min at 4°C to sediment the nuclei.
  • the polysomes are isolated by mixing 2 ml of the supernatant with 6 ml of 2.5 M sucrose in TK 150 M and layering this mixture over 4 ml of 2.5 M sucrose in TK, 50 M in a 38 ml polyallomer tube. Two additional sucrose TK I50 M solutions are successively layered onto the extract fraction; a first layer of 13 ml 2.05 M sucrose followed by a second layer of 6 ml of 1.3 M sucrose. The polysomes are isolated by centrifuging the gradient at 90,000 x g for 5 hr at 4°C.
  • the fraction then is taken from the 1.3 M sucrose/2.05 M sucrose interface with a siliconized pasteur pipette and diluted in an equal volume of TE (10 mM Tris-HCl, pH 7.4, 1 mM EDTA).
  • TE 10 mM Tris-HCl, pH 7.4, 1 mM EDTA.
  • An equal volume of 90°C SDS buffer 1% SDS, 200 mM NaCl, 20 mM Tris-HCl, pH 7.4
  • Proteins next are digested with a Proteinase K digestion (50 mg/ml) for 15 min at 37°C.
  • the mRNA is purified with 3 equal volumes of phenol-chloroform extractions followed by precipitation with 0.1 volume of 2 M sodium acetate (pH 5.2) and 2 volumes of 100% ethanol at -20°C overnight.
  • the precipitated RNA is recovered by centrifugation at 12,000 x g for 10 min at 4°C.
  • the RNA is dried and resuspended in TE (pH 7.4) or distilled water. The resuspended RNA then can be used in a slot blot or dot blot hybridization assay to check for the presence of PS213 mRNA (see Example 6).
  • nucleic acid and proteins are dependent on the method of preparation used. Each sample may require a different preparation technique to maximize isolation efficiency of the target molecule. These preparation techniques are within the skill of the ordinary artisan.
  • Example 4 Ribonuclease Protection Assay A. Synthesis of Labeled Complementary RNA (cRNA) Hybridization Probe and Unlabeled Sense Strand.
  • Labeled antisense and unlabeled sense riboprobes are transcribed from the PS213 gene cDNA sequence which contains a 5' RNA polymerase promoter such as SP6 or T7.
  • the sequence may be from a vector containing the appropriate PS213 cDNA insert, or from a PCR-generated product of the insert using PCR primers which incorporate a 5' RNA polymerase promoter sequence.
  • a plasmid containing the PS213 gene cDNA sequence, or fragment thereof, flanked by opposed SP6 and T7 or other RNA polymerase promoters is purified using a Qiagen Plasmid Purification Kit (Qiagen, Chatsworth, CA). Then 10 ⁇ g of the plasmid DNA are linearized by cutting with an appropriate restriction enzyme such as Dde I for 1 hr at 37°C.
  • the linearized plasmid DNA is purified using the QIAprep Kit (Qiagen, Chatsworth, CA) and used for the synthesis of antisense transcript from the appropriate promoter using the Riboprobe ® in vitro Transcription System (Promega Corporation, Madison, WI), as described by the supplier's instructions, incorporating either (alpha 32 P) CTP (Amersham Life Sciences, Inc. Arlington Heights, IL) or biotinylated CTP as a label.
  • 10 ⁇ g of the purified plasmid DNA are cut with restriction enzymes, such as Xba I and Not I, and transcribed as above from the appropriate promoter. Both sense and antisense strands are isolated by spin column chromatography. Unlabeled sense strand is quantitated by UV absorption at 260 nm.
  • solubilized tissue or diluted sense strand is mixed directly with either ; 1) 1 xlO 5 cpm of radioactively labeled probe, or 2) 250 pg of non-isotopically labeled probe in 5 ⁇ l of lysis buffer. Hybridization is allowed to proceed overnight at 37°C. See, T. Kaabache et al., Anal. Biochem. 232:225-230 (1995).
  • RNA that is not hybridized to probe is removed from the reaction as per the Direct ProtectTM protocol using a solution of RNase A and RNase Tl for 30 min at 37°C, followed by removal of RNase by Proteinase K digestion in the presence of sodium sarcosyl. Hybridized fragments protected from digestion are then precipitated by the addition of an equal volume of isopropanol and placed at -70°C for 3 hr. The precipitates are collected by centrifugation at 12,000 x g for 20 min.
  • the precipitates are dissolved in denaturing gel loading dye (80% formamide, 10 mM EDTA (pH 8.0), 1 mg/ml xylene cyanol, 1 mg/ml bromophenol blue), heat denatured, and electrophoresed in 6% polyacrylamide TBE, 8 M urea denaturing gels.
  • denaturing gel loading dye 80% formamide, 10 mM EDTA (pH 8.0), 1 mg/ml xylene cyanol, 1 mg/ml bromophenol blue
  • the gels are imaged and analyzed using the STORMTM storage phosphor autoradiography system (Molecular Dynamics, Sunnyvale. CA). Quantitation of protected fragment bands, expressed in femtograms (fg), is achieved by comparing the peak areas obtained from the test samples to those from the known dilutions of the positive control sense strand (see Section B, supra).
  • hybrids are transferred from the gels to membranes (nylon or nitrocellulose) by blotting and then analyzed using detection systems that employ streptavidin alkaline phosphatase conjugates and chemiluminesence or chemifluoresence reagents.
  • Detection of a product comprising a sequence of the PS213 polynucleotide (SEQUENCE ID NO 1), or fragments or complements thereof, is indicative of the presence of PS213 mRNA(s), suggesting a diagnosis of a prostrate tissue disease or condition, such as prostrate cancer.
  • Example 5 Northern Blotting The Northern blot technique is used to identify a specific size RNA fragment from a complex population of RNA using gel electrophoresis and nucleic acid hybridization. Northern blotting is well-known technique in the art.
  • RNA 5-10 ⁇ g of total RNA (see Example 3) are incubated in 15 ⁇ l of a solution containing 40 mM morphilinopropanesulfonic acid (MOPS) (pH 7.0), 10 mM sodium acetate, 1 mM EDTA, 2.2 M formaldehyde, 50% v/v formamide for 15 min at 65°C.
  • MOPS morphilinopropanesulfonic acid
  • RNA is mixed with 2 ⁇ l of loading buffer (50% glycerol, 1 mM EDTA, 0.4% bromophenol blue, 0.4% xylene cyanol) and loaded into a denaturing 1.0% agarose gel containing 40 mM MOPS (pH 7.0), 10 mM sodium acetate, 1 mM EDTA and 2.2 M formaldehyde.
  • the gel is electrophoresed at 60 V for 1.5 hr and rinsed in RNAse free water.
  • RNA is transferred from the gel onto nylon membranes (Brightstar-Plus, Ambion, Inc., Austin, TX) for 1.5 hours using the downward alkaline capillary transfer method (Chomczynski, Anal. Biochem.
  • RNA is crosslinked to the filter using a StratalinkerTM (Stratagene, Inc., La Jolla, CA) on the autocrosslinking mode and dried for 15 min.
  • the membrane is then placed into a hybridization tube containing 20 ml of preheated prehybridization solution (5X SSC, 50% formamide, 5X Denhardt's solution, 100 ⁇ g/ml denatured salmon sperm DNA) and incubated in a 42°C hybridization oven for at least 3 hr.
  • preheated prehybridization solution 5X SSC, 50% formamide, 5X Denhardt's solution, 100 ⁇ g/ml denatured salmon sperm DNA
  • a 32 P-labeled random-primed probe is generated using a PS213 insert fragment using Random Primer DNA Labeling System (Life Technologies, Inc., Gaithersburg, MD) according to the manufacturer's instructions.
  • Half of the probe is boiled for 10 min, quick chilled on ice and added to the hybridization tube.
  • Hybridization is carried out at 42°C for at least 12 hr.
  • the hybridization solution is discarded and the filter is washed in 30 ml of 3X SSC, 0.1 % SDS at 42°C for 15 min, followed by 30 ml of 3X SSC, 0.1 % SDS at 42°C for 15 min.
  • the filter is wrapped in Saran Wrap, exposed to Kodak XAR- Omat film for 8-96 hr, and the film is developed for analysis.
  • High level of expression of mRNA corresponding to a sequence of the PS213 polynucleotide (SEQUENCE ID NO 1), or fragments or complements thereof, is an indication of the presence of PS213 mRNA, suggesting a diagnosis of a prostrate tissue disease or condition, such as prostrate cancer.
  • Dot and slot blot assays are quick methods to evaluate the presence of a specific nucleic acid sequence in a complex mix of nucleic acid.
  • RNA are mixed in 50 ⁇ l of 50% formamide, 7% formaldehyde, IX SSC, incubated 15 min at 68°C, and then cooled on ice. Then, 100 ⁇ l of 20X SSC are added to the RNA mixture and loaded under vacuum onto a manifold apparatus that has a prepared nitrocellulose or nylon membrane.
  • the membrane is soaked in water, 20X SSC for 1 hour, placed on two sheets of 20X SSC prewet Whatman #3 filter paper, and loaded into a slot blot or dot blot vacuum manifold apparatus.
  • the slot blot is analyzed with probes prepared and labeled as described in Example 4, supra. Detection of mRNA corresponding to a sequence of the PS213 polynucleotide (SEQUENCE ID NO 1), or fragments or complements thereof, is an indication of the presence of PS213, suggesting a diagnosis of a prostrate tissue disease or condition, such as prostrate cancer.
  • This method is useful to directly detect specific target nucleic acid sequences in cells using detectable nucleic acid hybridization probes.
  • Tissues are prepared with cross-linking fixative agents such as paraformaldehyde or glutaraldehyde for maximum cellular RNA retention. See, L. Angerer et al., Methods in Cell Biol. 35:37-71 (1991). Briefly, the tissue is placed in greater than 5 volumes of 1 % glutaraldehyde in 50 mM sodium phosphate, pH 7.5 at 4°C for 30 min. The solution is changed with fresh glutaraldehyde solution (1% glutaraldehyde in 50mM sodium phosphate, pH 7.5) for a further 30 min fixing. The fixing solution should have an osmolality of approximately 0.375% NaCl. The tissue is washed once in isotonic NaCl to remove the phosphate.
  • cross-linking fixative agents such as paraformaldehyde or glutaraldehyde for maximum cellular RNA retention. See, L. Angerer et al., Methods in Cell Biol. 35:37-
  • the fixed tissues then are embedded in paraffin as follows.
  • the tissue is dehydrated though a series of increasing ethanol concentrations for 15 min each: 50% (twice), 70% (twice), 85%, 90% and then 100% (twice).
  • the tissue is soaked in two changes of xylene for 20 min each at room temperature.
  • the tissue is then soaked in two changes of a 1:1 mixture of xylene and paraffin for 20 min each at 60°C; and then in three final changes of paraffin for 15 min each.
  • tissue is cut in 5 ⁇ m sections using a standard microtome and placed on a slide previously treated with a tissue adhesive such as 3- aminopropyltriethoxysilane.
  • Paraffin is removed from the tissue by two 10 min xylene soaks and rehydrated in a series of decreasing ethanol concentrations: 99% twice, 95%, 85%, 70%, 50%, 30%, and then distilled water twice.
  • the sections are pre-treated with 0.2 M HC1 for 10 min and permeabilized with 2 ⁇ g/ml Proteinase K at 37°C for 15 min.
  • Labeled riboprobes transcribed from the PS213 gene plasmid are hybridized to the prepared tissue sections and incubated overnight at 56°C in 3X standard saline extract and 50% formamide. Excess probe is removed by washing in 2X standard saline citrate and 50% formamide followed by digestion with 100 ⁇ g/ml RNase A at 37°C for 30 min. Fluorescence probe is visualized by illumination with ultraviolet (UV) light under a microscope. Fluorescence in the cytoplasm is indicative of PS213 mRNA. Alternatively, the sections can be visualized by autoradiography.
  • UV ultraviolet
  • Target-specific primers are designed to detect the above-described target sequences by reverse transcription PCR using methods known in the art.
  • One step RT-PCR is a sequential procedure that performs both RT and PCR in a single reaction mixture.
  • the procedure is performed in a 200 ⁇ l reaction mixture containing 50 mM (N,N,-bis[2- Hydroxyethyl]glycine), pH 8.15, 81.7 mM KOAc, 33.33 mM KOH, 0.01 mg/ml bovine serum albumin, 0.1 mM ethylene diaminetetraacetic acid, 0.02 mg/ml NaN3 ; 8% w/v glycerol, 150 ⁇ M each of dNTP, 0.25 ⁇ M each primer, 5U rTth polymerase, 3.25 mM Mn(OAc) 2 and 5 ⁇ l of target RNA (see Example 3).
  • RNA and the rTth polymerase enzyme are unstable in the presence of Mn(OAc) 2 , the Mn(OAc) 2 should be added just before target addition.
  • Optimal conditions for cDNA synthesis and thermal cycling readily can be determined by those skilled in the art. The reaction is incubated in a Perkin-Elmer Thermal Cycler 480.
  • RT-PCR also may be performed by using a dual enzyme procedure with Taq polymerase and a reverse transcriptase enzyme, such as MMLV (Moloney murine leukemia virus) or AMV (avian myeloblastosis virus) RT (reverse transcriptase) enzymes.
  • MMLV Microloney murine leukemia virus
  • AMV avian myeloblastosis virus
  • RNasin 1 ⁇ M antisense primer and 25 U AMV or MMLV reverse transcriptase.
  • Reverse transcription is performed at 37-45°C for 30-60 min, followed by further incubation at 95°C for 5 min to inactivate the RT.
  • PCR is performed using 10 ⁇ l of the cDNA reaction in a final PCR reaction volume of 50 ⁇ l containing 10 mM Tris- HCl (pH 8.3), 50 mM KCI, 2 mM MgCl 2 , 200 ⁇ M dNTP, 0.5 ⁇ M of each primer and 2.5 U of Taq polymerase.
  • Optimal conditions for cDNA synthesis and thermal cycling can be readily determined by those skilled in the art.
  • the reaction is incubated in a Perkin-Elmer Thermal Cycler 480 or other comparable instrument. Conditions which may be found useful include 30-45 cycles of amplification (94°C, 1 min; 55-70°C, 1 min; 72°C, 2 min), final extension (72°C, 10 min) and soak at 4°C.
  • PCR Fragment Analysis The correct products then can be verified by size determination using gel electrophoresis with SYBR ® Green I nucleic acid gel stain (Molecular Probes, Eugene, OR) and imaged using a STORM imaging system, or also verified by Southern, dot or slot blot analysis using a labeled probe against the internal sequences of the PCR product.
  • the probes also may be polynucleotides analogs, such as morpholinos or peptide nucleic acids analogs (PNAs).
  • Detection of a product comprising a sequence of the PS213 polynucleotide is indicative of the presence of PS213 mRNA(s), suggesting a diagnosis of a prostrate tissue disease or condition, such as prostrate cancer.
  • Target-specific primers and probes are designed to detect the above-described target sequences by oligonucleotide hybridization PCR.
  • a label-phosphoramidite reagent is prepared and used to add the label to the oligonucleotide during its synthesis. For example, see N. T. Thuong et al., Tet. Letters 29(46):5905-5908 (1988); or J. S.
  • probes are labeled at their 3' end to prevent participation in PCR and the formation of undesired extension products.
  • the probe should have a T at least 15°C below the TM of the primers.
  • the primers and probes are utilized as specific binding members, with or without detectable labels, using standard phosphoramidite chemistry and/or post- synthetic labeling methods which are well-known to one skilled in the art.
  • RNA and the rTth polymerase enzyme are unstable in the presence of Mn(OAc) 2 , the Mn(OAc) 2 should be added just before target addition.
  • the reaction is incubated in a Perkin-Elmer Thermal Cycler 480.
  • Optimal conditions for cDNA synthesis and thermal cycling can be readily determined by those skilled in the art. Conditions which may be found useful include cDNA synthesis (60°C, 30 min), 30-45 amplification cycles (94°C, 40 sec; 55-70°C, 60 sec), oligo-hybridization (97°C, 5 min; 15°C, 5 min; 15°C soak).
  • the correct reaction product contains at least one of the strands of the PCR product and an internally hybridized probe.
  • Example 10 Synthetic Peptide Production Synthetic peptides were modeled and then prepared based upon the published amino acid sequence of the PS213 polypeptide consensus sequence (see Example 1). In particular, a number of PS213 peptides derived from SEQUENCE ID NO 6 were prepared, including the peptides of SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, and SEQUENCE ID NO 10. The C-terminal amino acid of the synthetic peptide represented by SEQUENCE ID NO 10 differs from that shown at the corresponding position in SEQUENCE ID NO 6.
  • cleavage and deprotection conditions were as follows: a volume of 2.5 ml of cleavage reagent (77.5% v/v trifluoroacetic acid, 15% v/v ethanedithiol, 2.5% v/v water, 5% v/v thioanisole, 1-2% w/v phenol) were added to the resin, and agitated at room temperature for 2-4 hours.
  • cleavage reagent 77.5% v/v trifluoroacetic acid, 15% v/v ethanedithiol, 2.5% v/v water, 5% v/v thioanisole, 1-2% w/v phenol
  • the purified peptides were used to immunize animals (see Example 14).
  • Example 11a Expression of Protein in a Cell Line Using Plasmid 577 A. Construction of a PS213 Expression Plasmid. Plasmid 577, described in U.S. patent application Serial No. 08/478,073, filed June 7, 1995, has been constructed for the expression of secreted antigens in a permanent cell line.
  • This plasmid contains the following DNA segments: (a) a 2.3 kb fragment of pBR322 containing bacterial beta-lactamase and origin of DNA replication; (b) a 1.8 kb cassette directing expression of a neomycin resistance gene under control of HSV- 1 thymidine kinase promoter and poly-A addition signals; (c) a 1.9 kb cassette directing expression of a dihydrofolate reductase gene under the control of an Simian Virus 40 (SV40) promoter and poly-A addition signals; (d) a 3.5 kb cassette directing expression of a rabbit immunoglobulin heavy chain signal sequence fused to a modified hepatitis C virus (HCV) E2 protein under the control of the Simian Virus 40 T-Ag promoter and transcription enhancer, the hepatitis B virus surface antigen (HBsAg) enhancer I followed by a fragment of Herpes Simplex Virus- 1 (HSV-1)
  • Plasmids for the expression of secretable PS213 proteins are constructed by replacing the hepatitis C virus E2 protein coding sequence in plasmid 577 with that of a PS213 polynucleotide sequence selected from fragments or complements thereof, of the PS213 polynucleotide (SEQUENCE ID NO 1) as follows. Digestion of plasmid 577 with Xbal releases the hepatitis C virus E2 gene fragment. The resulting plasmid backbone allows insertion of the PS213 cDNA insert downstream of the rabbit immunoglobulin heavy chain signal sequence which directs the expressed proteins into the secretory pathway of the cell. The PS213 cDNA fragment is generated by PCR using standard procedures.
  • Encoded in the sense PCR primer sequence is an Xbal site, immediately followed by a 12 nucleotide sequence that encodes the amino acid sequence Ser-Asn-Glu-Leu ("SNEL") to promote signal protease processing, efficient secretion and final product stability in culture fluids.
  • the primer contains nucleotides complementary to template sequences encoding amino acids of the PS213 gene .
  • the antisense primer incorporates a sequence encoding the following eight amino acids just before the stop codons: Asp-Tyr-Lys- Asp-Asp- Asp- Asp-Lys (SEQUENCE ID NO 11).
  • a recognition site to aid in analysis and purification of the PS213 protein product.
  • a recognition site (termed "FLAG”) that is recognized by a commercially available monoclonal antibody designated anti-FLAG M2 (Eastman Kodak, Co., New Haven, CT) can be utilized, as well as other comparable sequences and their corresponding antibodies.
  • FLAG a recognition site that is recognized by a commercially available monoclonal antibody designated anti-FLAG M2 (Eastman Kodak, Co., New Haven, CT)
  • PCR is performed using GeneAmp ® reagents obtained from Perkin-Elmer-Cetus, as directed by the supplier's instructions. PCR primers are used at a final concentration of 0.5 ⁇ M.
  • PCR is performed on the PS213 plasmid template in a 100 ⁇ l reaction for 35 cycles (94°C, 30 seconds; 55°C, 30 seconds; 72°C, 90 seconds) followed by an extension cycle of 72°C for 10 min.
  • CHO/dhfr- cells are cultured in Ham's F-12 media supplemented with 10% fetal calf serum, L- glutamine (1 mM) and freshly seeded into a flask at a density of 5 - 8 x 10 5 cells per flask.
  • the cells are grown to a confluency of between 60 and 80% for transfection.
  • Twenty micrograms (20 ⁇ g) of plasmid DNA are added to 1.5 ml of Opti-MEM I medium and 100 ⁇ l of Lipofectin Reagent (Gibco-BRL; Grand Island, NY) are added to a second 1.5 ml portion of Opti-MEM I media. The two solutions are mixed and incubated at room temperature for 20 min.
  • the cells are rinsed 3 times with 5 ml of Opti-MEM I medium.
  • the Opti-MEM I-Lipofection-plasmid DNA solution then is overlaid onto the cells.
  • the cells are incubated for 3 hr at 37°C, after which time the Opti-MEM I-Lipofectin-DNA solution is replaced with culture medium for an additional 24 hr prior to selection.
  • F-12 minus medium G dhfr/G418 selection medium
  • Selection medium is Ham's F-12 with L-glutamine and without hypoxanthine, thymidine and glycine (JRH Biosciences, Lenexa, Kansas) and 300 ⁇ g per ml G418 (Gibco-BRL; Grand Island, NY). Media volume-to-
  • DHFR/G418 cells are expanded to allow passage and continuous maintenance in F-12 minus medium G.
  • Amplification of each of the transfected PS213 cDNA sequences is achieved by stepwise selection of DHFR , G418 cells with methotrexate (reviewed by R. Schimke, Cell 37:705-713 [1984]). Cells are incubated with F-12 minus medium G containing 150 nM methotrexate (MTX) (Sigma, St. Louis, MO) for approximately two weeks until resistant colonies appear. Further gene amplification is achieved by selection of 150 nM adapted cells with 5 ⁇ M MTX. P. Antigen Production. F-12 minus medium G supplemented with 5 ⁇ M MTX is overlaid onto just confluent monolayers for 12 to 24 hr at 37°C in 5% CO 2 .
  • methotrexate 150 nM methotrexate
  • the growth medium is removed and the cells are rinsed 3 times with Dulbecco's phosphate buffered saline (PBS) (with calcium and magnesium) (Gibco-BRL; Grand Island, NY) to remove the remaining media/serum which may be present.
  • PBS Dulbecco's phosphate buffered saline
  • VAS custom medium VAS custom formulation with L-glutamine with HEPES without phenol red, available from JRH Bioscience; Lenexa, KS, product number 52-08678P
  • VAS custom medium VAS custom formulation with L-glutamine with HEPES without phenol red, available from JRH Bioscience; Lenexa, KS, product number 52-08678P
  • VAS custom medium VAS custom formulation with L-glutamine with HEPES without phenol red, available from JRH Bioscience; Lenexa, KS, product number 52-08678P
  • VAS custom medium VAS custom formulation with L-glutamine with HEPES
  • protein in pooled VAS medium harvests from roller bottles is exchanged into 50 mM Tris-HCl (pH 7.5), 150 mM NaCl buffer using a Sephadex G-25 (Pharmacia Biotech Inc., Uppsala, Sweden) column. Protein in this buffer is applied to the anti-ET-AG M2 antibody affinity column. Non-binding protein is eluted by washing the column with 50 mM Tris-HCl (pH 7.5), 150 mM NaCl buffer. Bound protein is eluted using an excess of FLAG peptide in 50 mM Tris- HCl (pH 7.5), 150 mM NaCl.
  • the excess FLAG peptide can be removed from the purified PS213 protein by gel electrophoresis or HPLC.
  • plasmid 577 is utilized in this example, it is known to those skilled in the art that other comparable expression systems, such as CMV, can be utilized herein with appropriate modifications in reagent and/or techniques and are within the skill of the ordinary artisan.
  • the largest cloned insert containing the coding region of the PS213 gene is then sub-cloned into either (i) a eukaryotic expression vector which may contain, for example, a cytomegalovirus (CMV) promoter and/or protein fusible sequences which aid in protein expression and detection, or (ii) a bacterial expression vector containing a superoxide-dismutase (SOD) and CMP-KDO synthetase (CKS) or other protein fusion gene for expression of the protein sequence.
  • CMV cytomegalovirus
  • SOD superoxide-dismutase
  • CKS CMP-KDO synthetase
  • Example 1 lb Expression of Protein in a Cell Line Using pcDNA3.1/Myc-His A. Construction of a PS213 Expression Plasmid. Plasmid pcDNA3.1/Myc-His (Cat.# V855-20, Invitrogen, Carlsbad, CA) has been constructed, in the past, for the expression of secreted antigens by most mammalian cell lines. Expressed protein inserts are fused to a myc-his peptide tag.
  • the myc-his tag (SEQUENCE ED NO 12) comprises a c-myc oncoprotein epitope and a polyhistidine sequence which are useful for the purification of an expressed fusion protein by using either anti-myc or anti-his affinity columns, or metalloprotein binding columns.
  • Plasmids for the expression of secretable PS213 proteins are constructed by inserting a PS213 polynucleotide sequence, selected fragments or complements thereof, of the PS213 polynucleotide (SEQUENCE ID NO 1). Prior to construction of a PS213 expression plasmid, the PS213 cDNA sequence is first cloned into a pCR ® -Blunt vector as follows:
  • the PS213 cDNA fragment is generated by PCR using standard procedures. For example, PCR is performed using procedures and reagents from Stratagene ® , Inc. (La Jolla, CA), as directed by the manufacturer. PCR primers are used at a final concentration of 0.5 ⁇ M. PCR using 5 U of pfu polymerase (Stratagene, La Jolla, CA) is performed on the PS213 plasmid template (see Example 2) in a 50 ⁇ l reaction for 30 cycles (94°C, 1 min; 65°C, 1.5 min; 72°C, 3 min) followed by an extension cycle of 72°C for 8 min.
  • the sense PCR primer sequence comprises nucleotides which are either complementary to the pINCY vector directly upstream of the PS213 gene insert or which incorporate a 5' EcoRI restriction site, an adjacent downstream protein translation consensus initiator, and a 3' nucleic acid sequence which is the same sense as the 5'-most end of the PS213 cDNA insert.
  • the antisense PCR primer incorporates a 5' NotI restriction sequence and a sequence complementary to the 3' end of the PS213 cDNA insert just upstream of the 3'-most, in-frame stop codon.)
  • Five microliters (5 ⁇ l) of the resulting blunted-ended PCR product are ligated into 25 ng of linearized pCR ® - Blunt vector (Invitrogen, Carlsbad, CA) interrupting the lethal ccdB gene of the vector.
  • the resulting ligated vector is transformed into TOP10 E_. coH (Invitrogen, Carlsbad, CA) using a One ShotTM Transformation Kit (Invitrogen, Carlsbad, CA) following manufacturer's instructions.
  • the transformed cells are grown on LB- Kan (50 ⁇ g/ml kanamycin) selection plates at 37°C. Only cells containing a plasmid with an interrupted ccdB gene will grow after transformation [Grant, S.G.N., Proc. Natl. Acad. Sci. USA 87:4645-4649 (1990)1. Transformed colonies are picked and grown up in 3 ml of LB-Kan broth at 37°C. Plasmid DNA is isolated by using a QIAprep ® (Qiagen Inc., Santa Clarita, CA) procedure, as directed by the manufacturer. The DNA is cut with EcoRI or SnaBI, and NotI restriction enzymes to release the PS213 insert fragment.
  • QIAprep ® Qiagen Inc., Santa Clarita, CA
  • the fragment is run on 1% Seakem ® LE agarose/0.5 ⁇ g/ml ethidium bromide/TE gel, visualized by UV irradiation, excised and purified using QIAquickTM (Qiagen Inc., Santa Clarita, CA) procedures, as directed by the supplier's instructions.
  • QIAquickTM Qiagen Inc., Santa Clarita, CA
  • the pcDNA3.1/Myc-His plasmid DNA is linearized by digestion with EcoRI or SnaBI, and NotI in the polylinker region of the plasmid DNA.
  • the resulting plasmid DNA backbone allows insertion of the PS213 purified cDNA fragment, supra, downstream of a CMV promoter which directs expression of the proteins in mammalian cells.
  • the ligated plasmid is transformed into DH5 alphaTM cells (GibcoBRL Grand Island, NY), as directed by the manufacturer. Briefly, 10 ng of pcDNA3.1/Myc-His containing a PS213 insert are added to 50 ⁇ l of competent DH5 alpha cells, and the contents are mixed gently.
  • the mixture is incubated on ice for 30 min, heat shocked for 20 sec at 37°C, and placed on ice for an additional 2 min.
  • Upon addition of 0.95 ml of LB medium the mixture is incubated for 1 hr at 37°C while shaking at 225 rpm.
  • the transformed cells then are plated onto 100 mm LB/Amp (50 ⁇ g/ml ampicillin) plates and grown at 37°C. Colonies are picked and grown in 3 ml of LB/Amp broth. Plasmid DNA is purified using a QIAprep Kit. The presence of the insert is confirmed using techniques known to those skilled in the art, including, but not limited to restriction digestion and gel analysis. (J. Sambrook et al., supra. )
  • the PS213 expression plasmid described in section A, supra is retransformed into DH5 alpha cells, plated onto LB/ampicillin agar, and grown up in 10 ml of LB/ampicillin broth, as described hereinabove.
  • the plasmid is purified using a QIAfilterTM Maxi Kit (Qiagen, Chatsworth, CA) and is transfected into HEK293 cells [F.L. Graham et al., J. Gen. Vir. 36:59-72 (1977)1. These cells are available from the A.T.C.C, 12301 Parklawn Drive, Rockville, MD 20852, under Accession No. CRL 1573.
  • Transfection is carried out using the cationic lipofectamine-mediated procedure described by P. Hawley-Nelson et al., Focus 15.73 (1993). Particularly, HEK293 cells are cultured in 10 ml DMEM media supplemented with 10% fetal bovine serum (FBS), L-glutamine (2 mM) and freshly seeded into 100 mm culture plates at a density of 9 x 10 6 cells per plate. The cells are grown at 37 °C to a confluency of between 70% and 80% for transfection.
  • FBS fetal bovine serum
  • L-glutamine 2 mM
  • Opti-MEM I ® medium (Gibco-BRL, Grand Island, NY), and 48-96 ⁇ l of LipofectamineTM Reagent (Gibco-BRL, Grand
  • Opti-MEM I media 800 ⁇ l portion of Opti-MEM I media.
  • the two solutions are mixed and incubated at room temperature for 15-30 min.
  • the culture medium is removed from the cells, the cells are washed once with 10 ml of serum-free DMEM.
  • the Opti-MEM I-Lipofectamine-plasmid DNA solution is diluted with 6.4 ml of serum-free DMEM and then overlaid onto the cells.
  • the cells are incubated for 5 hr at 37°C, after which time, an additional 8 ml of DMEM with 20% FBS are added.
  • the old medium is aspirated, and the cells are overlaid with 5 ml of fresh DMEM with 5% FBS.
  • Supernatants and cell extracts are analyzed for PS213 gene activity 72 hr after transfection.
  • PS213 protein band can be visualized using Western blotting techniques with anti-myc epitope or anti-histidine monoclonal antibodies (Invitrogen, Carlsbad, CA) or anti-PS213 polyclonal serum (see Example 14).
  • the expressed PS213 recombinant protein can be analyzed by mass spectrometry (see Example 12).
  • Bound PS213 recombinant protein then is eluted from the column using either an excess of imidazole or histidine, or a low pH buffer.
  • the recombinant protein can also be purified by binding at the myc-his sequence to an affinity column consisting of either anti-myc or anti-histidine monoclonal antibodies conjugated through a hydrazide or other linkage to an agarose resin and eluting with an excess of myc peptide or histidine, respectively.
  • the purified recombinant protein can then be covalently cross-linked to a solid phase, such as N-hydroxysuccinimide-activated sepharose columns
  • pcDNA3.1/Myc-His is utilized in this example, it is known to those skilled in the art that other comparable expression systems can be utilized herein with appropriate modifications in reagent and/or techniques and are within the skill of one of ordinary skill in the art.
  • the largest cloned insert containing the coding region of the PS213 gene is sub-cloned into either (i) a eukaryotic expression vector which may contain, for example, a cytomegalovirus (CMV) promoter and/or protein fusible sequences which aid in protein expression and detection, or (ii) a bacterial expression vector containing a superoxide-dismutase (SOD) and CMP-KDO synthetase (CKS) or other protein fusion gene for expression of the protein sequence.
  • CMV cytomegalovirus
  • SOD superoxide-dismutase
  • CKS CMP-KDO synthetase
  • Example 12 Chemical Analysis of Prostrate Tissue Proteins A. Analysis of Trvptic Peptide Fragments Using MS. Sera from patients with prostrate disease, such as prostrate cancer, sera from patients with no prostrate disease, extracts of prostrate tissues or cells from patients with prostrate disease, such as prostrate cancer, extracts of prostrate tissues or cells from patients with no prostrate disease, and extracts of tissues or cells from other non-diseased or diseased organs of patients are run on a polyacrylamide gel using standard procedures and stained with Coomassie Blue. Sections of the gel suspected of containing the unknown polypeptide are excised and subjected to an in-gel reduction, acetamidation and tryptic digestion. P. Jeno et al., Anal. Bio.
  • the peptides are adsorbed to approximately 0.1 ⁇ l of POROS R2 sorbent (Perseptive Biosystems, Framingham, Massachusetts) trapped in the tip of a drawn gas chromatography capillary tube by dissolving them in 10 ⁇ l of 5% formic acid and passing it through the capillary.
  • the adsorbed peptides are washed with water and eluted with 5% formic acid in 60% methanol.
  • the eluant is passed directly into the spraying capillary of an API III mass spectrometer (Perkin- Elmer Sciex, Thornhill, Ontario, Canada) for analysis by nano-electrospray mass spectrometry. M. Wilm et al., Int. J. Mass Spectrom.
  • the polypeptides are electroblotted to a cationic membrane and stained with Coomassie Blue. Following staining, the membranes are washed and sections thought to contain the unknown polypeptides are cut out and dissected into small pieces. The membranes are placed in 500 ⁇ l microcentrifuge tubes and immersed in 10 to 20 ⁇ l of proteolytic digestion buffer (100 mM Tris-HCl, pH 8.2, containing 0.1 M NaCl, 10% acetonitrile, 2 mM
  • proteolytic digestion buffer 100 mM Tris-HCl, pH 8.2, containing 0.1 M NaCl, 10% acetonitrile, 2 mM
  • Example 13 Gene Immunization Protocol A. In Vivo Antigen Expression. Gene immunization circumvents protein purification steps by directly expressing an antigen jn vivo after inoculation of the appropriate expression vector. Also, production of antigen by this method may allow correct protein folding and glycosylation since the protein is produced in mammalian tissue.
  • the method utilizes insertion of the gene sequence into a plasmid which contains a CMV promoter, expansion and purification of the plasmid and injection of the plasmid DNA into the muscle tissue of an animal. Preferred animals include mice and rabbits. See, for example, H. Davis et al., Human Molecular Genetics 2:1847-1851 (1993).
  • PS213 cDN A sequences are generated from the PS213 cDNA-containing vector using appropriate PCR primers containing suitable 5' restriction sites following the procedures described in Example 11. The PCR product is cut with appropriate restriction enzymes and inserted into a vector which contains the CMV promoter (for example, pRc/CMV or pcDNA3 vectors from Invitrogen, San Diego, CA).
  • CMV promoter for example, pRc/CMV or pcDNA3 vectors from Invitrogen, San Diego, CA.
  • This plasmid then is expanded in the appropriate bacterial strain and purified from the cell lysate using a CsCl gradient or a Qiagen plasmid DNA purification column. All these techniques are familiar to one of ordinary skill in the art of molecular biology.
  • Example 14 Production of Antibodies against PS213 A. Production of Polyclonal Antisera. Antiserum against PS213 was prepared by injecting rabbits with peptides whose sequences were derived from that of the predicted amino acid sequence of the PS213 published nucleotide sequence (SEQUENCE ID NO 1). The synthesis of peptides (SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10) is described in Example 10. Peptides used as immunogens were not conjugated to a carrier such as keyhole limpet hemocyanine, KLH, (i.e., they were unconjugated.).
  • a carrier such as keyhole limpet hemocyanine, KLH
  • SEQUENCE ID NO 9 and SEQUENCE ID NO 10, were used to prepare the primary immunogen by emulsifying 0.5 ml of the peptide at a concentration of 2 mg/ml in PBS (pH 7.2) which contained 0.5 ml of complete Freund's adjuvant (CFA) (Difco, Detroit, MI).
  • CFA complete Freund's adjuvant
  • the immunogen was injected into several sites of the animal via subcutaneous, intraperitoneal, and intramuscular routes of administration. Four weeks following the primary immunization, a booster immunization was administered.
  • the immunogen used for the booster immunization dose was prepared by emulsifying 0.5 ml of the same unconjugated peptide used for the primary immunogen, except that the peptide now was diluted to 1 mg/ml with 0.5 ml of incomplete Freund's adjuvant (IF A) (Difco, Detroit, MI). Again, the booster dose was administered into several sites via subcutaneous, intraperitoneal and intramuscular types of injections. The animals were bled (5 ml) two weeks after the booster immunizations and each serum was tested for immunoreactivity to the peptide as described below. The booster and bleed schedule was repeated at 4 week intervals until an adequate titer was obtained.
  • IF A incomplete Freund's adjuvant
  • the titer or concentration of antiserum was determined by using unconjugated peptides in a microtiter EIA as described in Example 17, below.
  • An antibody titer of 1 :500 or greater was considered an adequate titer for further use and study.
  • mice are immunized using peptides which can either be conjugated to a carrier such as KLH [prepared as described hereinbelow, or unconjugated (i.e., not conjugated to a carrier such as KLH)] except that the amount of the unconjugated or conjugated peptide for monoclonal antibody production in mice is one-tenth the amount used to produce polyclonal antisera in rabbits.
  • a carrier such as KLH [prepared as described hereinbelow, or unconjugated (i.e., not conjugated to a carrier such as KLH)] except that the amount of the unconjugated or conjugated peptide for monoclonal antibody production in mice is one-tenth the amount used to produce polyclonal antisera in rabbits.
  • the primary immunogen consists of 100 ⁇ g of unconjugated or conjugated peptide in 0.1 ml of CFA emulsion while the immunogen used for booster immunizations consists of 50
  • Hybridomas for the generation of monoclonal antibodies are prepared and screened using standard techniques.
  • the methods used for monoclonal antibody development follow procedures known in the art such as those detailed in Kohler and Milstein, Nature 256:494 (1975) and reviewed in J.G.R. Hurrel, ed., Monoclonal Hybridoma Antibodies: Techniques and Applications. CRC Press, Inc., Boca Raton, FL (1982).
  • Another method of monoclonal antibody development which is based on the Kohler and Milstein method is that of L.T. Mimms et al., Virology 176:604-619 (1990).
  • the immunization regimen (per mouse) consists of a primary immunization with additional booster immunizations.
  • the primary immunogen used for the primary immunization consists of 100 ⁇ g of unconjugated or conjugated peptide in 50 ⁇ l of PBS (pH 7.2) previously emulsified in 50 ⁇ l of CFA.
  • Booster immunizations performed at approximately two weeks and four weeks post primary immunization consist of 50 ⁇ g of unconjugated or conjugated peptide in 50 ⁇ l of PBS (pH 7.2) emulsified with 50 ⁇ l IFA.
  • a total of 100 ⁇ l of this immunogen are inoculated intraperitoneally and subcutaneously into each mouse.
  • mice are screened for immune response by microtiter plate enzyme immunoassay (EIA) as described in Example 17 approximately four weeks after the third immunization.
  • EIA microtiter plate enzyme immunoassay
  • splenocytes are fused with, for example, Sp2/0-Agl4 myeloma cells (Milstein Laboratories, England) using the polyethylene glycol (PEG) method.
  • the fusions are cultured in Iscove's Modified Dulbecco's Medium (IMDM) containing 10% fetal calf serum (FCS), plus 1 % hypoxan thine, aminopterin and thymidine (HAT).
  • IMDM Iscove's Modified Dulbecco's Medium
  • FCS fetal calf serum
  • HAT aminopterin and thymidine
  • Clones reactive with the peptide used an immunogen and non-reactive with other peptides are selected for final expansion. Clones thus selected are expanded, aliquoted and frozen in IMDM containing 10% FCS and 10% dimethyl sulfoxide, (DMSO).
  • Peptide Conjugation Peptide is conjugated to maleimide activated KLH (commercially available as Imject ® , available from Pierce Chemical Company, Rockford, IL). Imject ® contains about 250 moles of reactive maleimide groups per mole of hemocyanine.
  • the activated KLH is dissolved in phosphate buffered saline (PBS, pH 8.4) at a concentration of about 7.7 mg/ml.
  • PBS phosphate buffered saline
  • the peptide is conjugated through cysteines occurring in the peptide sequence, or to a cysteine previously added to the synthesized peptide in order to provide a point of attachment.
  • the peptide is dissolved in DMSO (Sigma Chemical Company, St.
  • conjugation reaction described hereinbelow is based on obtaining 3 mg of KLH peptide conjugate ("conjugated peptide"), which contains about 0.77 ⁇ moles of reactive maleimide groups. This quantity of peptide conjugate usually is adequate for one primary injection and four booster injections for production of polyclonal antisera in a rabbit.
  • conjugated peptide usually is adequate for one primary injection and four booster injections for production of polyclonal antisera in a rabbit.
  • peptide is dissolved in DMSO at a concentration of 1.16 ⁇ moles/100 ⁇ l of DMSO.
  • One hundred microliters (100 ⁇ l) of the DMSO solution are added to 380 ⁇ l of the activated KLH solution prepared as described hereinabove, and 20 ⁇ l of PBS (pH 8.4) are added to bring the volume to 500 ⁇ l.
  • the reaction is incubated overnight at room temperature with stirring.
  • the extent of reaction is determined by measuring the amount of unreacted thiol in the reaction mixture.
  • the difference between the starting concentration of thiol and the final concentration is assumed to be the concentration of peptide which has coupled to the activated KLH.
  • the amount of remaining thiol is measured using Ellman's reagent (5,5'-dithiobis(2-nitrobenzoic acid), Pierce Chemical Company, Rockford, IL).
  • Cysteine standards are made at a concentration of 0, 0.1, 0.5, 2, 5 and 20 mM by dissolving 35 mg of cysteine HC1 (Pierce Chemical Company, Rockford, IL) in 10 ml of PBS (pH 7.2) and diluting the stock solution to the desired concentration(s).
  • the photometric determination of the concentration of thiol is accomplished by placing 200 ⁇ l of PBS (pH 8.4) in each well of an Immulon 2 ® microwell plate (Dynex Technologies, Chantilly, VA). Next, 10 ⁇ l of standard or reaction mixture are added to each well. Finally, 20 ⁇ l of Ellman's reagent at a concentration of 1 mg/ml in PBS (pH 8.4) are added to each well. The wells are incubated for 10 minutes at room temperature, and the absorbance of all wells is read at 415 nm with a microplate reader (such as the
  • Frozen hybridoma cells prepared as described hereinabove are thawed and placed into expansion culture.
  • Viable hybridoma cells are inoculated intraperitoneally into Pristane treated mice.
  • Ascitic fluid is removed from the mice, pooled, filtered through a 0.2 ⁇ filter and subjected to an immunoglobulin class G (IgG) analysis to determine the volume of the Protein A column required for the purification.
  • IgG immunoglobulin class G
  • the volume of the Protein A column is determined by the quantity of IgG present in the ascites fluid.
  • the eluate then is dialyzed against PBS (pH 7.2) overnight at 2-8°C.
  • the dialyzed monoclonal antibody is sterile filtered and dispensed in aliquots.
  • the immunoreactivity of the purified monoclonal antibody is confirmed by determining its ability to specifically bind to the peptide used as the immunogen by use of the EIA microtiter plate assay procedure of Example 17.
  • the specificity of the purified monoclonal antibody is confirmed by determining its lack of binding to irrelevant peptides such as peptides of PS213 not used as the immunogen.
  • the purified anti-PS213 monoclonal thus prepared and characterized is placed at either 2-8°C for short term storage or at -80°C for long term storage. 5. Further Characterization of Monoclonal Antibody.
  • the isotype and subtype of the monoclonal antibody produced as described hereinabove can be determined using commercially available kits (available from Amersham. Inc., Arlington Heights, IL). Stability testing also can be performed on the monoclonal antibody by placing an aliquot of the monoclonal antibody in continuous storage at 2-8°C and assaying optical density (OD) readings throughout the course of a given period of time.
  • Immune sera obtained as described hereinabove in Examples 13 and/or 14, is affinity purified using immobilized synthetic peptides prepared as described in Example 10, or recombinant proteins prepared as described in Example 11.
  • An IgG fraction of the antiserum is obtained by passing the diluted, crude antiserum over a Protein A column (Affi-Gel protein A, Bio-Rad, Hercules, CA). Elution with a buffer (Binding Buffer, supplied by the manufacturer) removes substantially all proteins that are not immunoglobulins. Elution with 0.1M buffered glycine (pH 3) gives an immunoglobulin preparation that is substantially free of albumin and other serum proteins.
  • Immunoaffinity chromatography is performed to obtain a preparation with a higher fraction of specific antigen-binding antibody.
  • the peptide used to raise the antiserum is immobilized on a chromatography resin, and the specific antibodies directed against its epitopes are adsorbed to the resin. After washing away non- binding components, the specific antibodies are eluted with 0.1 M glycine buffer, pH 2.3. Antibody fractions are immediately neutralized with 1.0M Tris buffer (pH 8.0) to preserve immunoreactivity.
  • the chromatography resin chosen depends on the reactive groups present in the peptide.
  • a resin such as Affi-Gel 10 or Affi-Gel 15 is used (Bio-Rad, Hercules, CA). If coupling through a carboxy group on the peptide is desired, Affi-Gel 102 can be used (Bio-Rad, Hercules, CA). If the peptide has a free sulfhydryl group, an organomercurial resin such as Affi-Gel 501 can be used (Bio-Rad, Hercules, CA). Alternatively, spleens can be harvested and used in the production of hybridomas to produce monoclonal antibodies following routine methods known in the art as described hereinabove.
  • Example 16 Western Blotting of Tissue Samples Protein extracts were prepared by homogenizing tissue samples in 0.1 M Tris-HCl (pH 7.5), 15% (w/v) glycerol, 0.2 mM EDTA, 1.0 mM 1 ,4- dithiothreitol, 10 ⁇ g/ml leupeptin, 0.1% Triton X-100 , and 1.0 mM phenylmethylsulfonylfluoride [Kain et al., Biotechniques. 17:982 (1994)]. Following homogenization, the homogenates were centrifuged at 4°C for 5 minutes to separate supernatant from debris. For protein quantitation, 3-10 ⁇ l of supernatant were added to 1.5 ml of bicinchoninic acid reagent (Sigma, St. Louis, MO), and the resulting absorbance at 562 nm were measured.
  • Tris-HCl pH 7.5
  • 0.2 mM EDTA 1.0
  • Chemiluminesent bands were visualized by exposing the developed membranes to Hyperfilm ECL (Amersham, Arlington Heights, IL). The bands were also visualized directly on the membranes by the addition and development of chromogenic substrate 5-bromo-4-chloro-3-indolyl phosphate (BCIP). This chromogenic solution contains 0.016% BCIP, 100 mM NaCl, 5 mM MgCl2, and 100 mM Tris-HCl, pH 9.5. The filter was incubated in the chromogenic solution at room temperature until the bands developed to the desired intensity. Molecular mass determination was made based upon the mobility of pre- stained molecular weight standards (Novex, San Diego, CA) and biotinylated molecular weight standards (Tropix, Bedford, MA).
  • FIG 1 shows the results of the Western blot performed on a panel of tissue extracts using antiserum against PS213 synthetic peptide, SEQUENCE ID NO 10 (see Example 14).
  • Each lane of Figure 1 represents a different tissue protein extract: 1, endometrial cancer; 2, ovarian cancer; 3, normal bladder; 4, lung cancer; 5, normal colon; 6, BPH; 7, BPH; 8, prostate cancer; 9, blank; 10 molecular weight markers (kD).
  • a strong band appears between 148 kD and 60 kD in the prostate cancer (lane 8, indicated by an arrow) and one of the two BPH extracts (lane 6).
  • a faint band is seen at the same position in the other BPH sample (lane 7) and also in the lung cancer (lane 4).
  • Example 17 EIA Microtiter Plate Assay The immunoreactivity of antiserum preferably obtained from rabbits as described in Example 14 was determined by means of a microtiter plate EIA, as follows. Briefly, synthetic peptides, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, and SEQUENCE ID NO 10, prepared as described in Example 10, were dissolved in carbonate buffer (50 mM, pH 9.6) to a final concentration of 2 ⁇ g/ml. Next, 100 ⁇ l of the peptide or protein solution were placed in each well of an Immulon 2 ® microtiter plate (Dynex Technologies, Chantilly, VA). The plate was incubated overnight at room temperature and then washed four times with deionized water.
  • Immulon 2 ® microtiter plate Dynex Technologies, Chantilly, VA
  • a suitable protein blocking agent such as Superblock ® (Pierce Chemical Company, Rockford, IL)
  • Superblock ® Pieris Chemical Company, Rockford, IL
  • a protein blocking agent e.g., a 3% Superblock ® solution
  • Tween-20 ® monolaurate polyoxyethylene ether
  • Example 18 Coating of Solid Phase Particles
  • One coating method comprises coating ED AC (l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (Aldrich Chemical Co., Milwaukee, WI) activated carboxylated latex microparticles with antibodies which specifically bind to PS213 protein, as follows. Briefly, a final 0.375% solid suspension of resin washed carboxylated latex microparticles (available from Bangs Laboratories, Carmel, IN or Serodyn, Indianapolis, IN) are mixed in a solution containing 50 mM MES buffer, pH 4.0 and 150 mg/1 of affinity purified anti-PS213 antibody (see Example 14) for 15 min in an appropriate container. ED AC coupling agent is added to a final concentration of 5.5 ⁇ g/ml to the mixture and mixed for 2.5 hr at room temperature.
  • ED AC l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • microparticles then are washed with 8 volumes of a Tween 20 ® /sodium phosphate wash buffer (pH 7.2) by tangential flow filtration using a 0.2 ⁇ m Microgon Filtration module. Washed microparticles are stored in an appropriate buffer which usually contains a dilute surfactant and irrelevant protein as a blocking agent, until needed.
  • Antibodies which specifically bind to PS213-antigen also may be coated on the surface of 1/4 inch polystyrene beads by routine methods known in the art (Snitman et al., US Patent 5,273,882) and used in competitive binding or EIA sandwich assays.
  • Polystyrene beads first are cleaned by ultrasonicating them for about 15 seconds in 10 mM NaHCO3 buffer at pH 8.0. The beads then are washed in deionized water until all fines are removed. Beads then are immersed in an antibody solution in 10 mM carbonate buffer, pH 8 to 9.5.
  • the antibody solution can be as dilute as 1 ⁇ g/ml in the case of high affinity monoclonal antibodies or as concentrated as about 500 ⁇ g/ml for polyclonal antibodies which have not been affinity purified.
  • Beads are coated for at least 12 hours at room temperature, and then they are washed with deionized water. Beads may be air dried or stored wet (in PBS, pH 7.4). They also may be overcoated with protein stabilizers (such as sucrose) or protein blocking agents used as non-specific binding blockers (such as irrelevant proteins, Carnation skim milk, Superblock ® , or the like).
  • Example 19 Microparticle Enzyme Immunoassay (MEIA)
  • PS213 antigens are detected in patient test samples by performing a standard antigen competition EIA or antibody sandwich EIA and utilizing a solid phase such as microparticles (MEIA).
  • the assay can be performed on an automated analyzer such as the IMx ® Analyzer (Abbott Laboratories, Abbott Park, IL).
  • A. Antibody Sandwich EIA Briefly, samples suspected of containing PS213 antigen are incubated in the presence of anti-PS213 antibody-coated microparticles (prepared as described in Example 17) in order to form antigen/antibody complexes. The microparticles then are washed and an indicator reagent comprising an antibody conjugated to a signal generating compound (i.e., enzymes such as alkaline phosphatase or horseradish peroxide) is added to the antigen/antibody complexes or the microparticles and incubated.
  • a signal generating compound i.e., enzymes such as alkaline phosphatase or horseradish peroxide
  • the microparticles are washed and the bound antibody/antigen/antibody complexes are detected by adding a substrate (e.g., 4-methyl umbelliferyl phosphate (MUP), or OPD/peroxide, respectively), that reacts with the signal generating compound to generate a measurable signal.
  • a substrate e.g., 4-methyl umbelliferyl phosphate (MUP), or OPD/peroxide, respectively
  • MUP 4-methyl umbelliferyl phosphate
  • OPD/peroxide e.g., OPD/peroxide
  • the competitive binding assay uses a peptide or protein that generates a measurable signal when the labeled peptide is contacted with an anti-peptide antibody coated microparticle. This assay can be performed on the IMx ® Analyzer (available from Abbott Laboratories, Abbott Park, IL).
  • the labeled peptide is added to the PS213 antibody -coated microparticles (prepared as described in Example 17) in the presence of a test sample suspected of containing PS213 antigen, and incubated for a time and under conditions sufficient to form labeled PS213 peptide (or labeled protein) / bound antibody complexes and/or patient PS213 antigen / bound antibody complexes.
  • PS213 antigen in the test sample competes with the labeled PS213 peptide (or PS213 protein) for binding sites on the microparticle.
  • PS213 antigen in the test sample results in a lowered binding of labeled peptide and antibody coated microparticles in the assay since antigen in the test sample and the PS213 peptide or PS213 protein compete for antibody binding sites.
  • a lowered signal indicates the presence of PS213 antigen in the test sample.
  • the presence of PS213 antigen suggests the diagnosis of a prostrate disease or condition, such as prostrate cancer.
  • the PS213 polynucleotides and the proteins encoded thereby which are provided and discussed hereinabove are useful as markers of prostrate tissue disease, especially prostrate cancer. Tests based upon the appearance of this marker in a test sample such as blood, plasma or serum can provide low cost, non- invasive, diagnostic information to aid the physician to make a diagnosis of cancer, to help select a therapy protocol, or to monitor the success of a chosen therapy.
  • This marker may appear in readily accessible body fluids such as blood, urine or stool as antigens derived from the diseased tissue which are detectable by immunological methods. This marker may be elevated in a disease state, altered in a disease state, or be a normal protein of the prostrate which appears in an inappropriate body compartment.
  • Example 20 Immunohistochemical Detection of PS213 Protein Antiserum against a PS213 synthetic peptide derived from the consensus peptide sequence (SEQUENCE ED NO 10) described in Example 14, above, is to immunohistochemically stain a variety of normal and diseased tissues using standard procedures. Briefly, frozen blocks of tissue are cut into 6 micron sections, and placed on microscope slides. After fixation in cold acetone, the sections are dried at room temperature, then washed with phosphate buffered saline and blocked.
  • SEQUENCE ED NO 10 consensus peptide sequence
  • the slides are incubated with the antiserum against a synthetic peptide derived from the consensus PS213 peptide sequence (SEQUENCE ED NO 10) at a dilution of 1:500, washed, incubated with biotinylated goat anti-rabbit antibody, washed again, and incubated with avidin labeled with horseradish peroxidase. After a final wash, the slides are incubated with 3-amino-9- ethylcarbazole substrate which gives a red stain. The slides are counterstained with hematoxylin, mounted, and examined under a microscope by a pathologist.

Abstract

Sequences useful for the detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining the predisposition of an individual to diseases and conditions of the prostate, such as prostate cancer, are described. These sequences are designated as PS213 sequences herein. Also provided are antibodies which specifically bind to PS213-encoded polypeptide or protein, and agonists or inhibitors which prevent action of the tissue-specific PS213 polypeptide, which molecules are useful for the therapeutic treatment of prostate diseases, tumors or metastases.

Description

REAGENTS AND METHODS USEFUL FOR DETECTING DISEASES OF THE PROSTATE
Background of the Invention
This invention relates generally to detecting diseases of the prostate. Furthermore, the invention also relates to reagents and methods for detecting diseases of the prostate. More particularly, the present invention relates to reagents such as polynucleotide sequences and the polypeptide sequences encoded thereby, as well as methods which utilize these sequences. The polynucleotide and polypeptide sequences are useful for detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining predisposition to diseases or conditions of the prostate such as prostate cancer. Prostate cancer is the most common form of cancer occurring in males in the United States, with projections of 184,500 new cases diagnosed and 39,200 related deaths to occur during 1998 (American Cancer Society). Prostate cancer also has shown the largest increase in incidence as compared to other types of cancer, increasing 142% from 1992 to 1996. Procedures used for detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining predisposition to diseases or conditions of the prostate such as prostate cancer are of critical importance to the outcome of the patient. For example, patients diagnosed with localized prostate cancer have greater than a 90% five-year survival rate compared to a rate of 25 to 31 % for patients diagnosed with distant metastasis. (American Cancer Society statistics). A diagnostic procedure for early detection of prostate cancer should, therefore, specifically detect this disease and be capable of detecting the presence of prostate cancer before symptoms appear.
Such procedures could include assays based upon the appearance of various disease markers in test samples such as blood, plasma, serum, or urine obtained by minimally invasive procedures which are detectable by immunological methods. These procedures would provide information to aid the physician in managing the patient with disease of the prostate and at low cost to the patient. Markers such as the prostate specific antigen (PSA) exist and are used clinically for screening patients for prostate cancer. Elevated levels of PSA protein in serum can be used as a marker in the early detection of prostate cancer in asymptomatic men. G.E. Hanks, et al., In: Cancer: Principles and Practice of Oncology. Vol. 1, Fourth Edition, pp. 1073-1 113, Philadelphia, PA: J.B. Lippincott Co. (1993.). PSA normally is secreted by the prostate at high levels into the seminal fluid, but is present in very low levels in the blood of men with normal prostates. However, in patients with diseases of the prostate including benign prostatic hyperplasia (BPH) and adenocarcinoma of the prostate, the level of PSA can be markedly elevated in the blood and thus be useful as an indicator of prostate disease. PSA, however, cannot differentiate between BPH and prostate cancer, which reduces its specificity as a marker for prostate cancer. M.K. Schwartz, et al., In: Cancer: Principles and Practice of Oncology. Vol. 1, Fourth Edition, pp. 531- 542, Philadelphia, PA: J.B. Lippincott Co. 1993. New markers which are more specific for prostate cancer thus would be beneficial in the initial detection of this disease.
A critical step in managing patients with prostate cancer is the presurgical staging of the cancer to provide prognostic value and criteria for designing optimal therapy. Improved procedures for accurately staging prostate cancer prior to surgery are needed. One study demonstrated that current methods of staging prostate cancer prior to surgery were incorrect approximately fifty percent (50%) of the time. F. Labrie, et al., Urology 44 (Symposium Issue):29-37 (1994). Prostate cancer management also could be improved by utilizing new markers found in an inappropriate body compartment. Such markers could be mRNA or protein markers expressed by cells originating from the primary prostate tumor but residing in blood, bone marrow or lymph nodes and could be sensitive indicators for metastasis to these distal organs. For example, in patients with metastatic prostate cancer, PSA protein has been detected by immunohistochemical techniques in bone marrow, and PSA mRNA has been detected by RT-PCR in cells of blood, lymph nodes and bone marrow. K. Pantel, et al., Onkologie 18:394-401 (1995).
New markers which could predict the biologic behavior of early prostate cancers would also be of significant value. Early prostate cancers that threaten or will threaten the life of the patient are more clinically important than those that do not or will not be a threat. G.E. Hanks, supra. A need therefore exists for new markers which can differentiate between the clinically important and unimportant prostate cancers. Such markers would allow the clinician to accurately identify and effectively treat early cancers localized to the prostate which could otherwise metastasize and kill the patient. Further, if one could show that such a marker characteristic of aggressive cancer was absent, the patient could be spared expensive and non-beneficial treatment. It also would be beneficial to find a prostate associated marker which is more sensitive in detecting recurrence of prostate cancer than PSA and which is not affected by androgens. To date, PSA has proven to be the most sensitive marker for detecting recurrent disease. However, in some cases tumor progression occurs without PSA elevation due to hormonal therapy utilized for treating the cancer. Although the decrease in androgen results in a concomitant decrease in PSA, it does not necessarily reflect a decrease in tumor metastasis. This complication is the result of androgen-stimulated PSA expression. Part of the decline in PSA observed after androgen ablation is due not to tumor cell death but to diminished PSA expression. G.E. Hanks, supra.
It therefore would be advantageous to provide specific methods and reagents for detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining predisposition to diseases and conditions of the prostate, or to indicate possible predisposition to these conditions. Such methods would include assaying a test sample for products of a gene which are overexpressed in prostate diseases and conditions such as cancer. Such methods may also include assaying a test sample for products of a gene alteration associated with prostate disease or condition. Such methods may further include assaying a test sample for products of a gene whose distribution among the various tissues and compartments of the body have been altered by a prostate- associated disease or condition such as cancer. Useful reagents include polynucleotide(s), or fragment(s) thereof which may be used in diagnostic methods such as reverse transcriptase-polymerase chain reaction (RT-PCR), PCR, or hybridization assays of mRNA extracted from biopsied tissue, blood or other test samples; polypeptides or proteins which are the translation products of such mRNAs; or antibodies directed against these polypeptides or proteins. Drug treatment or gene therapy for diseases or conditions of the prostate can then be based on these identified gene sequences or their expressed proteins and efficacy of any particular therapy can be monitored. Furthermore, it would be advantageous to have available alternative, non-surgical diagnostic methods capable of detecting early stage prostate disease such as cancer.
Summary of the Invention
This invention relates to uses for a newly discovered polynucleotide (depicted in SEQUENCE ID NO 1 and described by Brash, A. R. et al., (1997) Proc Natl Acad Sci USA 94(12):6148-6152) and polypeptide encoded by this polynucleotide(depicted in SEQUENCE ID NO 6 herein and also described by Brash et al., supra). This polynucleotide represents a second human 15S- lipoxygenase. This polynucleotide and polypeptide will be hereinafter referred to as PS213.
The present invention provides a method of detecting a target PS213 polynucleotide in a test sample which comprises contacting the test sample with at least one PS213-specific polynucleotide, wherein the PS213 polynucleotide has at least 50% identity with (i) SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1 , or (iii) complements of (i) or (ii), and detecting the presence of the target PS213 polynucleotides in the test sample which bind to the PS213-specific polynucleotide. Also, the PS213-specific polynucleotide may be attached to a solid phase prior to performing the method.
The present invention also provides a method for detecting PS213 mRNA in a test sample, which comprises performing reverse transcription (RT) with at least one primer in order to produce cDNA, amplifying the cDNA so obtained using PS213 oligonucleotides as sense and antisense primers to obtain PS213 amplicon, and detecting the presence of the PS213 amplicon as an indication of the presence of PS213 mRNA in the test sample. The PS213 oligonucleotides utilized in the method have at least 50% identity with (i) SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1, or (iii) complements of (i) or (ii). Amplification can be performed by the polymerase chain reaction. Also, the test sample can be reacted with a solid phase prior to performing the method, prior to amplification or prior to detection. This reaction can be a direct or an indirect reaction. Further, the detection step can comprise utilizing a detectable label capable of generating a measurable signal. The detectable label can be attached to a solid phase. The present invention further provides a method of detecting a target PS213 polynucleotide in a test sample suspected of containing target PS213 polynucleotides, which comprises (a) contacting the test sample with at least one PS213 oligonucleotide as a sense primer and at least one PS213 oligonucleotide as an anti-sense primer, and amplifying same to obtain a first stage reaction product; (b) contacting the first stage reaction product with at least one other PS213 oligonucleotide to obtain a second stage reaction product, with the proviso that the other PS213 oligonucleotide is located 3' to the PS213 oligonucleotides utilized in step (a) and is complementary to the first stage reaction product; and (c) detecting the second stage reaction product as an indication of the presence of a target PS213 polynucleotide in the test sample. The PS213 oligonucleotides used in the method have at least 50% identity with (i) SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1, or (iii) complements of (i) or (ii). Amplification may be performed by the polymerase chain reaction. The test sample can be reacted either directly or indirectly with a solid phase prior to performing the method, or prior to amplification, or prior to detection. The detection step may also comprise utilizing a detectable label capable of generating a measurable signal; further, the detectable label can be attached to a solid phase. Test kits useful for detecting target PS213 polynucleotides in a test sample are also provided which comprise a container containing at least one PS213- specific polynucleotide. The polynucleotide has at least 50% identity with (i) SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1, or (iii) complements of (i) or (ii). These test kits further comprise containers with tools useful for collecting test samples (such as, for example, blood, urine, saliva and stool). Such tools include lancets and absorbent paper or cloth for collecting and stabilizing blood; swabs for collecting and stabilizing saliva; and cups for collecting and stabilizing urine or stool samples. Collection materials, such as, papers, cloths, swabs, cups, and the like, may optionally be treated to avoid denaturation or irreversible adsorption of the sample. The collection materials also may be treated with or contain preservatives, stabilizers or antimicrobial agents to help maintain the integrity of the specimens.
The present invention also provides a purified polynucleotide or fragment thereof derived from a PS213 gene. The purified polynucleotide encodes a polypeptide having at least 50% identity with a polypeptide having an amino acid sequence selected from the group consisting of SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof. Further, the purified polynucleotide can be produced by recombinant and/or synthetic techniques. The purified recombinant polynucleotide can be contained within a recombinant vector. The invention further comprises a host cell transfected with the recombinant vector.
The present invention further provides a recombinant expression system comprising a nucleic acid sequence that includes an open reading frame derived from PS213. The nucleic acid sequence is operably linked to a control sequence compatible with a desired host. Also provided is a cell transfected with this recombinant expression system.
The present invention also provides a polypeptide encoded by PS213. The polypeptide can be produced by recombinant technology, provided in purified form, or produced by synthetic techniques. The polypeptide comprises an amino acid sequence which has at least 50% identity with an amino acid sequence selected from the group consisting of SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof. Also provided is a specific binding molecule which specifically binds to at least one PS213 epitope. The specific binding molecule can be an antibody. The epitope is derived from an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
Test kits for determining the presence of PS213 antigen or anti-PS213 antibody in a test sample are also included. In one embodiment, the test kits comprise a container containing at least one PS213 polypeptide having at least 50% identity with an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8,
SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof. Further, the test kit can comprise a container with tools useful for collecting test samples (such as blood, urine, saliva, and stool). Such tools include lancets and absorbent paper or cloth for collecting and stabilizing blood; swabs for collecting and stabilizing saliva; and cups for collecting and stabilizing urine or stool samples. Collection materials such as papers, cloths, swabs, cups, and the like, may optionally be treated to avoid denaturation or irreversible adsorption of the sample. These collection materials also may be treated with or contain preservatives, stabilizers or antimicrobiol agents to help maintain the integrity of the specimens. Also, the polypeptide can be attached to a solid phase.
In another embodiment of the invention, specific binding molecules, such as antibodies or fragments thereof against the PS213 antigen, can be used to detect antigen, or for image localization of the antigen, in a patient for the purpose of detecting or diagnosing a disease or condition. Such antibodies can be polyclonal or monoclonal, or made by molecular biology techniques, and can be labeled with a variety of detectable labels, including but not limited to radioisotopes and paramagnetic metals. Furthermore, antibodies or fragments thereof, whether monoclonal, polyclonal, or made by molecular biology techniques, can be used as therapeutic agents for the treatment of diseases characterized by expression of the PS213 antigen. In the case of therapeutic applications, the antibody may be used without derivitization, or it may be derivitized with a cytotoxic agent such as a radioisotope, enzyme, toxin, drug, prodrug, or the like.
Another test kit for determining the presence of PS213 antigen or anti- PS213 antibody in a test sample comprises a container containing a specific binding molecule, such as an antibody, which specifically binds to a PS213 antigen, wherein the PS213 antigen comprises at least one PS213-encoded epitope. The PS213 antigen has at least about 50% identity to a sequence of a PS213- encoded antigen selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof. These test kits can further comprise containers with tools useful for collecting test samples (such as blood, urine, saliva, and stool). Such tools include lancets and absorbent paper or cloth for collecting and stabilizing blood; swabs for collecting and stabilizing saliva; cups for collecting and stabilizing urine or stool samples. Collection materials, such as papers, cloths, swabs, cups and the like, may optionally be treated to avoid denaturation or irreversible adsorption of the sample. These collection materials also may be treated with, or contain, preservatives, stabilizers or antimicrobial agents to help maintain the integrity of the specimens. The antibody can be attached to a solid phase.
A method for producing a polypeptide which contains at least one epitope of PS213 is provided, which method comprises incubating host cells transfected with an expression vector. This vector comprises a polynucleotide sequence encoding a polypeptide, wherein the polypeptide comprises an amino acid sequence having at least 50% identity with a PS213 amino acid sequence selected from the group consisting of SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
A method for detecting PS213 antigen in a test sample suspected of containing PS213 antigen also is provided. The method comprises contacting the test sample with a specific binding molecule, such as an antibody or fragment thereof, which specifically binds to at least one epitope of a PS213 antigen, for a time and under conditions sufficient for the formation of binding molecule/antigen complexes; and detecting the presence of such complexes as an indication of the presence of PS213 antigen in the test sample. The specific binding molecule can be attached to a solid phase antibody. Furthermore, the specific binding molecule specifically binds to at least one epitope of a PS213 antigen selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof. Another method is provided which detects antibodies which specifically bind to PS213 antigen in a test sample suspected of containing these antibodies. The method comprises contacting the test sample with a polypeptide which contains at least one PS213 epitope, wherein the PS213 epitope comprises an amino acid sequence having at least 50% identity with an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments. Contacting is performed for a time and under conditions sufficient to allow antigen/antibody complexes to form. The method further entails detecting complexes which contain the polypeptide. The polypeptide can be attached to a solid phase. Further, the polypeptide can be a recombinant protein or a synthetic peptide.
The present invention provides a cell transfected with a PS213 polynucleotide as described above.
A method for producing antibodies to PS213 antigen also is provided, which method comprises administering to an individual an isolated immunogenic polypeptide or fragment thereof, wherein the isolated immunogenic polypeptide comprises at least one PS213 epitope. The immunogenic polypeptide is administered in an amount sufficient to produce an immune response. The isolated, immunogenic polypeptide comprises an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof. Another method for producing antibodies which specifically bind to PS213 antigen is disclosed, which method comprises administering to an individual a plasmid comprising a nucleic acid sequence which encodes at least one PS213 epitope derived from an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof. The plasmid is administered in an amount such that the plasmid is taken up by cells in the individual and expressed at levels sufficient to produce an immune response.
Brief Description of the Drawing The following drawing is illustrative of embodiments of the invention and is not meant to limit the scope of the invention as encompassed by the claims.
Figure 1 shows the results of the Western blot performed on a panel of tissue protein extracts probed with antiserum against a PS213 synthetic peptide
(SEQUENCE ID NO 10).
Detailed Description of the Invention
The present invention provides methods for assaying a test sample for products of a prostate tissue gene designated as PS213, which comprises making cDNA from mRNA in the test sample, and detecting the cDNA as an indication of the presence of prostate tissue gene PS213. The method may include an amplification step, wherein one or more portions of the mRNA from PS213 corresponding to the gene or fragments thereof, is amplified. Methods also are provided for assaying for the translation products of PS213. Test samples which may be assayed by the methods provided herein include tissues, cells, body fluids and secretions. The present invention also provides reagents such as oligonucleotide primers and polypeptides which are useful in performing these methods. Portions of the nucleic acid sequences disclosed herein are useful as primers for the reverse transcription of RNA or for the amplification of cDNA; or as probes to determine the presence of certain mRNA sequences in test samples. Also disclosed are nucleic acid sequences which permit the production of encoded polypeptide sequences which are useful as standards or reagents in diagnostic immunoassays, as targets for pharmaceutical screening assays and/or as components or as target sites for various therapies. Monoclonal and polyclonal antibodies directed against at least one epitope contained within these polypeptide sequences are useful as delivery agents for therapeutic agents as well as for diagnostic tests and for screening for diseases or conditions associated with PS213, especially prostate cancer. Isolation of sequences of other portions of the gene of interest can be accomplished utilizing probes or PCR primers derived from these nucleic acid sequences. This allows additional probes of the mRNA or cDNA of interest to be established, as well as corresponding encoded polypeptide sequences. These additional molecules are useful in detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining the predisposition to diseases and conditions of the prostate, such as prostate cancer, characterized by PS213, as disclosed herein.
Techniques for determining amino acid sequence "similarity" are well- known in the art. In general, "similarity" means the exact amino acid to amino acid comparison of two or more polypeptides at the appropriate place, where amino acids are identical or possess similar chemical and/or physical properties such as charge or hydrophobicity. "Percent similarity" then can be determined between the compared polypeptide sequences. Techniques for determining nucleic acid and amino acid sequence identity also are well known in the art and include determining the nucleotide sequence of the mRNA for that gene (usually via a cDNA intermediate) and determining the amino acid sequence encoded thereby, and comparing this to a second amino acid sequence. In general, "identity" refers to an exact nucleotide to nucleotide or amino acid to amino acid correspondence of two polynucleotides or polypeptide sequences, respectively. Two or more polynucleotide sequences can be compared by determining their "percent identity." Two or more amino acid sequences likewise can be compared by determining their "percent identity." The percent identity of two sequences, whether nucleic acid or peptide sequences, is the number of exact matches between two aligned sequences divided by the length of the shorter sequences and multiplied by 100. An approximate alignment for nucleic acid sequences is provided by the local homology algorithm of Smith and Waterman, Advances in Applied Mathematics 2:482-489 (1981). This algorithm can be extended to use with peptide sequences using the scoring matrix developed by Dayhoff. Atlas of Protein Sequences and Structure. M.O. Dayhoff ed., 5 suppl. 3:353-358, National Biomedical Research Foundation, Washington, D.C., USA, and normalized by Gribskov, Nucl. Acids Res. 14(6): 6745-6763 (1986). An implementation of this algorithm for nucleic acid and peptide sequences is provided by the Genetics Computer Group (Madison, WI) in their BestFit utility application. The default parameters for this method are described in the Wisconsin Sequence Analysis Package, Program Manual. Version 8 (1995) (available from Genetics Computer Group, Madison, WI). Other equally suitable programs for calculating the percent identity or similarity between sequences are generally known in the art. The compositions and methods described herein will enable the identification of certain markers as indicative of a prostate tissue disease or condition; the information obtained therefrom will aid in the detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining diseases or conditions associated with PS213, especially prostate cancer. Test methods include, for example, probe assays which utilize the sequence(s) provided herein and which also may utilize nucleic acid amplification methods such as the polymerase chain reaction (PCR), the ligase chain reaction (LCR), and hybridization.
In addition, the nucleotide sequences provided herein contain open reading frames from which an immunogenic epitope may be found. This epitope is believed to be unique to the disease state or condition associated with PS213. It also is thought that the polynucleotides or polypeptides and protein encoded by the PS213 gene are useful as a marker. This marker is either elevated in disease such as prostate cancer, altered in disease such as prostate cancer, or present as a normal protein but appearing in an inappropriate body compartment. The uniqueness of the epitope may be determined by (i) its immunological reactivity and specificity with antibodies directed against proteins and polypeptides encoded by the PS213 gene, and (ii) its nonreactivity with any other tissue markers. Methods for determining immunological reactivity are well-known and include, but are not limited to, for example, radioimmunoassay (R A), enzyme-linked immunoabsorbent assay (ELISA), hemagglutination (HA), fluorescence polarization immunoassay (FPIA), chemiluminescent immunoassay (CLIA) and others. Several examples of suitable methods are described herein. Unless otherwise stated, the following terms shall have the following meanings:
A polynucleotide "derived from" or "specific for" a designated sequence refers to a polynucleotide sequence which comprises a contiguous sequence of approximately at least about 6 nucleotides, preferably at least about 8 nucleotides, more preferably at least about 10-12 nucleotides, and even more preferably at least about 15-20 nucleotides corresponding, i.e., identical or complementary to, a region of the designated nucleotide sequence. The sequence may be complementary or identical to a sequence which is unique to a particular polynucleotide sequence as determined by techniques known in the art.
Comparisons to sequences in databanks, for example, can be used as a method to determine the uniqueness of a designated sequence. Regions from which sequences may be derived, include but are not limited to, regions encoding specific epitopes, as well as non-translated and/or non-transcribed regions. The derived polynucleotide will not necessarily be derived physically from the nucleotide sequence of interest under study, but may be generated in any manner, including, but not limited to, chemical synthesis, replication, reverse transcription or transcription, which is based on the information provided by the sequence of bases in the region(s) from which the polynucleotide is derived. As such, it may represent either a sense or an antisense orientation of the original polynucleotide. In addition, combinations of regions corresponding to that of the designated sequence may be modified in ways known in the art to be consistent with the intended use.
A "fragment" of a specified polynucleotide refers to a polynucleotide sequence which comprises a contiguous sequence of approximately at least about 6 nucleotides, preferably at least about 8 nucleotides, more preferably at least about 10-12 nucleotides, and even more preferably at least about 15-20 nucleotides corresponding, i.e., identical or complementary to, a region of the specified nucleotide sequence. The term "primer" denotes a specific oligonucleotide sequence which is complementary to a target nucleotide sequence and used to hybridize to the target nucleotide sequence. A primer serves as an initiation point for nucleotide polymerization catalyzed by either DNA polymerase, RNA polymerase or reverse transcriptase. The term "probe" denotes a defined nucleic acid segment (or nucleotide analog segment, e.g., PNA as defined hereinbelow) which can be used to identify a specific polynucleotide present in samples bearing the complementary sequence. "Encoded by" refers to a nucleic acid sequence which codes for a polypeptide sequence, wherein the polypeptide sequence or a portion thereof contains an amino acid sequence of at least 3 to 5 amino acids, more preferably at least 8 to 10 amino acids, and even more preferably at least 15 to 20 amino acids from a polypeptide encoded by the nucleic acid sequence. Also encompassed are polypeptide sequences which are immunologically identifiable with a polypeptide encoded by the sequence. Thus, a "polypeptide," "protein," or "amino acid" sequence has at least about 50% identity, preferably about 60% identity, more preferably about 75-85% identity, and most preferably about 90-95% or more identity with a PS213 amino acid sequence. Further, the PS213 "polypeptide," "protein," or "amino acid" sequence may have at least about 60% similarity, preferably at least about 75% similarity, more preferably about 85% similarity, and most preferably about 95% or more similarity to a polypeptide or amino acid sequence of PS213. This amino acid sequence can be selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
A "recombinant polypeptide," "recombinant protein," or "a polypeptide produced by recombinant techniques," which terms may be used interchangeably herein, describes a polypeptide which by virtue of its origin or manipulation is not associated with all or a portion of the polypeptide with which it is associated in nature and/or is linked to a polypeptide other than that to which it is linked in nature. A recombinant or encoded polypeptide or protein is not necessarily translated from a designated nucleic acid sequence. It also may be generated in any manner, including chemical synthesis or expression of a recombinant expression system.
The term "synthetic peptide" as used herein means a polymeric form of amino acids of any length, which may be chemically synthesized by methods well- known to the routineer. These synthetic peptides are useful in various applications. The term "polynucleotide" as used herein means a polymeric form of nucleotides of any length, either ribonucleotides or deoxyribonucleotides. This term refers only to the primary structure of the molecule. Thus, the term includes double- and single-stranded DNA, as well as double- and single-stranded RNA. It also includes modifications, such as methylation or capping and unmodified forms of the polynucleotide. The terms "polynucleotide," "oligomer," "oligonucleotide," and "oligo" are used interchangeably herein.
"A sequence corresponding to a cDNA" means that the sequence contains a polynucleotide sequence that is identical or complementary to a sequence in the designated DNA. The degree (or "percent") of identity or complementarity to the cDNA will be approximately 50% or greater, preferably at least about 70% or greater, and more preferably at least about 90% or greater. The sequence that corresponds to the identified cDNA will be at least about 50 nucleotides in length, preferably at least about 60 nucleotides in length, and more preferably at least about 70 nucleotides in length. The correspondence between the gene or gene fragment of interest and the cDNA can be determined by methods known in the art and include, for example, a direct comparison of the sequenced material with the cDNAs described, or hybridization and digestion with single strand nucleases, followed by size determination of the digested fragments.
"Purified polynucleotide" refers to a polynucleotide of interest or fragment thereof which is essentially free, e.g., contains less than about 50%, preferably less than about 70%, and more preferably less than about 90%, of the protein with which the polynucleotide is naturally associated. Techniques for purifying polynucleotides of interest are well-known in the art and include, for example, disruption of the cell containing the polynucleotide with a chaotropic agent and separation of the polynucleotide(s) and proteins by ion-exchange chromatography, affinity chromatography and sedimentation according to density.
"Purified polypeptide" or "purified protein" means a polypeptide of interest or fragment thereof which is essentially free of, e.g., contains less than about 50%, preferably less than about 70%, and more preferably less than about 90%, cellular components with which the polypeptide of interest is naturally associated. Methods for purifying polypeptides of interest are known in the art.
The term "isolated" means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring). For example, a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or DNA or polypeptide, which is separated from some or all of the coexisting materials in the natural system, is isolated. Such polynucleotide could be part of a vector and/or such polynucleotide or polypeptide could be part of a composition, and still be isolated in that the vector or composition is not part of its natural environment.
"Polypeptide" and "protein" are used interchangeably herein and indicate at least one molecular chain of amino acids linked through covalent and or non- covalent bonds. The terms do not refer to a specific length of the product. Thus peptides, oligopeptides and proteins are included within the definition of polypeptide. The terms include post-translational modifications of the polypeptide, for example, glycosylations, acetylations, phosphorylations and the like. In addition, protein fragments, analogs, mutated or variant proteins, fusion proteins and the like are included within the meaning of polypeptide.
A "fragment" of a specified polypeptide refers to an amino acid sequence which comprises at least about 3-5 amino acids, more preferably at least about 8-10 amino acids, and even more preferably at least about 15-20 amino acids derived from the specified polypeptide.
"Recombinant host cells," "host cells," "cells," "cell lines," "cell cultures," and other such terms denoting microorganisms or higher eukaryotic cell lines cultured as unicellular entities refer to cells which can be, or have been, used as recipients for recombinant vector or other transferred DNA, and include the original progeny of the original cell which has been transfected.
As used herein "replicon" means any genetic element, such as a plasmid, a chromosome or a virus, that behaves as an autonomous unit of polynucleotide replication within a cell. A "vector" is a replicon in which another polynucleotide segment is attached, such as to bring about the replication and/or expression of the attached segment.
The term "control sequence" refers to a polynucleotide sequence which is necessary to effect the expression of a coding sequence to which it is ligated. The nature of such control sequences differs depending upon the host organism. In prokaryotes, such control sequences generally include a promoter, a ribosomal binding site and terminators; in eukaryotes, such control sequences generally include promoters, terminators and, in some instances, enhancers. The term "control sequence" thus is intended to include at a minimum all components whose presence is necessary for expression, and also may include additional components whose presence is advantageous, for example, leader sequences.
"Operably linked" refers to a situation wherein the components described are in a relationship permitting them to function in their intended manner. Thus, for example, a control sequence "operably linked" to a coding sequence is ligated in such a manner that expression of the coding sequence is achieved under conditions compatible with the control sequence.
The term "open reading frame" or "ORF" refers to a region of a polynucleotide sequence which encodes a polypeptide. This region may represent a portion of a coding sequence or a total coding sequence. A "coding sequence" is a polynucleotide sequence which is transcribed into mRNA and translated into a polypeptide when placed under the control of appropriate regulatory sequences. The boundaries of the coding sequence are determined by a translation start codon at the 5' -terminus and a translation stop codon at the 3' -terminus. A coding sequence can include, but is not limited to, mRNA, cDNA and recombinant polynucleotide sequences.
The term "immunologically identifiable with/as" refers to the presence of epitope(s) and polypeptide(s) which also are present in and are unique to the designated polypeptide(s). Immunological identity may be determined by antibody binding and/or competition in binding. These techniques are known to the routineer and also are described herein. The uniqueness of an epitope also can be determined by computer searches of known data banks, such as GenBank, for the polynucleotide sequence which encodes the epitope and by amino acid sequence comparisons with other known proteins.
As used herein, "epitope" means an antigenic determinant of a polypeptide or protein. Conceivably, an epitope can comprise three amino acids in a spatial conformation which is unique to the epitope. Generally, an epitope consists of at least five such amino acids and more usually, it consists of at least eight to ten amino acids. Methods of examining spatial conformation are known in the art and include, for example, x-ray crystallography and two-dimensional nuclear magnetic resonance.
A "conformational epitope" is an epitope that is comprised of a specific juxtaposition of amino acids in an immunologically recognizable structure, such amino acids being present on the same polypeptide in a contiguous or noncontiguous order or present on different polypeptides.
A polypeptide is "immunologically reactive" with an antibody when it binds to an antibody due to antibody recognition of a specific epitope contained within the polypeptide. Immunological reactivity may be determined by antibody binding, more particularly, by the kinetics of antibody binding, and/or by competition in binding using as competitor(s) a known polypeptide(s) containing an epitope against which the antibody is directed. The methods for determining whether a polypeptide is immunologically reactive with an antibody are known in the art.
As used herein, the term "immunogenic polypeptide containing an epitope of interest" means naturally occurring polypeptides of interest or fragments thereof, as well as polypeptides prepared by other means, for example, by chemical synthesis or the expression of the polypeptide in a recombinant organism. The term "transfection" refers to the introduction of an exogenous polynucleotide into a prokaryotic or eucaryotic host cell, irrespective of the method used for the introduction. The term "transfection" refers to both stable and transient introduction of the polynucleotide, and encompasses direct uptake of polynucleotides, transformation, transduction, and f-mating. Once introduced into the host cell, the exogenous polynucleotide may be maintained as a non-integrated replicon, for example, a plasmid, or alternatively, may be integrated into the host genome.
"Treatment" refers to prophylaxis and/or therapy. The term "individual" as used herein refers to vertebrates, particularly members of the mammalian species and includes, but is not limited to, domestic animals, sports animals, primates and humans; more particularly, the term refers to humans.
The term "sense strand" or "plus strand" (or "+") as used herein denotes a nucleic acid that contains the sequence that encodes the polypeptide. The term "antisense strand" or "minus strand" (or "-") denotes a nucleic acid that contains a sequence that is complementary to that of the "plus" strand.
The term "test sample" refers to a component of an individual's body which is the source of the analyte (such as antibodies of interest or antigens of interest). These components are well known in the art. A test sample is typically anything suspected of containing a target sequence. Test samples can be prepared using methodologies well known in the art such as by obtaining a specimen from an individual and, if necessary, disrupting any cells contained thereby to release target nucleic acids. These test samples include biological samples which can be tested by the methods of the present invention described herein and include human and animal body fluids such as whole blood, serum, plasma, cerebrospinal fluid, sputum, bronchial washing, bronchial aspirates, urine, lymph fluids, and various external secretions of the respiratory, intestinal and genitourinary tracts, tears, saliva, milk, white blood cells, myelomas and the like; biological fluids such as cell culture supernatants; tissue specimens which may be fixed; and cell specimens which may be fixed.
"Purified product" refers to a preparation of the product which has been isolated from the cellular constituents with which the product is normally associated and from other types of cells which may be present in the sample of interest. "PNA" denotes a "peptide nucleic acid analog" which may be utilized in a procedure such as an assay described herein to determine the presence of a target. "MA" denotes a "morpholino analog" which may be utilized in a procedure such as an assay described herein to determine the presence of a target. See, for example, U.S. Patent No. 5,378,841. PNAs are neutrally charged moieties which can be directed against RNA targets or DNA. PNA probes used in assays in place of, for example, the DNA probes of the present invention, offer advantages not achievable when DNA probes are used. These advantages include manufacturability, large scale labeling, reproducibility, stability, insensitivity to changes in ionic strength and resistance to enzymatic degradation which is present in methods utilizing DNA or RNA. These PNAs can be labeled with ("attached to") such signal generating compounds as fluorescein, radionucleotides, chemiluminescent compounds and the like. PNAs or other nucleic acid analogs such as MAs thus can be used in assay methods in place of DNA or RNA. Although assays are described herein utilizing DNA probes, it is within the scope of the routineer that PNAs or MAs can be substituted for RNA or DNA with appropriate changes if and as needed in assay reagents.
"Analyte," as used herein, is the substance to be detected which may be present in the test sample. The analyte can be any substance for which there exists a naturally occurring specific binding member (such as an antibody), or for which a specific binding member can be prepared. Thus, an analyte is a substance that can bind to one or more specific binding members in an assay. "Analyte" also includes any antigenic substances, haptens, antibodies and combinations thereof. As a member of a specific binding pair, the analyte can be detected by means of naturally occurring specific binding partners (pairs) such as the use of intrinsic factor protein as a member of a specific binding pair for the determination of Vitamin B12, the use of folate-binding protein to determine folic acid, or the use of a lectin as a member of a specific binding pair for the determination of a carbohydrate. The analyte can include a protein, a polypeptide, an amino acid, a nucleotide target and the like. The analyte can be soluble in a body fluid such as blood, blood plasma or serum, urine or the like. The analyte can be in a tissue, either on a cell surface or within a cell. The analyte can be on or in a cell dispersed in a body fluid such as blood, urine, breast aspirate, or obtained as a biopsy sample.
The terms "diseases of the prostate," "prostate disease," and "conditions of the prostate" are used interchangeably herein to refer to any disease or condition of the prostate including, but not limited to, benign prostatic hyperplasia (BPH), prostatitis, prostatic intraepithelial neoplasia (PIN) and cancer. "Prostate cancer," as used herein, refers to any malignant disease of the prostate including, but not limited to, adenocarcinoma, small cell undifferentiated carcinoma and mucinous (colloid) cancer.
An "Expressed Sequence Tag" or "EST" refers to the partial sequence of a cDNA insert which has been made by reverse transcription of mRNA extracted from a tissue followed by insertion into a vector.
A "transcript image" refers to a table or list giving the quantitative distribution of ESTs in a library and represents the genes active in the tissue from which the library was made. The present invention provides assays which utilize specific binding members. A "specific binding member," as used herein, is a member of a specific binding pair. That is, two different molecules where one of the molecules, through chemical or physical means, specifically binds to the second molecule. Therefore, in addition to antigen and antibody specific binding pairs of common immunoassays, other specific binding pairs can include biotin and avidin, carbohydrates and lectins, complementary nucleotide sequences, effector and receptor molecules, cofactors and enzymes, enzyme inhibitors, and enzymes and the like. Furthermore, specific binding pairs can include members that are analogs of the original specific binding members, for example, an analyte-analog.
Immunoreactive specific binding members include antigens, antigen fragments, antibodies and antibody fragments, both monoclonal and polyclonal and complexes thereof, including those formed by recombinant DNA molecules.
Specific binding members include "specific binding molecules." A "specific binding molecule" intends any specific binding member, particularly an immunoreactive specific binding member. As such, the term "specific binding molecule" encompasses antibody molecules (obtained from both polyclonal and monoclonal preparations), as well as, the following: hybrid (chimeric) antibody molecules (see, for example, Winter, et al, Nature 349:293-299 (1991), and U.S. Patent No. 4,816,567); F(ab')2 and F(ab) fragments; Fv molecules [non-covalent heterodimers, see, for example, Inbar, et al., Proc. Natl. Acad. Sci. USA 69:2659-2662 (1972), and Ehrlich, et al., Biochem. 19:4091-4096 (1980)]; single chain Fv molecules (sFv) [see, for example, Huston, et al., Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988)]; humanized antibody molecules (see, for example, Riechmann, et al., Nature 332:323-327 (1988), Verhoeyan, et al., Science
239: 1534-1536 (1988), and UK Patent Publication No. GB 2,276,169, published 21 September 1994)]; and any functional fragments obtained from such molecules, wherein such fragments retain immunological binding properties of the parent antibody molecule. The term "hapten," as used herein, refers to a partial antigen or non-protein binding member which is capable of binding to an antibody, but which is not capable of eliciting antibody formation unless coupled to a carrier protein.
A "capture reagent," as used herein, refers to an unlabeled specific binding member which is specific either for the analyte as in a sandwich assay, for the indicator reagent or analyte as in a competitive assay, or for an ancillary specific binding member, which itself is specific for the analyte, as in an indirect assay. The capture reagent can be directly or indirectly bound to a solid phase material before the performance of the assay or during the performance of the assay, thereby enabling the separation of immobilized complexes from the test sample.
The "indicator reagent" comprises a "signal-generating compound" ("label") which is capable of generating and generates a measurable signal detectable by external means, conjugated ("attached") to a specific binding member. In addition to being an antibody member of a specific binding pair, the indicator reagent also can be a member of any specific binding pair, including either hapten-anti-hapten systems such as biotin or anti-biotin, avidin or biotin, a carbohydrate or a lectin, a complementary nucleotide sequence, an effector or a receptor molecule, an enzyme cofactor and an enzyme, an enzyme inhibitor or an enzyme, and the like. An immunoreactive specific binding member can be an antibody, an antigen, or an antibody/antigen complex that is capable of binding either to the polypeptide of interest as in a sandwich assay, to the capture reagent as in a competitive assay, or to the ancillary specific binding member as in an indirect assay. When describing probes and probe assays, the term "reporter molecule" may be used. A reporter molecule comprises a signal generating compound as described hereinabove conjugated to a specific binding member of a specific binding pair, such as carbazole or adamantane.
The various "signal-generating compounds" (labels) contemplated include chromagens, catalysts such as enzymes, luminescent compounds such as fluorescein and rhodamine, chemiluminescent compounds such as dioxetanes, acridiniums, phenanthridiniums and luminol, radioactive elements and direct visual labels. Examples of enzymes include alkaline phosphatase, horseradish peroxidase, beta-galactosidase and the like. The selection of a particular label is not critical, but it must be capable of producing a signal either by itself or in conjunction with one or more additional substances.
"Solid phases" ("solid supports") are known to those in the art and include the walls of wells of a reaction tray, test tubes, polystyrene beads, magnetic or non-magnetic beads, nitrocellulose strips, membranes, microparticles such as latex particles, sheep (or other animal) red blood cells and Duracytes® (red blood cells "fixed" by pyruvic aldehyde and formaldehyde, available from Abbott Laboratories, Abbott Park, IL) and others. The "solid phase" is not critical and can be selected by one skilled in the art. Thus, latex particles, microparticles, magnetic or non-magnetic beads, membranes, plastic tubes, walls of microtiter wells, glass or silicon chips, sheep (or other suitable animal's) red blood cells and Duracytes® are all suitable examples. Suitable methods for immobilizing peptides on solid phases include ionic, hydrophobic, covalent interactions and the like. A "solid phase," as used herein, refers to any material which is insoluble, or can be made insoluble by a subsequent reaction. The solid phase can be chosen for its intrinsic ability to attract and immobilize the capture reagent. Alternatively, the solid phase can retain an additional receptor which has the ability to attract and immobilize the capture reagent. The additional receptor can include a charged substance that is oppositely charged with respect to the capture reagent itself or to a charged substance conjugated to the capture reagent. As yet another alternative, the receptor molecule can be any specific binding member which is immobilized upon (attached to) the solid phase and which has the ability to immobilize the capture reagent through a specific binding reaction. The receptor molecule enables the indirect binding of the capture reagent to a solid phase material before the performance of the assay or during the performance of the assay. The solid phase thus can be a plastic, derivatized plastic, magnetic or non-magnetic metal, glass or silicon surface of a test tube, microtiter well, sheet, bead, microparticle, chip, sheep (or other suitable animal's) red blood cells, Duracytes® and other configurations known to those of ordinary skill in the art.
It is contemplated and within the scope of the present invention that the solid phase also can comprise any suitable porous material with sufficient porosity to allow access by detection antibodies and a suitable surface affinity to bind antigens. Microporous structures generally are preferred, but materials with a gel structure in the hydrated state may be used as well. Such useful solid supports include, but are not limited to, nitrocellulose and nylon. It is contemplated that such porous solid supports described herein preferably are in the form of sheets of thickness from about 0.01 to 0.5 mm, preferably about 0.1 mm. The pore size may vary within wide limits and preferably is from about 0.025 to 15 microns, especially from about 0.15 to 15 microns. The surface of such supports may be activated by chemical processes which cause covalent linkage of the antigen or antibody to the support. The irreversible binding of the antigen or antibody is obtained, however, in general, by adsorption on the porous material by poorly understood hydrophobic forces. Other suitable solid supports are known in the art.
Reagents.
The present invention provides reagents such as polynucleotide sequences derived from a prostate tissue of interest and designated as PS213, polypeptides encoded thereby and antibodies specific for these polypeptides. The present invention also provides reagents such as oligonucleotide fragments derived from the disclosed polynucleotides and nucleic acid sequences complementary to these polynucleotides. The polynucleotides, polypeptides, or antibodies of the present invention may be used to provide information leading to the detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating of, or determining the predisposition to, diseases and conditions of the prostate, such as prostate cancer. The sequences disclosed herein represent unique polynucleotides which can be used in assays or for producing a specific profile of gene transcription activity. Such assays are disclosed in European Patent Number 0373203B1 and International Publication No. WO 95/1 1995.
Selected PS213-derived polynucleotides can be used in the methods described herein for the detection of normal or altered gene expression. Such methods may employ PS213 polynucleotides or oligonucleotides, fragments or derivatives thereof, or nucleic acid sequences complementary thereto.
The polynucleotides disclosed herein, their complementary sequences, or fragments of either, can be used in assays to detect, amplify or quantify genes, nucleic acids, cDNAs or mRNAs relating to prostate tissue disease and conditions associated therewith. They also can be used to identify an entire or partial coding region of a PS213 polypeptide. They further can be provided in individual containers in the form of a kit for assays, or provided as individual compositions. If provided in a kit for assays, other suitable reagents such as buffers, conjugates and the like may be included.
The polynucleotide may be in the form of RNA or DNA. Polynucleotides in the form of DNA, cDNA, genomic DNA, nucleic acid analogs and synthetic DNA are within the scope of the present invention. The DNA may be double- stranded or single-stranded, and if single stranded, may be the coding (sense) strand or non-coding (anti-sense) strand. The coding sequence which encodes the polypeptide may be identical to the coding sequence provided herein or may be a different coding sequence which coding sequence, as a result of the redundancy or degeneracy of the genetic code, encodes the same polypeptide as the DNA provided herein.
This polynucleotide may include only the coding sequence for the polypeptide, or the coding sequence for the polypeptide and an additional coding sequence such as a leader or secretory sequence or a proprotein sequence, or the coding sequence for the polypeptide (and optionally an additional coding sequence) and non-coding sequence, such as a non-coding sequence 5' and/or 3' of the coding sequence for the polypeptide.
In addition, the invention includes variant polynucleotides containing modifications such as polynucleotide deletions, substitutions or additions; and any polypeptide modification resulting from the variant polynucleotide sequence. A polynucleotide of the present invention also may have a coding sequence which is a naturally occurring allelic variant of the coding sequence provided herein. In addition, the coding sequence for the polypeptide may be fused in the same reading frame to a polynucleotide sequence which aids in expression and secretion of a polypeptide from a host cell, for example, a leader sequence which functions as a secretory sequence for controlling transport of a polypeptide from the cell. The polypeptide having a leader sequence is a preprotein and may have the leader sequence cleaved by the host cell to form the polypeptide. The polynucleotides may also encode for a proprotein which is the protein plus additional 5' amino acid residues. A protein having a prosequence is a proprotein and may, in some cases, be an inactive form of the protein. Once the prosequence is cleaved, an active protein remains. Thus, the polynucleotide of the present invention may encode for a protein, or for a protein having a prosequence, or for a protein having both a presequence (leader sequence) and a prosequence.
The polynucleotides of the present invention may also have the coding sequence fused in frame to a marker sequence which allows for purification of the polypeptide of the present invention. The marker sequence may be a hexa-histidine tag supplied by a pQE-9 vector to provide for purification of the polypeptide fused to the marker in the case of a bacterial host, or, for example, the marker sequence may be a hemagglutinin (HA) tag when a mammalian host, e.g. a COS-7 cell line, is used. The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein. See, for example, I. Wilson et al., Cell 37:767 (1984).
It is contemplated that polynucleotides will be considered to hybridize to the sequences provided herein if there is at least 50%, preferably at least 70%, and more preferably at least 90% identity between the polynucleotide and the sequence. The degree of sequence identity between two nucleic acid molecules greatly affects the efficiency and strength of hybridization events between such molecules. A partially identical nucleic acid sequence is one that will at least partially inhibit a completely identical sequence from hybridizing to a target molecule. Inhibition of hybridization of the completely identical sequence can be assessed using hybridization assays that are well known in the art (e.g., Southern blot, Northern blot, solution hybridization, in situ hybridization, or the like, see Sambrook, et al., Molecular Cloning: A Laboratory Manual. Second Edition, (1989) Cold Spring Harbor, N.Y.). Such assays can be conducted using varying degrees of selectivity, for example, using conditions varying from low to high stringency. If conditions of low stringency are employed, the absence of non-specific binding can be assessed using a secondary probe that lacks even a partial degree of sequence identity (for example, a probe having less than about 30% sequence identity with the target molecule), such that, in the absence of non-specific binding events, the secondary probe will not hybridize to the target. When utilizing a hybridization-based detection system, a nucleic acid probe is chosen that is complementary to a target nucleic acid sequence, and then by selection of appropriate conditions the probe and the target sequence "selectively hybridize," or bind, to each other to form a hybrid molecule. In one embodiment of the present invention, a nucleic acid molecule is capable of hybridizing selectively to a target sequence under moderately stringent hybridization conditions. In the context of the present invention, moderately stringent hybridization conditions allow detection of a target nucleic acid sequence of at least 14 nucleotides in length having at least approximately 70% sequence identity with the sequence of the selected nucleic acid probe. In another embodiment, such selective hybridization is performed under stringent hybridization conditions. Stringent hybridization conditions allow detection of target nucleic acid sequences of at least 14 nucleotides in length having a sequence identity of greater than 90% with the sequence of the selected nucleic acid probe. Hybridization conditions useful for probe/target hybridization where the probe and target have a specific degree of sequence identity, can be determined as is known in the art (see, for example, Nucleic Acid Hybridization: A Practical Approach, editors B.D. Hames and S.J. Higgins, (1985) Oxford; Washington, DC; IRL Press). Hybrid molecules can be formed, for example, on a solid support, in solution, and in tissue sections. The formation of hybrids can be monitored by inclusion of a reporter molecule, typically, in the probe. Such reporter molecules, or detectable elements include, but are not limited to, radioactive elements, fluorescent markers, and molecules to which an enzyme-conjugated ligand can bind.
With respect to stringency conditions for hybridization, it is well known in the art that numerous equivalent conditions can be employed to establish a particular stringency by varying, for example, the following factors: the length and nature of probe and target sequences, base composition of the various sequences, concentrations of salts and other hybridization solution components, the presence or absence of blocking agents in the hybridization solutions (e.g., formamide, dextran sulfate, and polyethylene glycol), hybridization reaction temperature and time parameters, as well as, varying wash conditions. The selection of a particular set of hybridization conditions is well within the skill of the routineer in the art (see, for example, Sambrook, et al., Molecular Cloning: A Laboratory Manual, Second Edition, (1989) Cold Spring Harbor, N.Y.). The present invention also provides an antibody produced by using a purified PS213 polypeptide of which at least a portion of the polypeptide is encoded by a PS213 polynucleotide selected from the polynucleotides provided herein. These antibodies may be used in the methods provided herein for the detection of PS213 antigen in test samples. The presence of PS213 antigen in the test samples is indicative of the presence of a prostate disease or condition. The antibody also may be used for therapeutic purposes, for example, in neutralizing the activity of PS213 polypeptide in conditions associated with altered or abnormal expression.
The present invention further relates to a PS213 polypeptide which has the amino acid sequence as provided herein, as well as fragments, analogs and derivatives of such polypeptide. The polypeptide for use with the present invention may be a recombinant polypeptide, a natural purified polypeptide or a synthetic polypeptide. The fragment, derivative or analog of the PS213 polypeptide may be one in which one or more of the amino acid residues is substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code; or it may be one in which one or more of the amino acid residues includes a substituent group; or it may be one in which the polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol); or it may be one in which the additional amino acids are fused to the polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification of the polypeptide or a proprotein sequence. Such fragments, derivatives and analogs are within the scope of the present invention. The polypeptides and polynucleotides of the present invention are provided preferably in an isolated form and preferably purified.
Thus, a polypeptide of the present invention may have an amino acid sequence that is identical to that of the naturally occurring polypeptide or that is different by minor variations due to one or more amino acid substitutions. The variation may be a "conservative change" typically in the range of about 1 to 5 amino acids, wherein the substituted amino acid has similar structural or chemical properties, e.g., replacement of leucine with isoleucine or threonine with serine. In contrast, variations may include nonconservative changes, e.g., replacement of a glycine with a tryptophan. Similar minor variations may also include amino acid deletions or insertions, or both. Guidance in determining which and how many amino acid residues may be substituted, inserted or deleted without changing biological or immunological activity may be found using computer programs well known in the art, for example, DNASTAR software (DNASTAR Inc., Madison WI).
Probes constructed according to the polynucleotide sequences of the present invention can be used in various assay methods to provide various types of analysis. For example, such probes can be used in fluorescent in situ hybridization (FISH) technology to perform chromosomal analysis, and used to identify cancer- specific structural alterations in the chromosomes, such as deletions or translocations that are visible from chromosome spreads or detectable using PCR- generated and/or allele specific oligonucleotides probes, allele specific amplification or by direct sequencing. Probes also can be labeled with radioisotopes, directly- or indirectly- detectable haptens, or fluorescent molecules, and utilized for in situ hybridization studies to evaluate the mRNA expression of the gene comprising the polynucleotide in tissue specimens or cells. This invention also provides teachings as to the production of the polynucleotides and polypeptides provided herein. Probe Assays
The sequences provided herein may be used to produce probes which can be used in assays for the detection of nucleic acids in test samples. The probes may be designed from conserved nucleotide regions of the polynucleotides of interest or from non-conserved nucleotide regions of the polynucleotide of interest. The design of such probes for optimization in assays is within the skill of the routineer. Generally, nucleic acid probes are developed from non-conserved or unique regions when maximum specificity is desired, and nucleic acid probes are developed from conserved regions when assaying for nucleotide regions that are closely related to, for example, different members of a multi-gene family or in related species like mouse and man.
The polymerase chain reaction (PCR) is a technique for amplifying a desired nucleic acid sequence (target) contained in a nucleic acid or mixture thereof. In PCR, a pair of primers are employed in excess to hybridize to the complementary strands of the target nucleic acid. The primers are each extended by a polymerase using the target nucleic acid as a template. The extension products become target sequences themselves, following dissociation from the original target strand. New primers then are hybridized and extended by a polymerase, and the cycle is repeated to geometrically increase the number of target sequence molecules. PCR is disclosed in U.S. Patents 4,683,195 and 4,683,202.
The Ligase Chain Reaction (LCR) is an alternate method for nucleic acid amplification. In LCR, probe pairs are used which include two primary (first and second) and two secondary (third and fourth) probes, all of which are employed in molar excess to target. The first probe hybridizes to a first segment of the target strand, and the second probe hybridizes to a second segment of the target strand, the first and second segments being contiguous so that the primary probes abut one another in 5' phosphate-3' hydroxyl relationship, and so that a ligase can covalently fuse or ligate the two probes into a fused product. In addition, a third (secondary) probe can hybridize to a portion of the first probe and a fourth (secondary) probe can hybridize to a portion of the second probe in a similar abutting fashion. Of course, if the target is initially double stranded, the secondary probes also will hybridize to the target complement in the first instance. Once the ligated strand of primary probes is separated from the target strand, it will hybridize with the third and fourth probes which can be ligated to form a complementary, secondary ligated product. It is important to realize that the ligated products are functionally equivalent to either the target or its complement. By repeated cycles of hybridization and ligation, amplification of the target sequence is achieved. This technique is described more completely in EP-A- 320 308 to K. Backman published June 16, 1989 and EP-A-439 182 to K. Backman et al., published July 31 , 1991.
For amplification of mRNAs, it is within the scope of the present invention to reverse transcribe mRNA into cDNA followed by polymerase chain reaction (RT-PCR); or, to use a single enzyme for both steps as described in U.S. Patent No. 5,322,770; or reverse transcribe mRNA into cDNA followed by asymmetric gap ligase chain reaction (RT-AGLCR) as described by R.L. Marshall et al., PCR Methods and Applications 4:80-84 (1994). Other known amplification methods which can be utilized herein include but are not limited to the so-called "NASBA" or "3SR" technique described by J.C. Guatelli et al., Proc. Natl. Acad. Sci. USA 87: 1874-1878 (1990) and also described by J. Compton, Nature 350 (No. 6313):91-92 (1991); Q-beta amplification as described in published European Patent Application (EPA) No. 4544610; strand displacement amplification (as described in G.T. Walker et al., Clin. Chem. 42:9-13 [1996]) and European Patent Application No. 684315; and target mediated amplification, as described in International Publication No. WO 93/22461.
Detection of PS213 may be accomplished using any suitable detection method, including those detection methods which are currently well known in the art, as well as detection strategies which may evolve later. See, for example, Caskey et al, U.S. Patent No. 5,582,989, Gelfand et al., U.S. Patent No. 5,210,015. Examples of such detection methods include target amplification methods as well as signal amplification technologies. An example of presently known detection methods would include the nucleic acid amplification technologies referred to as PCR, LCR, NASBA, SDA, RCR and TMA. See, for example, Caskey et al., U.S. Patent No. 5,582,989, Gelfand et al., U.S. Patent No. 5,210,015. Detection may also be accomplished using signal amplification such as that disclosed in Snitman et al., U.S. Patent No. 5,273,882. While the amplification of target or signal is preferred at present, it is contemplated and within the scope of the present invention that ultrasensitive detection methods which do not require amplification can be utilized herein. Detection, both amplified and non-amplified, may be performed using a variety of heterogeneous and homogeneous detection formats. Examples of heterogeneous detection formats are disclosed in Snitman et al., U.S. Patent No. 5,273,882, Albarella et al in EP-841 14441.9, Urdea et al., U.S. Patent No. 5,124,246, Ullman et al. U.S. Patent No. 5,185,243 and Kourilsky et al., U.S. Patent No. 4,581,333. Examples of homogeneous detection formats are disclosed in, Caskey et al., U.S. Patent No. 5,582,989, Gelfand et al., U.S. Patent No. 5,210,015. Also contemplated and within the scope of the present invention is the use of multiple probes in the hybridization assay, which use improves sensitivity and amplification of the PS213 signal. See, for example, Caskey et al., U.S. Patent No. 5,582,989, Gelfand et al., U.S. Patent No. 5,210,015.
In one embodiment, the present invention generally comprises the steps of contacting a test sample suspected of containing a target polynucleotide sequence with amplification reaction reagents comprising an amplification primer, and a detection probe that can hybridize with an internal region of the amplicon sequences. Probes and primers employed according to the method provided herein are labeled with capture and detection labels, wherein probes are labeled with one type of label and primers are labeled with another type of label. Additionally, the primers and probes are selected such that the probe sequence has a lower melt temperature than the primer sequences. The amplification reagents, detection reagents and test sample are placed under amplification conditions whereby, in the presence of target sequence, copies of the target sequence (an amplicon) are produced. In the usual case, the amplicon is double stranded because primers are provided to amplify a target sequence and its complementary strand. The double stranded amplicon then is thermally denatured to produce single stranded amplicon members. Upon formation of the single stranded amplicon members, the mixture is cooled to allow the formation of complexes between the probes and single stranded amplicon members.
As the single stranded amplicon sequences and probe sequences are cooled, the probe sequences preferentially bind the single stranded amplicon members. This finding is counterintuitive given that the probe sequences generally are selected to be shorter than the primer sequences and therefore have a lower melt temperature than the primers. Accordingly, the melt temperature of the amplicon produced by the primers should also have a higher melt temperature than the probes. Thus, as the mixture cools, the re-formation of the double stranded amplicon would be expected. As previously stated, however, this is not the case. The probes are found to preferentially bind the single stranded amplicon members. Moreover, this preference of probe/single stranded amplicon binding exists even when the primer sequences are added in excess of the probes.
After the probe/single stranded amplicon member hybrids are formed, they are detected. Standard heterogeneous assay formats are suitable for detecting die hybrids using the detection labels and capture labels present on the primers and probes. The hybrids can be bound to a solid phase reagent by virtue of the capture label and detected by virtue of the detection label. In cases where the detection label is directly detectable, the presence of the hybrids on the solid phase can be detected by causing the label to produce a detectable signal, if necessary, and detecting the signal. In cases where the label is not directly detectable, the captured hybrids can be contacted with a conjugate, which generally comprises a binding member attached to a directly detectable label. The conjugate becomes bound to the complexes and the conjugate's presence on the complexes can be detected with the directly detectable label. Thus, the presence of the hybrids on the solid phase reagent can be determined. Those skilled in the art will recognize that wash steps may be employed to wash away unhybridized amplicon or probe as well as unbound conjugate.
In one embodiment, the heterogeneous assays can be conveniently performed using a solid phase support that carries an array of nucleic acid molecules. Such arrays are useful for high-throughput and/or multiplexed assay formats. Various methods for forming such arrays from pre-formed nucleic acid molecules, or methods for generating the array using in situ synthesis techniques, are generally known in the art. [See, for example, Dattagupta, et al., EP Publication No. 0 234, 726A3; Southern, U.S. Patent No. 5,700,637; Pirrung, et al., U.S. Patent No. 5,143,854; PCT International Publication No. WO 92/10092; and, Fodor, et al., Science 251:767-777 (1991).] Although the target sequence is described as single stranded, it also is contemplated to include the case where the target sequence is actually double stranded but is merely separated from its complement prior to hybridization with the amplification primer sequences. In the case where PCR is employed in this method, the ends of the target sequences are usually known. In cases where LCR or a modification thereof is employed in the preferred method, the entire target sequence is usually known. Typically, the target sequence is a nucleic acid sequence such as, for example, RNA or DNA. The method provided herein can be used in well-known amplification reactions that include thermal cycle reaction mixtures, particularly in PCR and gap LCR (GLCR). Amplification reactions typically employ primers to repeatedly generate copies of a target nucleic acid sequence, which target sequence is usually a small region of a much larger nucleic acid sequence. Primers are themselves nucleic acid sequences that are complementary to regions of a target sequence. Under amplification conditions, these primers hybridize or bind to the complementary regions of the target sequence. Copies of the target sequence typically are generated by the process of primer extension and/or ligation which utilizes enzymes with polymerase or ligase activity, separately or in combination, to add nucleotides to the hybridized primers and/or ligate adjacent probe pairs. The nucleotides that are added to the primers or probes, as monomers or preformed oligomers, are also complementary to the target sequence. Once the primers or probes have been sufficiently extended and/or ligated, they are separated from the target sequence, for example, by heating the reaction mixture to a "melt temperature" which is one in which complementary nucleic acid strands dissociate. Thus, a sequence complementary to the target sequence is formed.
A new amplification cycle then can take place to further amplify the number of target sequences by separating any double stranded sequences, allowing primers or probes to hybridize to their respective targets, extending and/or Hgating the hybridized primers or probes and re-separating. The complementary sequences that are generated by amplification cycles can serve as templates for primer extension or filling the gap of two probes to further amplify the number of target sequences. Typically, a reaction mixture is cycled between 20 and 100 times, more typically, a reaction mixture is cycled between 25 and 50 times. The numbers of cycles can be determined by the routineer. In this manner, multiple copies of the target sequence and its complementary sequence are produced. Thus, primers initiate amplification of the target sequence when it is present under amplification conditions. Generally, two primers which are complementary to a portion of a target strand and its complement are employed in PCR. For LCR, four probes, two of which are complementary to a target sequence and two of which are similarly complementary to the target's complement, are generally employed. In addition to the primer sets and enzymes previously mentioned, a nucleic acid amplification reaction mixture may also comprise other reagents which are well known and include but are not limited to: enzyme cofactors such as manganese; magnesium; salts; nicotinamide adenine dinucleotide (NAD); and deoxynucleotide triphosphates (dNTPs) such as, for example, deoxyadenine triphosphate, deoxyguanine triphosphate, deoxycytosine triphosphate and deoxythymine triphosphate.
While the amplification primers initiate amplification of the target sequence, the detection (or hybridization) probe is not involved in amplification. Detection probes are generally nucleic acid sequences or uncharged nucleic acid analogs such as, for example, peptide nucleic acids which are disclosed in International Publication No. WO 92/20702; morpholino analogs which are described in U.S. Patents Nos 5,185,444, 5,034,506 and 5,142,047; and the like. Depending upon the type of label carried by the probe, the probe is employed to capture or detect the amplicon generated by the amplification reaction. The probe is not involved in amplification of the target sequence and therefore may have to be rendered "non- extendible" in that additional dNTPs cannot be added to the probe. In and of themselves, analogs usually are non-extendible and nucleic acid probes can be rendered non-extendible by modifying the 3' end of the probe such that the hydroxyl group is no longer capable of participating in elongation. For example, the 3' end of the probe can be functionalized with the capture or detection label to thereby consume or otherwise block the hydroxyl group. Alternatively, the 3' hydroxyl group simply can be cleaved, replaced or modified. U.S. Patent Application Serial No. 07/049,061 filed April 19, 1993 describes modifications which can be used to render a probe non-extendible.
The ratio of primers to probes is not important. Thus, either the probes or primers can be added to the reaction mixture in excess whereby the concentration of one would be greater than the concentration of the other. Alternatively, primers and probes can be employed in equivalent concentrations. Preferably, however, the primers are added to the reaction mixture in excess of the probes. Thus, primer to probe ratios of, for example, 5: 1 and 20:1, are preferred.
While the length of the primers and probes can vary, the probe sequences are selected such that they have a lower melt temperature than the primer sequences. Hence, the primer sequences are generally longer than the probe sequences. Typically, the primer sequences are in the range of between 20 and 50 nucleotides long, more typically in the range of between 20 and 30 nucleotides long. The typical probe is in the range of between 10 and 25 nucleotides long.
Various methods for synthesizing primers and probes are well known in the art. Similarly, methods for attaching labels to primers or probes are also well known in the art. For example, it is a matter of routine to synthesize desired nucleic acid primers or probes using conventional nucleotide phosphoramidite chemistry and instruments available from Applied Biosystems, Inc., (Foster City, CA), DuPont (Wilmington, DE), or Milligen (Bedford, MA). Many methods have been described for labeling oligonucleotides such as the primers or probes of the present invention. Enzo Biochemical (New York, NY) and Clontech (Palo Alto, CA) both have described and commercialized probe labeling techniques. For example, a primary amine can be attached to a 3' oligo terminus using 3'-Amine- ON CPG™ (Clontech, Palo Alto, CA). Similarly, a primary amine can be attached to a 5' oligo terminus using Aminomodifier II® (Clontech). The amines can be reacted to various haptens using conventional activation and linking chemistries. In addition, copending applications U.S. Serial Nos. 625,566, filed December 11, 1990 and 630,908, filed December 20, 1990, teach methods for labeling probes at their 5' and 3' termini, respectively. International Publication Nos WO 92/10505, published 25 June 1992, and WO 92/11388, published 9 July 1992, teach methods for labeling probes at their 5' and 3' ends, respectively. According to one known method for labeling an oligonucleotide, a label-phosphoramidite reagent is prepared and used to add the label to the oligonucleotide during its synthesis. See, for example, N.T. Thuong et al., Tet. Letters 29(46):5905-5908 (1988); or J.S.
Cohen et al., published U.S. Patent Application 07/246,688 (NTIS ORDER No. PAT-APPL-7-246,688) (1989). Preferably, probes are labeled at their 3' and 5' ends.
A capture label is attached to the primers or probes and can be a specific binding member which forms a binding pair with the solid phase reagent's specific binding member. It will be understood that the primer or probe itself may serve as the capture label. For example, in the case where a solid phase reagent's binding member is a nucleic acid sequence, it may be selected such that it binds a complementary portion of the primer or probe to thereby immobilize the primer or probe to the solid phase. In cases where the probe itself serves as the binding member, those skilled in the art will recognize that the probe will contain a sequence or "tail" that is not complementary to the single stranded amplicon members. In the case where the primer itself serves as the capture label, at least a portion of the primer will be free to hybridize with a nucleic acid on a solid phase because the probe is selected such that it is not fully complementary to the primer sequence.
Generally, probe/single stranded amplicon member complexes can be detected using techniques commonly employed to perform heterogeneous immunoassays. Preferably, in this embodiment, detection is performed according to the protocols used by the commercially available Abbott LCx® instrumentation (Abbott Laboratories, Abbott Park, IL). The primers and probes disclosed herein are useful in typical PCR assays, wherein the test sample is contacted with a pair of primers, amplification is performed, the hybridization probe is added, and detection is performed.
Another method provided by the present invention comprises contacting a test sample with a plurality of polynucleotides, wherein at least one polynucleotide is a PS213 molecule as described herein, hybridizing the test sample with the plurality of polynucleotides and detecting hybridization complexes. Hybridization complexes are identified and quantitated to compile a profile which is indicative of prostate tissue disease, such as prostate cancer. Expressed RNA sequences may further be detected by reverse transcription and amplification of the DNA product by procedures well-known in the art, including polymerase chain reaction (PCR). Drug Screening and Gene Therapy.
The present invention also encompasses the use of gene therapy methods for the introduction of anti-sense PS213 derived molecules, such as polynucleotides or oligonucleotides of the present invention, into patients with conditions associated with abnormal expression of polynucleotides related to a prostate tissue disease or condition especially prostate cancer. These molecules, including antisense RNA and DNA fragments and ribozymes, are designed to inhibit the translation of PS213 mRNA, and may be used therapeutically in the treatment of conditions associated with altered or abnormal expression of PS213 polynucleotide.
Alternatively, the oligonucleotides described above can be delivered to cells by procedures known in the art such that the anti-sense RNA or DNA may be expressed in vivo to inhibit production of a PS213 polypeptide in the manner described above. Antisense constructs to a PS213 polynucleotide, therefore, reverse the action of PS213 transcripts and may be used for treating prostate tissue disease conditions, such as prostate cancer. These antisense constructs may also be used to treat tumor metastases.
The present invention also provides a method of screening a plurality of compounds for specific binding to PS213 polypeptide(s), or any fragment thereof, to identify at least one compound which specifically binds the PS213 polypeptide. Such a method comprises the steps of providing at least one compound; combining the PS213 polypeptide with each compound under suitable conditions for a time sufficient to allow binding; and detecting the PS213 polypeptide binding to each compound.
The polypeptide or peptide fragment employed in such a test may either be free in solution, affixed to a solid support, borne on a cell surface or located intracellularly. One method of screening utilizes eukaryotic or prokaryotic host cells which are stably transfected with recombinant nucleic acids which can express the polypeptide or peptide fragment. A drug, compound, or any other agent may be screened against such transfected cells in competitive binding assays. For example, the formation of complexes between a polypeptide and the agent being tested can be measured in either viable or fixed cells.
The present invention thus provides methods of screening for drugs, compounds, or any other agent which can be used to treat diseases associated with PS213. These methods comprise contacting the agent with a polypeptide or fragment thereof and assaying for either the presence of a complex between the agent and the polypeptide, or for the presence of a complex between the polypeptide and the cell. In competitive binding assays, the polypeptide typically is labeled. After suitable incubation, free (or uncomplexed) polypeptide or fragment thereof is separated from that present in bound form, and the amount of free or uncomplexed label is used as a measure of the ability of the particular agent to bind to the polypeptide or to interfere with the polypeptide/cell complex.
The present invention also encompasses the use of competitive screening assays in which neutralizing antibodies capable of binding polypeptide specifically compete with a test agent for binding to the polypeptide or fragment thereof. In this manner, the antibodies can be used to detect the presence of any polypeptide in the test sample which shares one or more antigenic determinants with a PS213 polypeptide as provided herein.
Another technique for screening provides high throughput screening for compounds having suitable binding affinity to at least one polypeptide of PS213 disclosed herein. Briefly, large numbers of different small peptide test compounds are synthesized on a solid phase, such as plastic pins or some other surface. The peptide test compounds are reacted with polypeptide and washed. Polypeptide thus bound to the solid phase is detected by methods well-known in the art. Purified polypeptide can also be coated directly onto plates for use in the screening techniques described herein. In addition, non-neutralizing antibodies can be used to capture the polypeptide and immobilize it on the solid support. See, for example, EP 84/03564, published on September 13, 1984.
The goal of rational drug design is to produce structural analogs of biologically active polypeptides of interest or of the small molecules including agonists, antagonists, or inhibitors with which they interact. Such structural analogs can be used to design drugs which are more active or stable forms of the polypeptide or which enhance or interfere with the function of a polypeptide in vivo. J. Hodgson. Bio/Technology 9: 19-21 (19911. For example, in one approach, the three-dimensional structure of a polypeptide, or of a polypeptide-inhibitor complex, is determined by x-ray crystallography, by computer modeling or, most typically, by a combination of the two approaches. Both the shape and charges of the polypeptide must be ascertained to elucidate the structure and to determine active site(s) of the molecule. Less often, useful information regarding the structure of a polypeptide may be gained by modeling based on the structure of homologous proteins. In both cases, relevant structural information is used to design analogous polypeptide-like molecules or to identify efficient inhibitors Useful examples of rational drug design may include molecules which have improved activity or stability as shown by S. Braxton et al., Biochemistry 31:7796-7801 (1992), or which act as inhibitors, agonists, or antagonists of native peptides as shown by S.B.P. Athauda et al., J Biochem. (Tokvol 1 13 (6):742-746 (1993). It also is possible to isolate a target-specific antibody selected by an assay as described hereinabove, and then to determine its crystal structure. In principle this approach yields a pharmacophore upon which subsequent drug design can be based. It further is possible to bypass protein crystallography altogether by generating anti-idiotypic antibodies ("anti-ids") to a functional, pharmacologically active antibody. As a mirror image of a mirror image, the binding site of the anti-id is an analog of the original receptor. The anti-id then can be used to identify and isolate peptides from banks of chemically or biologically produced peptides. The isolated peptides then can act as the pharmacophore (that is, a prototype pharmaceutical drug). A sufficient amount of a recombinant polypeptide of the present invention may be made available to perform analytical studies such as X-ray crystallography. In addition, knowledge of the polypeptide amino acid sequence which is derivable from the nucleic acid sequence provided herein will provide guidance to those employing computer modeling techniques in place of, or in addition to, x-ray crystallography.
Antibodies specific to a PS213 polypeptide (e.g., anti-PS213 antibodies) further may be used to inhibit the biological action of the polypeptide by binding to the polypeptide. In this manner, the antibodies may be used in therapy, for example, to treat prostate tissue diseases including prostate cancer and its metastases.
Further, such antibodies can detect the presence or absence of a PS213 polypeptide in a test sample and, therefore, are useful as diagnostic markers for the diagnosis of a prostate tissue disease or condition especially prostate cancer. Such antibodies may also function as a diagnostic marker for prostate tissue disease conditions, such as prostate cancer.
The present invention also is directed to antagonists and inhibitors of the PS213 polypeptides. The antagonists and inhibitors are those which inhibit or eliminate the function of the polypeptide. Thus, for example, an antagonist may bind to a polypeptide of the present invention and inhibit or eliminate its function. The antagonist, for example, could be an antibody against the polypeptide which eliminates the activity of a PS213 polypeptide by binding a PS213 polypeptide, or in some cases the antagonist may be an oligonucleotide. Examples of small molecule inhibitors include, but are not limited to, small peptides or peptide-like molecules.
The antagonists and inhibitors may be employed as a composition with a pharmaceutically acceptable carrier including, but not limited to. saline, buffered saline, dextrose, water, glycerol. ethanol and combinations thereof. Administration of PS213 polypeptide inhibitors is preferably systemic. The present invention also provides an antibody which inhibits the action of such a polypeptide.
Antisense technology can be used to reduce gene expression through triple- helix formation or antisense DNA or RNA, both of which methods are based on binding of a polynucleotide to DNA or RNA. For example, the 5' coding portion of the polynucleotide sequence, which encodes for the polypeptide of the present invention, is used to design an antisense RNA oligonucleotide of from 10 to 40 base pairs in length. A DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription, thereby preventing transcription and the production of the PS213 polypeptide. For triple helix, see. for example, Lee et al., Nuc. Acids Res. 6:3073 (1979); Cooney et al., Science 241:456 (1988); and Dervan et al., Science 251: 1360 (1991) The antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of a mRNA molecule into the PS213 polypeptide. For antisense, see, for example, Okano, J. Neurochem. 56:560 (1991): and Oligodeoxynucleotides as Antisense Inhibitors of Gene
Expression. CRC Press, Boca Raton, Fla. (1988). Antisense oligonucleotides act with greater efficacy when modified to contain artificial internucleotide linkages which render the molecule resistant to nucleolytic cleavage. Such artificial internucleotide linkages include, but are not limited to, methylphosphonate, phosphorothiolate and phosphoroamydate internucleotide linkages. Recombinant Technology.
The present invention provides host cells and expression vectors comprising PS213 polynucleotides of the present invention and methods for the production of the polypeptide(s) they encode. Such methods comprise culturing the host cells under conditions suitable for the expression of the PS213 polynucleotide and recovering the PS213 polypeptide from the cell culture. The present invention also provides vectors which include PS213 polynucleotides of the present invention, host cells which are genetically engineered with vectors of the present invention and the production of polypeptides of the present invention by recombinant techniques.
Host cells are genetically engineered (transfected, transduced or transformed) with the vectors of this invention which may be cloning vectors or expression vectors. The vector may be in the form of a plasmid, a viral particle, a phage, etc. The engineered host cells can be cultured in conventional nutrient media modified as appropriate for activating promoters, selecting transfected cells, or amplifying PS213 gene(s). The culture conditions, such as temperature, pH and the like, are those previously used with the host cell selected for expression, and will be apparent to the ordinarily skilled artisan.
The polynucleotides of the present invention may be employed for producing a polypeptide by recombinant techniques. Thus, the polynucleotide sequence may be included in any one of a variety of expression vehicles, in particular, vectors or plasmids for expressing a polypeptide. Such vectors include chromosomal, nonchromosomal and synthetic DNA sequences, e.g., derivatives of SV40; bacterial plasmids; phage DNA; yeast plasmids; vectors derived from combinations of plasmids and phage DNA, viral DNA such as vaccinia, adenovirus, fowl pox virus and pseudorabies. However, any other plasmid or vector may be used so long as it is replicable and viable in the host. The appropriate DNA sequence may be inserted into the vector by a variety of procedures. In general, the DNA sequence is inserted into appropriate restriction endonuclease sites by procedures known in the art. Such procedures and others are deemed to be within the scope of those skilled in the art. The DNA sequence in the expression vector is operatively linked to an appropriate expression control sequence(s) (promoter) to direct mRNA synthesis. Representative examples of such promoters include, but are not limited to, the LTR or the SV40 promoter, the I coji lac or tip, the phage lambda P sub L promoter and other promoters known to control expression of genes in prokaryotic or eukaryotic cells or their viruses. The expression vector also contains a ribosome binding site for translation initiation and a transcription terminator. The vector may also include appropriate sequences for amplifying expression. In addition, the expression vectors preferably contain a gene to provide a phenotypic trait for selection of transfected host cells such as dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, or such as tetracycline or ampicillin resistance in FL coli.
The vector containing the appropriate DNA sequence as hereinabove described, as well as an appropriate promoter or control sequence, may be employed to transfect an appropriate host to permit the host to express the protein. As representative examples of appropriate hosts, there may be mentioned: bacterial cells, such as IL coli. Salmonella typhimurium Streptomyces sp.: fungal cells, such as yeast; insect cells, such as Drosophila and Sf9; animal cells, such as CHO, COS or Bowes melanoma; plant cells, etc. The selection of an appropriate host is deemed to be within the scope of those skilled in the art from the teachings provided herein.
More particularly, the present invention also includes recombinant constructs comprising one or more of the sequences as broadly described above. The constructs comprise a vector, such as a plasmid or viral vector, into which a sequence of the invention has been inserted, in a forward or reverse orientation. In a preferred aspect of this embodiment, the construct further comprises regulatory sequences including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art and are commercially available. The following vectors are provided by way of example. Bacterial: pINCY (Incyte Pharmaceuticals Inc., Palo Alto, CA), pSPORTl (Life Technologies, Gaithersburg, MD), pQE70, pQE60, pQE-9 (Qiagen) pBs, phagescript, psiX174, pBluescript SK, pBsKS, pNH8a, pNHlόa, pNH18a, pNH46a (Stratagene); pTrc99A, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia); Eukaryotic: pWLneo, pSV2cat, pOG44, pXTl, pSG (Stratagene) pSVK3, pBPV, pMSG, pSVL (Pharmacia). However, any other plasmid or vector may be used as long as it is replicable and viable in the host.
Plasmid pINCY is generally identical to the plasmid pSPORTl (available from Life Technologies, Gaithersburg, MD) with the exception that it has two modifications in the polylinker (multiple cloning site). These modifications are (1) it lacks a Hindffl restriction site and (2) its EcoRI restriction site lies at a different location. pINCY is created from pSPORTl by cleaving pSPORTl with both Hindiπ and EcoRI and replacing the excised fragment of the polylinker with synthetic DNA fragments (SEQUENCE ID NO 2 and SEQUENCE ID NO 3). This replacement may be made in any manner known to those of ordinary skill in the art. For example, the two nucleotide sequences, SEQUENCE ID NO 2 and SEQUENCE ID NO 3, may be generated synthetically with 5' terminal phosphates, mixed together, and then ligated under standard conditions for performing staggered end ligations into the pSPORTl plasmid cut with Hindiπ and EcoRI. Suitable host cells (such as EL coH DH5μ cells) then are transfected with the ligated DNA and recombinant clones are selected for ampicillin resistance. Plasmid DNA then is prepared from individual clones and subjected to restriction enzyme analysis or DNA sequencing in order to confirm the presence of insert sequences in the proper orientation. Other cloning strategies known to the ordinary artisan also may be employed.
Promoter regions can be selected from any desired gene using CAT (chloramphenicol transferase) vectors or other vectors with selectable markers. Two appropriate vectors are pKK232-8 and pCM7. Particular named bacterial promoters include lad, lacZ, T3, SP6, T7, gpt, lambda P sub R, P sub L and tip. Eukaryotic promoters include cytomegalovirus (CMV) immediate early, herpes simplex virus (HSV) thymidine kinase, early and late SV40, LTRs from retroviruses and mouse metallothionein-I. Selection of the appropriate vector and promoter is well within the level of ordinary skill in the art. In a further embodiment, the present invention provides host cells containing the above-described construct. The host cell can be a higher eukaryotic cell, such as a mammalian cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-Dextran mediated transfection, or electroporation [L. Davis et al, Basic Methods in Molecular Biology. 2nd edition, Appleton and Lang, Paramount Publishing, East Norwalk, CT (1994)].
The constructs in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence. Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
Recombinant proteins can be expressed in mammalian cells, yeast, bacteria, or other cells, under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors- for use with prokaryotic and eukaryotic hosts are described by Sambrook et al., Molecular Cloning: A Laboratory Manual. Second Edition, (Cold Spring Harbor, NY, 1989).
Transcription of a DNA encoding the polypeptide(s) of the present invention by higher eukaryotes is increased by inserting an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription. Examples include the SV40 enhancer on the late side of the replication origin (bp 100 to 270), a cytomegalovirus early promoter enhancer, a polyoma enhancer on the late side of the replication origin and adenovirus enhancers.
Generally, recombinant expression vectors will include origins of replication and selectable markers permitting transfection of the host cell, e.g., the ampicillin resistance gene of EL coH and S cerevisiae TRP 1 gene, and a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence. Such promoters can be derived from operons encoding glycolytic enzymes such as 3-phosphoglycerate kinase (PGK), alpha factor, acid phosphatase, or heat shock proteins, among others. The heterologous structural sequence is assembled in appropriate phase with translation initiation and termination sequences, and preferably, a leader sequence capable of directing secretion of translated protein into the periplasmic space or extracellular medium. Optionally, the heterologous sequence can encode a fusion protein including an N- terminal identification peptide imparting desired characteristics, e.g., stabilization or simplified purification of expressed recombinant product.
Useful expression vectors for bacterial use are constructed by inserting a structural DNA sequence encoding a desired protein together with suitable translation initiation and termination signals in operable reading phase with a functional promoter. The vector will comprise one or more phenotypic selectable markers and an origin of replication to ensure maintenance of the vector and to, if desirable, provide amplification within the host. Suitable prokaryotic hosts for transfection include EL coH, Bacillus subtilis. Salmonella typhimurium and various species within the genera Pseudomonas. Strep tomyces and Staphylococcus. although others may also be employed as a routine matter of choice. Useful expression vectors for bacterial use comprise a selectable marker and bacterial origin of replication derived from plasmids comprising genetic elements of the well-known cloning vector pBR322 (ATCC 37017). Other vectors include but are not limited to PKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEM1 (Promega Biotec, Madison, WI). These pBR322 "backbone" sections are combined with an appropriate promoter and the structural sequence to be expressed.
Following transfection of a suitable host and growth of the host to an appropriate cell density, the selected promoter is derepressed by appropriate means (e.g., temperature shift or chemical induction), and cells are cultured for an additional period. Cells are typically harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification. Microbial cells employed in expression of proteins can be disrupted by any convenient method including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents. Such methods are well-known to the ordinary artisan.
Various mammalian cell culture systems can also be employed to express recombinant protein. Examples of mammalian expression systems include the COS-7 lines of monkey kidney fibroblasts described by Gluzman, Cell 23: 175 (1981), and other cell lines capable of expressing a compatible vector, such as the C127, HEK-293, 3T3, CHO, HeLa and BHK cell lines. Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer and also any necessary ribosome binding sites, polyadenylation sites, splice donor and acceptor sites, transcriptional termination sequences and 5' flanking nontranscribed sequences. DNA sequences derived from the SV40 viral genome, for example, SV40 origin, early promoter, enhancer, splice, and polyadenylation sites may be used to provide the required nontranscribed genetic elements. Representative, useful vectors include pRc/CMV and pcDNA3 (available from Invitrogen, San Diego, CA).
PS213 polypeptides are recovered and purified from recombinant cell cultures by known methods including affinity chromatography, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, hydroxyapatite chromatography or lectin chromatography. It is preferred to have low concentrations (approximately 0.1-5 mM) of calcium ion present during purification [Price, et al., J. Biol. Chem. 244:917 (1969)]. Protein refolding steps can be used, as necessary, in completing configuration of the polypeptide. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps.
Thus, polypeptides of the present invention may be naturally purified products expressed from a high expressing cell line, or a product of chemical synthetic procedures, or produced by recombinant techniques from a prokaryotic or eukaryotic host (for example, by bacterial, yeast, higher plant, insect and mammalian cells in culture). Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated with mammalian or other eukaryotic carbohydrates or may be non- glycosylated. The polypeptides of the invention may also include an initial methionine amino acid residue. The starting plasmids can be constructed from available plasmids in accord with published, known procedures. In addition, equivalent plasmids to those described are known in the art and will be apparent to one of ordinary skill in the art. The following is the general procedure for the isolation and analysis of cDNA clones. In a particular embodiment disclosed herein, mRNA is isolated from prostate tissue and used to generate the cDNA library. Prostate tissue is obtained from patients by surgical resection and is classified as tumor or non-tumor tissue by a pathologist.
The cDNA inserts from random isolates of the prostate tissue libraries are sequenced in part, analyzed in detail as set forth in the Examples.
Methods for DNA sequencing are well known in the art. Conventional enzymatic methods employ DNA polymerase, Klenow fragment, Sequenase (US Biochemical Corp, Cleveland, OH) or Taq polymerase to extend DNA chains from an oligonucleotide primer annealed to the DNA template of interest. Methods have been developed for the use of both single-stranded and double-stranded templates. The chain termination reaction products may be electrophoresed on urea/polyacrylamide gels and detected either by autoradiography (for radionucleotide labeled precursors) or by fluorescence (for fluorescent-labeled precursors). Recent improvements in mechanized reaction preparation, sequencing and analysis using the fluorescent detection method have permitted expansion in the number of sequences that can be determined per day using machines such as the Applied Biosystems 377 DNA Sequencers (Applied Biosystems, Foster City, CA).
The reading frame of the nucleotide sequence can be ascertained by several types of analyses. First, reading frames contained within the coding sequence can be analyzed for the presence of start codon ATG and stop codons TGA, TAA or TAG. Typically, one reading frame will continue throughout the major portion of a cDNA sequence while other reading frames tend to contain numerous stop codons. In such cases, reading frame determination is straightforward. In other more difficult cases, further analysis is required.
Algorithms have been created to analyze the occurrence of individual nucleotide bases at each putative codon triplet. See, for example J.W. Fickett, Nuc. Acids Res. 10:5303 (1982). Coding DNA for particular organisms (bacteria, plants and animals) tends to contain certain nucleotides within certain triplet periodicities, such as a significant preference for pyrimidines in the third codon position. These preferences have been incorporated into widely available software which can be used to determine coding potential (and frame) of a given stretch of DNA. The algorithm-derived information combined with start stop codon information can be used to determine proper frame with a high degree of certainty. This, in turn, readily permits cloning of the sequence in the correct reading frame into appropriate expression vectors. The nucleic acid sequences disclosed herein may be joined to a variety of other polynucleotide sequences and vectors of interest by means of well- established recombinant DNA techniques. See J. Sambrook et al., supra. Vectors of interest include cloning vectors, such as plasmids, cosmids, phage derivatives, phagemids, as well as sequencing, replication and expression vectors, and the like. In general, such vectors contain an origin of replication functional in at least one organism, convenient restriction endonuclease digestion sites and selectable markers appropriate for particular host cells. The vectors can be transferred by a variety of means known to those of skill in the art into suitable host cells which then produce the desired DNA, RNA or polypeptides.
Occasionally, sequencing or random reverse transcription errors will mask the presence of the appropriate open reading frame or regulatory element. In such cases, it is possible to determine the correct reading frame by attempting to express the polypeptide and determining the amino acid sequence by standard peptide mapping and sequencing techniques. See. F.M. Ausubel et al.. Current Protocols in Molecular Biology. John Wiley & Sons, New York, NY (1989). Additionally, the actual reading frame of a given nucleotide sequence may be determined by transfection of host cells with vectors containing all three potential reading frames. Only those cells with the nucleotide sequence in the correct reading frame will produce a peptide of the predicted length.
The nucleotide sequences provided herein have been prepared by current, state-of-the-art, automated methods and, as such, may contain unidentified nucleotides. These will not present a problem to those skilled in the art who wish to practice the invention. Several methods employing standard recombinant techniques, described in J. Sambrook (supra) or periodic updates thereof, may be used to complete the missing sequence information. The same techniques used for obtaining a full length sequence, as described herein, may be used to obtain nucleotide sequences.
Expression of a particular cDNA may be accomplished by subcloning the cDNA into an appropriate expression vector and transfecting this vector into an appropriate expression host. The cloning vector used for the generation of the prostate tissue cDNA library can be used for transcribing mRNA of a particular cDNA and contains a promoter for beta-galactosidase, an amino-terminal met and the subsequent seven amino acid residues of beta-galactosidase. Immediately following these eight residues is an engineered bacteriophage promoter useful for artificial priming and transcription, as well as a number of unique restriction sites, including EcoRI, for cloning. The vector can be transfected into an appropriate host strain of EL coli. Induction of the isolated bacterial strain with isopropylthiogalactoside (IPTG) using standard methods will produce a fusion protein which contains the first seven residues of beta-galactosidase, about 15 residues of linker and the peptide encoded within the cDNA. Since cDNA clone inserts are generated by an essentially random process, there is one chance in three that the included cDNA will lie in the correct frame for proper translation. If the cDNA is not in the proper reading frame, the correct frame can be obtained by deletion or insertion of an appropriate number of bases by well known methods including in vitro mutagenesis, digestion with exonuclease III or mung bean nuclease, or oligonucleotide linker inclusion.
The cDNA can be shuttled into other vectors known to be useful for expression of protein in specific hosts. Oligonucleotide primers containing cloning sites and segments of DNA sufficient to hybridize to stretches at both ends of the target cDNA can be synthesized chemically by standard methods. These primers can then be used to amplify the desired gene segments by PCR. The resulting new gene segments can be digested with appropriate restriction enzymes under standard conditions and isolated by gel electrophoresis. Alternately, similar gene segments can be produced by digestion of the cDNA with appropriate restriction enzymes and filling in the missing gene segments with chemically synthesized oligonucleotides. Segments of the coding sequence from more than one gene can be ligated together and cloned in appropriate vectors to optimize expression of recombinant sequence.
Suitable expression hosts for such chimeric molecules include, but are not limited to, mammalian cells, such as Chinese Hamster Ovary (CHO) and human embryonic kidney (HEK) 293 cells, insect cells, such as Sf9 cells, yeast cells, such as Saccharomyces cerevisiae and bacteria, such as IL coli. For each of these cell systems, a useful expression vector may also include an origin of replication to allow propagation in bacteria and a selectable marker such as the beta-lactamase antibiotic resistance gene to allow selection in bacteria. In addition, the vectors may include a second selectable marker, such as the neomycin phosphotransferase gene, to allow selection in transfected eukaryotic host cells. Vectors for use in eukaryotic expression hosts may require the addition of 3' poly A tail if the sequence of interest lacks poly A.
Additionally, the vector may contain promoters or enhancers which increase gene expression. Such promoters are host specific and include, but are not limited to, MMTV, SV40, or metallothionine promoters for CHO cells; tip, lac, tac or T7 promoters for bacterial hosts; or alpha factor, alcohol oxidase or PGH promoters for yeast. Adenoviral vectors with or without transcription enhancers, such as the Rous sarcoma virus (RSV) enhancer, may be used to drive protein expression in mammalian cell lines. Once homogeneous cultures of recombinant cells are obtained, large quantities of recombinantly produced protein can be recovered from the conditioned medium and analyzed using chromatographic methods well known in the art. An alternative method for the production of large amounts of secreted protein involves the transfection of mammalian embryos and the recovery of the recombinant protein from milk produced by transgenic cows, goats, sheep, etc. Polypeptides and closely related molecules may be expressed recombinantly in such a way as to facilitate protein purification. One approach involves expression of a chimeric protein which includes one or more additional polypeptide domains not naturally present on human polypeptides. Such purification-facilitating domains include, but are not limited to, metal-chelating peptides such as histidine-tryptophan domains that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp, Seattle, WA). The inclusion of a cleavable linker sequence such as Factor XA or enterokinase from Invitrogen (San Diego, CA) between the polypeptide sequence and the purification domain may be useful for recovering the polypeptide.
Immunoassays.
PS213 polypeptides, including fragments, derivatives, and analogs thereof, or cells expressing such polypeptides, can be utilized in a variety of assays, many of which are described herein, for the detection of antibodies to prostate tissue. They also can be used as immunogens to produce antibodies. These antibodies can be, for example, polyclonal or monoclonal antibodies, chimeric, single chain and humanized antibodies, as well as Fab fragments, or the product of an Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments. For example, antibodies generated against a polypeptide comprising a sequence of the present invention can be obtained by direct injection of the polypeptide into an animal or by administering the polypeptide to an animal such as a mouse, rabbit, goat or human. A mouse, rabbit or goat is preferred. The polypeptide is selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9,
SEQUENCE ID NO 10, and fragments thereof. The antibody so obtained then will bind the polypeptide itself. In this manner, even a sequence encoding only a fragment of the polypeptide can be used to generate antibodies that bind the native polypeptide. Such antibodies then can be used to isolate the polypeptide from test samples such as tissue suspected of containing that polypeptide. For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique as described by Kohler and Milstein, Nature 256:495-497 (1975), the trioma technique, the human B-cell hybridoma technique as described by Kozbor et al., Immun. Today 4:72 (1983) and the EBV-hybridoma technique to produce human monoclonal antibodies as described by Cole et al., in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc, New York, NY, pp. 77-96 (1985). Techniques described for the production of single chain antibodies can be adapted to produce single chain antibodies to immunogenic polypeptide products of this invention. See, for example, U.S. Patent No. 4,946,778.
Various assay formats may utilize the antibodies of the present invention, including "sandwich" immunoassays and probe assays. For example, the antibodies of the present invention, or fragments thereof, can be employed in various assay systems to determine the presence, if any, of PS213 antigen in a test sample. For example, in a first assay format, a polyclonal or monoclonal antibody or fragment thereof, or a combination of these antibodies, which has been coated on a solid phase, is contacted with a test sample, to form a first mixture. This first mixture is incubated for a time and under conditions sufficient to form antigen/antibody complexes. Then, an indicator reagent comprising a monoclonal or a polyclonal antibody or a fragment thereof, or a combination of these antibodies, to which a signal generating compound has been attached, is contacted with the antigen/antibody complexes to form a second mixture. This second mixture then is incubated for a time and under conditions sufficient to form antibody /antigen/antibody complexes. The presence of PS213 antigen in the test sample and captured on the solid phase, if any, is determined by detecting the measurable signal generated by the signal generating compound. The amount of PS213 antigen present in the test sample is proportional to the signal generated. In an alternative assay format, a mixture is formed by contacting: (1) a polyclonal antibody, monoclonal antibody, or fragment thereof, which specifically binds to PS213 antigen, or a combination of such antibodies bound to a solid support; (2) the test sample; and (3) an indicator reagent comprising a monoclonal antibody, polyclonal antibody, or fragment thereof, which specifically binds to a different PS213 antigen (or a combination of these antibodies) to which a signal generating compound is attached. This mixture is incubated for a time and under conditions sufficient to form antibody/antigen/antibody complexes. The presence, if any, of PS213 antigen present in the test sample and captured on the solid phase is determined by detecting the measurable signal generated by the signal generating compound. The amount of PS213 antigen present in the test sample is proportional to the signal generated.
In another assay format, one or a combination of at least two monoclonal antibodies of the invention can be employed as a competitive probe for the detection of antibodies to PS213 antigen. For example, PS213 polypeptides such as the recombinant antigens disclosed herein, either alone or in combination, are coated on a solid phase. A test sample suspected of containing antibody to PS213 antigen then is incubated with an indicator reagent comprising a signal generating compound and at least one monoclonal antibody of the invention for a time and under conditions sufficient to form antigen/antibody complexes of either the test sample and indicator reagent bound to the solid phase or the indicator reagent bound to the solid phase. The reduction in binding of the monoclonal antibody to the solid phase can be quantitatively measured. In yet another detection method, each of the monoclonal or polyclonal antibodies of the present invention can be employed in the detection of PS213 antigens in tissue sections, as well as in cells, by immunohistochemical analysis. The tissue sections can be cut from either frozen or chemically fixed samples of tissue. If the antigens are to be detected in cells, the cells can be isolated from blood, urine, breast aspirates, or other bodily fluids. The cells may be obtained by biopsy, either surgical or by needle. The cells can be isolated by centrifugation or magnetic attraction after labeling with magnetic particles or ferrofluids so as to enrich a particular fraction of cells for staining with the antibodies of the present invention. Cytochemical analysis wherein these antibodies are labeled directly (with, for example, fluorescein, colloidal gold, horseradish peroxidase, alkaline phosphatase, etc.) or are labeled by using secondary labeled anti-species antibodies (with various labels as exemplified herein) to track the histopathology of disease also are within the scope of the present invention.
In addition, these monoclonal antibodies can be bound to matrices similar to CNBr-activated Sepharose and used for the affinity purification of specific PS213 polypeptides from cell cultures or biological tissues such as to purify recombinant and native PS213 proteins.
The monoclonal antibodies of the invention also can be used for the generation of chimeric antibodies for therapeutic use, or other similar applications. The monoclonal antibodies or fragments thereof can be provided individually to detect PS213 antigens. Combinations of the monoclonal antibodies (and fragments thereof) provided herein also may be used together as components in a mixture or "cocktail" of at least one PS213 antibody of the invention, along with antibodies which specifically bind to other PS213 regions, each antibody having different binding specificities. Thus, this cocktail can include the monoclonal antibodies of the invention which are directed to PS213 polypeptides disclosed herein and other monoclonal antibodies specific to other antigenic determinants of PS213 antigens or other related proteins.
The polyclonal antibody or fragment thereof which can be used in the assay formats should specifically bind to a PS213 polypeptide or other PS213 polypeptides additionally used in the assay. The polyclonal antibody used preferably is of mammalian origin such as, human, goat, rabbit or sheep polyclonal antibody which binds PS213 polypeptide. Most preferably, the polyclonal antibody is of rabbit origin. The polyclonal antibodies used in the assays can be used either alone or as a cocktail of polyclonal antibodies. Since the cocktails used in the assay formats are comprised of either monoclonal antibodies or polyclonal antibodies having different binding specificity to PS213 polypeptides, they are useful for the detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining the predisposition to, diseases and conditions of the prostate, such as prostate cancer. It is contemplated and within the scope of the present invention that PS213 antigen may be detectable in assays by use of a recombinant antigen as well as by use of a synthetic peptide or purified peptide, which peptide comprises an amino acid sequence of PS213. The amino acid sequence of such a polypeptide is selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID -NO 10, and fragments thereof. It also is within the scope of the present invention that different synthetic, recombinant or purified peptides, identifying different epitopes of PS213, can be used in combination in an assay for the detecting, diagnosing, staging, monitoring, prognosticating, in vivo imaging, preventing or treating, or determining the predisposition to diseases and conditions of the prostate, such as prostate cancer. In this case, all of these peptides can be coated onto one solid phase; or each separate peptide may be coated onto separate solid phases, such as microparticles, and then combined to form a mixture of peptides which can be later used in assays. Furthermore, it is contemplated that multiple peptides which define epitopes from different antigens may be used for the detection, diagnosis, staging, monitoring, prognosis, prevention or treatment of, or determining the predisposition to. diseases and conditions of the prostate, such as prostate cancer. Peptides coated on solid phases or labeled with detectable labels are then allowed to compete with those present in a patient sample (if any) for a limited amount of antibody. A reduction in binding of the synthetic, recombinant, or purified peptides to the antibody (or antibodies) is an indication of the presence of PS213 antigen in the patient sample. The presence of PS213 antigen indicates the presence of prostate tissue disease, especially prostate cancer, in the patient. Variations of assay formats are known to those of ordinary skill in the art and many are discussed herein below.
In another assay format, the presence of anti-PS213 antibody and/or PS213 antigen can be detected in a simultaneous assay, as follows. A test sample is simultaneously contacted with a capture reagent of a first analyte, wherein said capture reagent comprises a first binding member specific for a first analyte attached to a solid phase and a capture reagent for a second analyte, wherein said capture reagent comprises a first binding member for a second analyte attached to a second solid phase, to thereby form a mixture. This mixture is incubated for a time and under conditions sufficient to form capture reagent/first analyte and capture reagent/second analyte complexes. These so-formed complexes then are contacted with an indicator reagent comprising a member of a binding pair specific for the first analyte labeled with a signal generating compound and an indicator reagent comprising a member of a binding pair specific for the second analyte labeled with a signal generating compound to form a second mixture. This second mixture is incubated for a time and under conditions sufficient to form capture reagent first analyte/indicator reagent complexes and capture reagent/second analyte/indicator reagent complexes. The presence of one or more analytes is determined by detecting a signal generated in connection with the complexes formed on either or both solid phases as an indication of the presence of one or more analytes in the test sample. In this assay format, recombinant antigens derived from the expression systems disclosed herein may be utilized, as well as monoclonal antibodies produced from the proteins derived from the expression systems as disclosed herein. For example, in this assay system, PS213 antigen can be the first analyte. Such assay systems are described in greater detail in EP Publication No. 0473065. In yet other assay formats, the polypeptides disclosed herein may be utilized to detect the presence of antibody against PS213 antigen in test samples. For example, a test sample is incubated with a solid phase to which at least one polypeptide such as a recombinant protein or synthetic peptide has been attached. The polypeptide is selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9,
SEQUENCE ID NO 10, and fragments thereof. These are reacted for a time and under conditions sufficient to form antigen/antibody complexes. Following incubation, the antigen antibody complex is detected. Indicator reagents may be used to facilitate detection, depending upon the assay system chosen. In another assay format, a test sample is contacted with a solid phase to which a recombinant protein produced as described herein is attached, and also is contacted with a monoclonal or polyclonal antibody specific for the protein, which preferably has been labeled with an indicator reagent. After incubation for a time and under conditions sufficient for antibody/antigen complexes to form, the solid phase is separated from the free phase, and the label is detected in either the solid or free phase as an indication of the presence of antibody against PS213 antigen. Other assay formats utilizing the recombinant antigens disclosed herein are contemplated. These include contacting a test sample with a solid phase to which at least one antigen from a first source has been attached, incubating the solid phase and test sample for a time and under conditions sufficient to form antigen/antibody complexes, and then contacting the solid phase with a labeled antigen, which antigen is derived from a second source different from the first source. For example, a recombinant protein derived from a first source such as IL coH is used as a capture antigen on a solid phase, a test sample is added to the so-prepared solid phase, and following standard incubation and washing steps as deemed or required, a recombinant protein derived from a different source (i.e., non-E. coli) is utilized as a part of an indicator reagent which subsequently is detected. Likewise, combinations of a recombinant antigen on a solid phase and synthetic peptide in the indicator phase also are possible. Any assay format which utilizes an antigen specific for PS213 produced or derived from a first source as the capture antigen and an antigen specific for PS213 from a different second source is contemplated. Thus, various combinations of recombinant antigens, as well as the use of synthetic peptides. purified proteins and the like, are within the scope of this invention. Assays such as this and others are described in U.S. Patent No. 5,254,458. which enjoys common ownership.
Other embodiments which utilize various other solid phases also are contemplated and are within the scope of this invention. For example, ion capture procedures for immobilizing an immobilizable reaction complex with a negatively charged polymer (described in EP publication 0326100 and EP publication No. 0406473), can be employed according to the present invention to effect a fast solution-phase immunochemical reaction. An immobilizable immune complex is separated from the rest of the reaction mixture by ionic interactions between the negatively charged poly-anion/immune complex and the previously treated, positively charged porous matrix and detected by using various signal generating systems previously described, including those described in chemiluminescent signal measurements as described in EPO Publication No. 0 273,115. Also, the methods of the present invention can be adapted for use in systems which utilize microparticle technology including automated and semi- automated systems wherein the solid phase comprises a microparticle (magnetic or non-magnetic). Such systems include those described in, for example, published EPO applications Nos. EP 0 425 633 and EP 0 424 634, respectively.
The use of scanning probe microscopy (SPM) for immunoassays also is a technology to which the monoclonal antibodies of the present invention are easily adaptable. In scanning probe microscopy, particularly in atomic force microscopy, the capture phase, for example, at least one of the monoclonal antibodies of the invention, is adhered to a solid phase and a scanning probe microscope is utilized to detect antigen/antibody complexes which may be present on the surface of the solid phase. The use of scanning tunneling microscopy eliminates the need for labels which normally must be utilized in many immunoassay systems to detect antigen/antibody complexes. The use of SPM to monitor specific binding reactions can occur in many ways. In one embodiment, one member of a specific binding partner (analyte specific substance which is the monoclonal antibody of the invention) is attached to a surface suitable for scanning. The attachment of the analyte specific substance may be by adsorption to a test piece which comprises a solid phase of a plastic or metal surface, following methods known to those of ordinary skill in the art. Or, covalent attachment of a specific binding partner
(analyte specific substance) to a test piece which test piece comprises a solid phase of derivatized plastic, metal, silicon, or glass may be utilized. Covalent attachment methods are known to those skilled in the art and include a variety of means to irreversibly link specific binding partners to the test piece. If the test piece is silicon or glass, the surface must be activated prior to attaching the specific binding partner. Also, polyelectrolyte interactions may be used to immobilize a specific binding partner on a surface of a test piece by using techniques and chemistries. The preferred method of attachment is by covalent means. Following attachment of a specific binding member, the surface may be further treated with materials such as serum, proteins, or other blocking agents to minimize non-specific binding. The surface also may be scanned either at the site of manufacture or point of use to verify its suitability for assay purposes. The scanning process is not anticipated to alter the specific binding properties of the test piece.
While the present invention discloses the preference for the use of solid phases, it is contemplated that the reagents such as antibodies, proteins and peptides of the present invention can be utilized in non-solid phase assay systems. These assay systems are known to those skilled in the art, and are considered to be within the scope of the present invention. It is contemplated that the reagent employed for the assay can be provided in the form of a test kit with one or more containers such as vials or bottles, with each container containing a separate reagent such as a probe, primer, monoclonal antibody or a cocktail of monoclonal antibodies, or a polypeptide (e.g. recombinantly, synthetically produced or purified) employed in the assay. The polypeptide is selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof. Other components such as buffers, controls and the like, known to those of ordinary skill in art, may be included in such test kits. It also is contemplated to provide test kits which have means for collecting test samples comprising accessible body fluids, e.g., blood, urine, saliva and stool. Such tools useful for collection ("collection materials") include lancets and absorbent paper or cloth for collecting and stabilizing blood; swabs for collecting and stabilizing saliva; cups for collecting and stabilizing urine or stool samples. Collection materials, papers, cloths, swabs, cups and the like, may optionally be treated to avoid denaturation or irreversible adsorption of the sample. The collection materials also may be treated with or contain preservatives, stabilizers or antimicrobial agents to help maintain the integrity of the specimens. Test kits designed for the collection, stabilization and preservation of test specimens obtained by surgery or needle biopsy are also useful. It is contemplated that all kits may be configured in two components which can be provided separately; one component for collection and transport of the specimen and the other component for the analysis of the specimen. The collection component, for example, can be provided to the open market user while the components for analysis can be provided to others such as laboratory personnel for determination of the presence, absence or amount of analyte. Further, kits for the collection, stabilization and preservation of test specimens may be configured for use by untrained personnel and may be available in the open market for use at home with subsequent transportation to a laboratory for analysis of the test sample. In Vivo Antibody Use.
Antibodies of the present invention can be used in vivo: that is, they can be injected into patients suspected of having diseases of the prostate for diagnostic or therapeutic uses. The use of antibodies for in vivo diagnosis is well known in the art. Sumerdon et al, Nucl. Med. Biol 17:247-254 (1990) have described an optimized antibody-chelator for the radioimmunoscintographic imaging of carcinoembryonic antigen (CEA) expressing tumors using Indium- 111 as the label. Griffin et al. . J Clin One 9:631-640 (1991) have described the use of this agent in detecting tumors in patients suspected of having recurrent colorectal cancer. The use of similar agents with paramagnetic ions as labels for magnetic resonance imaging is known in the art (R. B. Lauffer, Magnetic Resonance in Medicine 22:339-342 (1991). It is anticipated that antibodies directed against PS213 antigen can be injected into patients suspected of having a disease of the prostate such as prostate cancer for the purpose of diagnosing or staging the disease status of the patient. The label used will depend on the imaging modality chosen. Radioactive labels such as Indium- 111, Technetium-99m, or Iodine-131 can be used for planar scans or single photon emission computed tomography (SPECT). Positron emitting labels such as Fluorine- 19 can also be used for positron emission tomography (PET). For MRI, paramagnetic ions such as Gadolinium (III) or Manganese (II) can be used. Localization of the label within the prostate or external to the prostate may allow determination of spread of the disease. The amount of label within the prostate may allow determination of the presence or absence of cancer of the prostate. For patients known to have a disease of the prostate, injection of an antibody directed against PS213 antigen may have therapeutic benefit. The antibody may exert its effect without the use of attached agents by binding to PS213 antigen expressed on or in the tissue or organ. Alternatively, the antibody may be conjugated to cytotoxic agents such as drugs, toxins, or radionuclides to enhance its therapeutic effect. Garnett and Baldwin, Cancer Research 46:2407- 2412 (1986) have described the preparation of a drug-monoclonal antibody conjugate. Pastan et al., Cell 47:641-648 (1986) have reviewed the use of toxins conjugated to monoclonal antibodies for the therapy of various cancers. Goodwin and Meares. Cancer Supplement 80:2675-2680 (1997) have described the use of Yittrium-90 labeled monoclonal antibodies in various strategies to maximize the dose to tumor while limiting normal tissue toxicity. Other known cytotoxic radionuclides include Copper-67, Iodine-131 , and Rhenium- 186 all of which can be used to label monoclonal antibodies directed against PS213 antigen for the treatment of cancer of the prostate. The present invention will now be described by way of examples, which are meant to illustrate, but not to limit, the scope of the present invention.
EXAMPLES
Example 1 : Identification of Prostrate Tissue Library PS213 Gene-Specific Clones Library Comparison of Expressed Sequence Tags (EST's) or Transcript
Images. Partial sequences of cDNA clone inserts, so-called "expressed sequence tags" (EST's), were derived from cDNA libraries made from prostrate tumor tissues, prostrate non-tumor tissues and numerous other tissues, both tumor and non-tumor and entered into a database (LEFESEQ™ database, available from Incyte Pharmaceuticals, Palo Alto, CA) as gene transcript images. See International Publication No. WO 95/20681. (A transcript image is a listing of the number of EST's for each of the represented genes in a given tissue library. EST's sharing regions of mutual sequence overlap are classified into clusters. A cluster is assigned a clone number from a representative 5' EST. Often, a cluster of interest can be extended by comparing its consensus sequence with sequences of other EST's which did not meet the criteria for automated clustering. The alignment of all available clusters and single EST's represent a contig from which a consensus sequence is derived.) The transcript images then were evaluated to identify EST sequences that were representative primarily of the prostrate tissue libraries. These target clones then were ranked according to their abundance (occurrence) in the target libraries and their absence from background libraries. Higher abundance clones with low background occurrence were given higher study priority. EST's corresponding to the sequence of PS213 (or fragments thereof) were found in 30.9% (13 of 42) of prostrate tissue libraries. EST's corresponding to the sequence of PS213 (or fragments thereof) were found in only 1.8% (13 of 717) of the other, non-prostrate, libraries of the data base. Therefore, the consensus sequence or fragment thereof was found more than 17 times more often in prostrate than non-prostrate tissues.
Overlapping clones were identified for further study. These represented the minimum number of clones that were needed to form the contig and from which a consensus sequence was derived. The nucleotide sequences of the clones were entered in the Sequencher™ Program (available from Gene Codes Corporation, Ann Arbor, MI. in order to generate a nucleotide alignment (contig map) and then generate their consensus sequence. Following this, a three-frame translation was performed on the consensus sequence. The first forward frame was found to have an open reading frame which was compared with published sequences using software and techniques known to those skilled in the art. The nucleotide sequence was found to be the same as that deposited with GenBank under Accession No.
U78294 and is represented by SEQUENCE ID NO 1 herein. The protein sequence corresponding to this nucleotide sequence is reported in Brash et al., Proc. Natl. Acad. Sci. USA 94:6148-6152 (1997) and is represented by SEQUENCE ID NO
6 herein. Example 2: Sequencing of PS213 EST-Specific Clones DNA sequences for clones which comprise the most upstream and downstream EST's of the PS213 gene contig are determined using dideoxy termination sequencing with dye terminators following known methods [F. Sanger et al., Proc. Natl. Acad. Sci. USA 74:5463 (1977)].
Because vectors such as pSPORTl (Life Technologies, Gaithersburg, MD) and pINCY (available from Incyte Pharmaceuticals, Inc., Palo Alto, CA) contain universal priming sites just adjacent to the 3' and 5' ligation junctions of the inserts, the inserts are sequenced in both directions using universal primers, SEQUENCE ED NO 4 and SEQUENCE ID NO 5 ( New England Biolabs,
Beverly, MA and Applied Biosystems Inc, Foster City, CA, respectively). The sequencing reactions are run on a polyacrylamide denaturing gel, and the sequences are determined by an Applied Biosystems 377 Sequencer (available from Applied Biosystems. Foster City, CA) or other sequencing apparatus.
Example 3: Nucleic Acid A. RNA Extraction from Tissue. Total RNA is isolated from prostrate tissues and from non-prostrate tissues. Various methods are utilized, including but not limited to the lithium chloride/urea technique, known in the art and described by Kato et al., (J. Virol. 61:2182-2191, 1987), and TRIzol™ (Gibco-BRL, Grand Island, NY).
Briefly, tissue is placed in a sterile conical tube on ice and 10-15 volumes of 3 M LiCl, 6 M urea, 5 mM EDTA, 0.1 M β-mercaptoethanol, 50 mM Tris-HCl
(pH 7.5) are added. The tissue is homogenized with a Polytron® homogenizer (Brinkman Instruments, Inc., Westbury, NY) for 30-50 sec on ice. The solution is transferred to a 15 ml plastic centrifuge tube and placed overnight at -20°C. The tube is centrifuged for 90 min at 9,000 x g at 0-4°C and the supernatant is immediately decanted. Ten ml of 3 M LiCl are added and the tube is vortexed for 5 sec. The tube is centrifuged for 45 min at 1 1,000 x g at 0-4°C. The decanting, resuspension in LiCl, and centrifugation is repeated and the final pellet is air dried and suspended in 2 ml of 1 mM EDTA, 0.5% SDS, 10 mM Tris (pH 7.5). Twenty microliters (20 μl) of Proteinase K (20 mg/ml) are added, and the solution is incubated for 30 min at 37°C with occasional mixing. One-tenth volume (0.22- 0.25 ml) of 3 M NaCl is added and the solution is vortexed before transfer into another tube containing 2 ml of phenol/chloroform/isoamyl alcohol (PCI). The tube is vortexed for 1-3 sec and centrifuged for 20 min at 3,000 x g at 10°C. The PCI extraction is repeated and followed by two similar extractions with chloroform isoamyl alcohol (Cl). The final aqueous solution is transferred to a prechilled 15 ml Corex glass tube containing 6 ml of absolute ethanol, the tube is covered with parafilm, and placed at -20°C overnight. The tube is centrifuged for 30 min at 10,000 x g at 0-4°C and the ethanol supernatant is decanted immediately. The RNA pellet is washed four times with 10 ml of 75% ice-cold ethanol and the final pellet is air dried for 15 min at room temperature. The RNA is suspended in 0.5 ml of 10 mM TE (pH 7.6, 1 mM EDTA) and its concentration is determined spectrophotometrically. RNA samples are aliquoted and stored at -70°C as ethanol precipitates. The quality of the RNA is determined by agarose gel electrophoresis (see
Example 5. Northern Blot Analysis) and staining with 0.5 μg/ml ethidium bromide for one hour. RNA samples that do not contain intact rRNAs are excluded from the study.
Alternatively, for RT-PCR analysis, 1 ml of Ultraspec RNA reagent is added to 120 mg of pulverized tissue in a 2.0 ml polypropylene microfuge tube, homogenized with a Polytron® homogenizer (Brinkman Instruments, Inc., Westbury, NY) for 50 sec and placed on ice for 5 min. Then, 0.2 ml of chloroform is added to each sample, followed by vortexing for 15 sec. The sample is placed on ice for another 5 min, followed by centrifugation at 12,000 x g for 15 min at 4°C. The upper layer is collected and transferred to another RNase-free 2.0 ml microfuge tube. An equal volume of isopropanol is added to each sample, and the solution is placed on ice for 10 min. The sample is centrifuged at 12,000 x g for 10 min at 4°C, and the supernatant is discarded. The remaining pellet is washed twice with cold 75% ethanol, resuspended by vortexing, and the resuspended material is then pelleted by centrifugation at 7500 x g for 5 min at 4°C. Finally, the RNA pellet is dried in a Speedvac (Savant, Farmingdale, NY) for 5 min and reconstituted in RNase-free water.
B. RNA Extraction from Blood Mononuclear Cells. Mononuclear cells are isolated from blood samples from patients by centrifugation using Ficoll-Hypaque as follows. A 10 ml volume of whole blood is mixed with an equal volume of
RPMI Medium (Gibco-BRL, Grand Island, NY). This mixture is then underlayed with 10 ml of Ficoll-Hypaque (Pharmacia, Piscataway, NJ) and centrifuged for 30 minutes at 200 x g. The buffy coat containing the mononuclear cells is removed, diluted to 50 ml with Dulbecco's PBS (Gibco-BRL, Grand Island, NY) and the mixture centrifuged for 10 minutes at 200 x g. After two washes, the resulting pellet is resuspended in Dulbecco's PBS to a final volume of 1 ml. RNA is prepared from the isolated mononuclear cells as described by N. Kato et al. J. Virology 61 : 2182-2191 (1987). Briefly, the pelleted mononuclear cells are brought to a final volume of 1 ml and then are resuspended in 250 μL of PBS and mixed with 2.5 ml of 3M LiCl, 6M urea, 5mM EDTA, 0.1M 2- mercaptoethanol, 50mM Tris-HCl (pH 7.5). The resulting mixture is homogenized and incubated at -20°C overnight. The homogenate is centrifuged at 8,000 RPM in a Beckman J2-21M rotor for 90 minutes at 0-4°C. The pellet is resuspended in 10 ml of 3M LiCl by vortexing and then centrifuged at 10,000 RPM in a Beckman J2-21M rotor centrifuge for 45 minutes at 0-4°C. The resuspending and pelleting steps then are repeated. The pellet is resuspended in 2 ml of 1 mM EDTA, 0.5% SDS, 10 mM Tris (pH 7.5) and 400 μg Proteinase K with vortexing and then it is incubated at 37°C for 30 minutes with shaking. One tenth volume of 3M NaCl then is added and the mixture is vortexed. Proteins are removed by two cycles of extraction with phenol/ chloroform/ isoamyl alcohol (PCI) followed by one extraction with chloroform/ isoamyl alcohol (Cl). RNA is precipitated by the addition of 6 ml of absolute ethanol followed by overnight incubation at -20°C. After the precipitated RNA is collected by centrifugation, the pellet is washed 4 times in 75% ethanol. The pelleted RNA is then dissolved in solution containing ImM EDTA, lOmM Tris-HCl (pH 7.5). Non-prostrate tissues are used as negative controls. The mRNA can be further purified from total RNA by using commercially available kits such as oligo dT cellulose spin columns (RediCol™ from Pharmacia, Uppsala, Sweden) for the isolation of poly-adenylated RNA. Total RNA or mRNA can be dissolved in lysis buffer (5M guanidine thiocyanate, 0.1M EDTA, pH 7.0) for analysis in the ribonuclease protection assay.
C. RNA Extraction from polysomes. Tissue is minced in saline at 4°C and mixed with 2.5 volumes of 0.8 M sucrose in a TK150M (150 mM KCI, 5 mM MgCl2, 50 mM Tris-HCl, pH 7.4) solution containing 6 mM 2-mercaptoethanol. The tissue is homogenized in a Teflon-glass Potter homogenizer with five strokes at 100-200 rpm followed by six strokes in a Dounce homogenizer, as described by B. Mechler, Methods in Enzymology 152:241-248 (1987). The homogenate then is centrifuged at 12,000 x g for 15 min at 4°C to sediment the nuclei. The polysomes are isolated by mixing 2 ml of the supernatant with 6 ml of 2.5 M sucrose in TK150M and layering this mixture over 4 ml of 2.5 M sucrose in TK,50M in a 38 ml polyallomer tube. Two additional sucrose TKI50M solutions are successively layered onto the extract fraction; a first layer of 13 ml 2.05 M sucrose followed by a second layer of 6 ml of 1.3 M sucrose. The polysomes are isolated by centrifuging the gradient at 90,000 x g for 5 hr at 4°C. The fraction then is taken from the 1.3 M sucrose/2.05 M sucrose interface with a siliconized pasteur pipette and diluted in an equal volume of TE (10 mM Tris-HCl, pH 7.4, 1 mM EDTA). An equal volume of 90°C SDS buffer (1% SDS, 200 mM NaCl, 20 mM Tris-HCl, pH 7.4) is added and the solution is incubated in a boiling water bath for 2 min. Proteins next are digested with a Proteinase K digestion (50 mg/ml) for 15 min at 37°C. The mRNA is purified with 3 equal volumes of phenol-chloroform extractions followed by precipitation with 0.1 volume of 2 M sodium acetate (pH 5.2) and 2 volumes of 100% ethanol at -20°C overnight. The precipitated RNA is recovered by centrifugation at 12,000 x g for 10 min at 4°C. The RNA is dried and resuspended in TE (pH 7.4) or distilled water. The resuspended RNA then can be used in a slot blot or dot blot hybridization assay to check for the presence of PS213 mRNA (see Example 6).
The quality of nucleic acid and proteins is dependent on the method of preparation used. Each sample may require a different preparation technique to maximize isolation efficiency of the target molecule. These preparation techniques are within the skill of the ordinary artisan.
Example 4: Ribonuclease Protection Assay A. Synthesis of Labeled Complementary RNA (cRNA) Hybridization Probe and Unlabeled Sense Strand. Labeled antisense and unlabeled sense riboprobes are transcribed from the PS213 gene cDNA sequence which contains a 5' RNA polymerase promoter such as SP6 or T7. The sequence may be from a vector containing the appropriate PS213 cDNA insert, or from a PCR-generated product of the insert using PCR primers which incorporate a 5' RNA polymerase promoter sequence. A plasmid containing the PS213 gene cDNA sequence, or fragment thereof, flanked by opposed SP6 and T7 or other RNA polymerase promoters, is purified using a Qiagen Plasmid Purification Kit (Qiagen, Chatsworth, CA). Then 10 μg of the plasmid DNA are linearized by cutting with an appropriate restriction enzyme such as Dde I for 1 hr at 37°C. The linearized plasmid DNA is purified using the QIAprep Kit (Qiagen, Chatsworth, CA) and used for the synthesis of antisense transcript from the appropriate promoter using the Riboprobe® in vitro Transcription System (Promega Corporation, Madison, WI), as described by the supplier's instructions, incorporating either (alpha32P) CTP (Amersham Life Sciences, Inc. Arlington Heights, IL) or biotinylated CTP as a label. To generate the sense strand, 10 μg of the purified plasmid DNA are cut with restriction enzymes, such as Xba I and Not I, and transcribed as above from the appropriate promoter. Both sense and antisense strands are isolated by spin column chromatography. Unlabeled sense strand is quantitated by UV absorption at 260 nm.
B. Hybridization of Labeled Probe to Target. Frozen tissue is pulverized to powder under liquid nitrogen and 100-500 mg are dissolved in 1 ml of lysis buffer, available as a component of the Direct Protect™ Lysate RNase Protection Kit (Ambion, Inc., Austin, TX). Further dissolution can be achieved using a tissue homogenizer. In addition, a dilution series of a known amount of sense strand in mouse liver lysate is made for use as a positive control. Finally, 45 μl of solubilized tissue or diluted sense strand is mixed directly with either ; 1) 1 xlO5 cpm of radioactively labeled probe, or 2) 250 pg of non-isotopically labeled probe in 5 μl of lysis buffer. Hybridization is allowed to proceed overnight at 37°C. See, T. Kaabache et al., Anal. Biochem. 232:225-230 (1995).
C. RNase Digestion. RNA that is not hybridized to probe is removed from the reaction as per the Direct Protect™ protocol using a solution of RNase A and RNase Tl for 30 min at 37°C, followed by removal of RNase by Proteinase K digestion in the presence of sodium sarcosyl. Hybridized fragments protected from digestion are then precipitated by the addition of an equal volume of isopropanol and placed at -70°C for 3 hr. The precipitates are collected by centrifugation at 12,000 x g for 20 min.
P. Fragment Analysis. The precipitates are dissolved in denaturing gel loading dye (80% formamide, 10 mM EDTA (pH 8.0), 1 mg/ml xylene cyanol, 1 mg/ml bromophenol blue), heat denatured, and electrophoresed in 6% polyacrylamide TBE, 8 M urea denaturing gels. The gels are imaged and analyzed using the STORM™ storage phosphor autoradiography system (Molecular Dynamics, Sunnyvale. CA). Quantitation of protected fragment bands, expressed in femtograms (fg), is achieved by comparing the peak areas obtained from the test samples to those from the known dilutions of the positive control sense strand (see Section B, supra). The results are expressed in molecules of PS213 RNA/cell and as a image rating score. In cases where non-isotopic labels are used, hybrids are transferred from the gels to membranes (nylon or nitrocellulose) by blotting and then analyzed using detection systems that employ streptavidin alkaline phosphatase conjugates and chemiluminesence or chemifluoresence reagents.
Detection of a product comprising a sequence of the PS213 polynucleotide (SEQUENCE ID NO 1), or fragments or complements thereof, is indicative of the presence of PS213 mRNA(s), suggesting a diagnosis of a prostrate tissue disease or condition, such as prostrate cancer. Example 5: Northern Blotting The Northern blot technique is used to identify a specific size RNA fragment from a complex population of RNA using gel electrophoresis and nucleic acid hybridization. Northern blotting is well-known technique in the art. Briefly, 5-10 μg of total RNA (see Example 3) are incubated in 15 μl of a solution containing 40 mM morphilinopropanesulfonic acid (MOPS) (pH 7.0), 10 mM sodium acetate, 1 mM EDTA, 2.2 M formaldehyde, 50% v/v formamide for 15 min at 65°C. The denatured RNA is mixed with 2 μl of loading buffer (50% glycerol, 1 mM EDTA, 0.4% bromophenol blue, 0.4% xylene cyanol) and loaded into a denaturing 1.0% agarose gel containing 40 mM MOPS (pH 7.0), 10 mM sodium acetate, 1 mM EDTA and 2.2 M formaldehyde. The gel is electrophoresed at 60 V for 1.5 hr and rinsed in RNAse free water. RNA is transferred from the gel onto nylon membranes (Brightstar-Plus, Ambion, Inc., Austin, TX) for 1.5 hours using the downward alkaline capillary transfer method (Chomczynski, Anal. Biochem. 201 : 134-139. 1992). The filter is rinsed with IX SSC, and RNA is crosslinked to the filter using a Stratalinker™ (Stratagene, Inc., La Jolla, CA) on the autocrosslinking mode and dried for 15 min. The membrane is then placed into a hybridization tube containing 20 ml of preheated prehybridization solution (5X SSC, 50% formamide, 5X Denhardt's solution, 100 μg/ml denatured salmon sperm DNA) and incubated in a 42°C hybridization oven for at least 3 hr. While the blot is prehybridizing, a 32P-labeled random-primed probe is generated using a PS213 insert fragment using Random Primer DNA Labeling System (Life Technologies, Inc., Gaithersburg, MD) according to the manufacturer's instructions. Half of the probe is boiled for 10 min, quick chilled on ice and added to the hybridization tube. Hybridization is carried out at 42°C for at least 12 hr. The hybridization solution is discarded and the filter is washed in 30 ml of 3X SSC, 0.1 % SDS at 42°C for 15 min, followed by 30 ml of 3X SSC, 0.1 % SDS at 42°C for 15 min. The filter is wrapped in Saran Wrap, exposed to Kodak XAR- Omat film for 8-96 hr, and the film is developed for analysis. High level of expression of mRNA corresponding to a sequence of the PS213 polynucleotide (SEQUENCE ID NO 1), or fragments or complements thereof, is an indication of the presence of PS213 mRNA, suggesting a diagnosis of a prostrate tissue disease or condition, such as prostrate cancer.
Example 6: Dot Blot/Slot Blot
Dot and slot blot assays are quick methods to evaluate the presence of a specific nucleic acid sequence in a complex mix of nucleic acid. To perform such assays, up to 50 μg of RNA are mixed in 50 μl of 50% formamide, 7% formaldehyde, IX SSC, incubated 15 min at 68°C, and then cooled on ice. Then, 100 μl of 20X SSC are added to the RNA mixture and loaded under vacuum onto a manifold apparatus that has a prepared nitrocellulose or nylon membrane. The membrane is soaked in water, 20X SSC for 1 hour, placed on two sheets of 20X SSC prewet Whatman #3 filter paper, and loaded into a slot blot or dot blot vacuum manifold apparatus. The slot blot is analyzed with probes prepared and labeled as described in Example 4, supra. Detection of mRNA corresponding to a sequence of the PS213 polynucleotide (SEQUENCE ID NO 1), or fragments or complements thereof, is an indication of the presence of PS213, suggesting a diagnosis of a prostrate tissue disease or condition, such as prostrate cancer.
Other methods and buffers which can be utilized in the methods described in Examples 5 and 6, but not specifically detailed herein, are known in the art and are described in J. Sambrook et al., supra.
Example 7: In Situ Hybridization
This method is useful to directly detect specific target nucleic acid sequences in cells using detectable nucleic acid hybridization probes.
Tissues are prepared with cross-linking fixative agents such as paraformaldehyde or glutaraldehyde for maximum cellular RNA retention. See, L. Angerer et al., Methods in Cell Biol. 35:37-71 (1991). Briefly, the tissue is placed in greater than 5 volumes of 1 % glutaraldehyde in 50 mM sodium phosphate, pH 7.5 at 4°C for 30 min. The solution is changed with fresh glutaraldehyde solution (1% glutaraldehyde in 50mM sodium phosphate, pH 7.5) for a further 30 min fixing. The fixing solution should have an osmolality of approximately 0.375% NaCl. The tissue is washed once in isotonic NaCl to remove the phosphate.
The fixed tissues then are embedded in paraffin as follows. The tissue is dehydrated though a series of increasing ethanol concentrations for 15 min each: 50% (twice), 70% (twice), 85%, 90% and then 100% (twice). Next, the tissue is soaked in two changes of xylene for 20 min each at room temperature. The tissue is then soaked in two changes of a 1:1 mixture of xylene and paraffin for 20 min each at 60°C; and then in three final changes of paraffin for 15 min each.
Next, the tissue is cut in 5 μm sections using a standard microtome and placed on a slide previously treated with a tissue adhesive such as 3- aminopropyltriethoxysilane. Paraffin is removed from the tissue by two 10 min xylene soaks and rehydrated in a series of decreasing ethanol concentrations: 99% twice, 95%, 85%, 70%, 50%, 30%, and then distilled water twice. The sections are pre-treated with 0.2 M HC1 for 10 min and permeabilized with 2 μg/ml Proteinase K at 37°C for 15 min.
Labeled riboprobes transcribed from the PS213 gene plasmid (see Example 4) are hybridized to the prepared tissue sections and incubated overnight at 56°C in 3X standard saline extract and 50% formamide. Excess probe is removed by washing in 2X standard saline citrate and 50% formamide followed by digestion with 100 μg/ml RNase A at 37°C for 30 min. Fluorescence probe is visualized by illumination with ultraviolet (UV) light under a microscope. Fluorescence in the cytoplasm is indicative of PS213 mRNA. Alternatively, the sections can be visualized by autoradiography.
Example 8: Reverse Transcription PCR
A. One Step RT-PCR Assay. Target-specific primers are designed to detect the above-described target sequences by reverse transcription PCR using methods known in the art. One step RT-PCR is a sequential procedure that performs both RT and PCR in a single reaction mixture. The procedure is performed in a 200 μl reaction mixture containing 50 mM (N,N,-bis[2- Hydroxyethyl]glycine), pH 8.15, 81.7 mM KOAc, 33.33 mM KOH, 0.01 mg/ml bovine serum albumin, 0.1 mM ethylene diaminetetraacetic acid, 0.02 mg/ml NaN3; 8% w/v glycerol, 150 μM each of dNTP, 0.25 μM each primer, 5U rTth polymerase, 3.25 mM Mn(OAc)2 and 5 μl of target RNA (see Example 3). Since RNA and the rTth polymerase enzyme are unstable in the presence of Mn(OAc)2, the Mn(OAc)2 should be added just before target addition. Optimal conditions for cDNA synthesis and thermal cycling readily can be determined by those skilled in the art. The reaction is incubated in a Perkin-Elmer Thermal Cycler 480.
Conditions which may be found useful include cDNA synthesis at 60°-70°C for 15- 45 min and 30-45 amplification cycles at 94°C, 1 min; 55°-70°C, 1 min; 72°C, 2 min. One step RT-PCR also may be performed by using a dual enzyme procedure with Taq polymerase and a reverse transcriptase enzyme, such as MMLV (Moloney murine leukemia virus) or AMV (avian myeloblastosis virus) RT (reverse transcriptase) enzymes.
B. Traditional RT-PCR. Alternatively, a traditional two-step RT-PCR reaction may be performed, as described by K.Q. Hu et al., Virology 181:721-726 (1991), as follows. The extracted mRNA is transcribed in a 25 μl reaction mixture containing 10 mM Tris-HCl, pH 8.3, 5 mM MgC12, 500 μM dNTP, 20 U
RNasin, 1 μM antisense primer and 25 U AMV or MMLV reverse transcriptase. Reverse transcription is performed at 37-45°C for 30-60 min, followed by further incubation at 95°C for 5 min to inactivate the RT. PCR is performed using 10 μl of the cDNA reaction in a final PCR reaction volume of 50 μl containing 10 mM Tris- HCl (pH 8.3), 50 mM KCI, 2 mM MgCl2, 200 μM dNTP, 0.5 μM of each primer and 2.5 U of Taq polymerase. Optimal conditions for cDNA synthesis and thermal cycling can be readily determined by those skilled in the art. The reaction is incubated in a Perkin-Elmer Thermal Cycler 480 or other comparable instrument. Conditions which may be found useful include 30-45 cycles of amplification (94°C, 1 min; 55-70°C, 1 min; 72°C, 2 min), final extension (72°C, 10 min) and soak at 4°C.
C. PCR Fragment Analysis. The correct products then can be verified by size determination using gel electrophoresis with SYBR® Green I nucleic acid gel stain (Molecular Probes, Eugene, OR) and imaged using a STORM imaging system, or also verified by Southern, dot or slot blot analysis using a labeled probe against the internal sequences of the PCR product. The probes also may be polynucleotides analogs, such as morpholinos or peptide nucleic acids analogs (PNAs). Detection of a product comprising a sequence of the PS213 polynucleotide (SEQUENCE ID NO 1), or fragments or complements thereof, is indicative of the presence of PS213 mRNA(s), suggesting a diagnosis of a prostrate tissue disease or condition, such as prostrate cancer.
Example 9: OH-PCR
A. Probe selection and Labeling. Target-specific primers and probes are designed to detect the above-described target sequences by oligonucleotide hybridization PCR. International Publication Nos WO 92/10505, published June 25, 1992, and WO 92/11388, published July 9, 1992, teach methods for labeling oligonucleotides at their 5' and 3' ends, respectively. According to one known method for labeling an oligonucleotide, a label-phosphoramidite reagent is prepared and used to add the label to the oligonucleotide during its synthesis. For example, see N. T. Thuong et al., Tet. Letters 29(46):5905-5908 (1988); or J. S. Cohen et al, published U.S. Patent Application 07/246,688 (NTIS ORDER No. PAT- APPL-7-246,688) (1989). Preferably, probes are labeled at their 3' end to prevent participation in PCR and the formation of undesired extension products. For one step OH-PCR, the probe should have a T at least 15°C below the TM of the primers. The primers and probes are utilized as specific binding members, with or without detectable labels, using standard phosphoramidite chemistry and/or post- synthetic labeling methods which are well-known to one skilled in the art.
B. One Step Oligo Hybridization PCR. OH-PCR is performed on a 200 μl reaction containing 50 mM (N,N,-bis[2-Hydroxyethyl]glycine), pH 8.15, 81.7 mM KOAc. 33.33 mM KOH, 0.01 mg/ml bovine serum albumin, 0.1 mM ethylene diaminetetraacetic acid, 0.02 mg/ml NaN3; 8% w/v glycerol, 150 μM each of dNTP, 0.25 μM each primer, 3.75 nM probe, 5U rTth polymerase, 3.25 mM Mn(OAc)2 and 5 μl blood equivalents of target (see Example 3). Since RNA and the rTth polymerase enzyme are unstable in the presence of Mn(OAc)2, the Mn(OAc)2 should be added just before target addition. The reaction is incubated in a Perkin-Elmer Thermal Cycler 480. Optimal conditions for cDNA synthesis and thermal cycling can be readily determined by those skilled in the art. Conditions which may be found useful include cDNA synthesis (60°C, 30 min), 30-45 amplification cycles (94°C, 40 sec; 55-70°C, 60 sec), oligo-hybridization (97°C, 5 min; 15°C, 5 min; 15°C soak). The correct reaction product contains at least one of the strands of the PCR product and an internally hybridized probe.
C. OH-PCR Product Analysis. Amplified reaction products are detected on an LCx® Analyzer system (available from Abbott Laboratories, Abbott Park, IL). Briefly, the correct reaction product is captured by an antibody labeled microparticle at a capturable site on either the PCR product strand or the hybridization probe, and the complex is detected by binding of a detectable antibody conjugate to either a detectable site on the probe or the PCR strand. Only a complex containing a PCR strand hybridized with the internal probe is detectable. The detection of this complex then is indicative of the presence of PS213 mRNA, suggesting a diagnosis of a prostrate disease or condition, such as prostrate cancer. Many other detection formats exist which can be used and/or modified by those skilled in the art to detect the presence of amplified or non-amplified PS213- derived nucleic acid sequences including, but not limited to, ligase chain reaction (LCR, Abbott Laboratories, Abbott Park, IL); Q-beta replicase (Gene-Trak™, Naperville, Illinois), branched chain reaction (Chiron, Emeryville, CA) and strand displacement assays (Becton Dickinson, Research Triangle Park, NC).
Example 10: Synthetic Peptide Production Synthetic peptides were modeled and then prepared based upon the published amino acid sequence of the PS213 polypeptide consensus sequence (see Example 1). In particular, a number of PS213 peptides derived from SEQUENCE ID NO 6 were prepared, including the peptides of SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, and SEQUENCE ID NO 10. The C-terminal amino acid of the synthetic peptide represented by SEQUENCE ID NO 10 differs from that shown at the corresponding position in SEQUENCE ID NO 6. All peptides were synthesized on a Symphony Peptide Synthesizer (available from Rainin Instrument Co, Emeryville, CA) or similar instrument, using FMOC chemistry, standard cycles and in-situ HBTU activation. Cleavage and deprotection conditions were as follows: a volume of 2.5 ml of cleavage reagent (77.5% v/v trifluoroacetic acid, 15% v/v ethanedithiol, 2.5% v/v water, 5% v/v thioanisole, 1-2% w/v phenol) were added to the resin, and agitated at room temperature for 2-4 hours. The filtrate was then removed and the peptide was precipitated from the cleavage reagent with cold diethyl ether. Each peptide was filtered, purified via reverse-phase preparative HPLC using a water/acetonitrile/0.1 % TFA gradient, and lyophilized. The product was confirmed by mass spectrometry.
The purified peptides were used to immunize animals (see Example 14).
Example 11a: Expression of Protein in a Cell Line Using Plasmid 577 A. Construction of a PS213 Expression Plasmid. Plasmid 577, described in U.S. patent application Serial No. 08/478,073, filed June 7, 1995, has been constructed for the expression of secreted antigens in a permanent cell line. This plasmid contains the following DNA segments: (a) a 2.3 kb fragment of pBR322 containing bacterial beta-lactamase and origin of DNA replication; (b) a 1.8 kb cassette directing expression of a neomycin resistance gene under control of HSV- 1 thymidine kinase promoter and poly-A addition signals; (c) a 1.9 kb cassette directing expression of a dihydrofolate reductase gene under the control of an Simian Virus 40 (SV40) promoter and poly-A addition signals; (d) a 3.5 kb cassette directing expression of a rabbit immunoglobulin heavy chain signal sequence fused to a modified hepatitis C virus (HCV) E2 protein under the control of the Simian Virus 40 T-Ag promoter and transcription enhancer, the hepatitis B virus surface antigen (HBsAg) enhancer I followed by a fragment of Herpes Simplex Virus- 1 (HSV-1) genome providing poly-A addition signals; and (e) a residual 0.7 kb fragment of SV40 genome late region of no function in this plasmid. All of the segments of the vector were assembled by standard methods known to those skilled in the art of molecular biology.
Plasmids for the expression of secretable PS213 proteins are constructed by replacing the hepatitis C virus E2 protein coding sequence in plasmid 577 with that of a PS213 polynucleotide sequence selected from fragments or complements thereof, of the PS213 polynucleotide (SEQUENCE ID NO 1) as follows. Digestion of plasmid 577 with Xbal releases the hepatitis C virus E2 gene fragment. The resulting plasmid backbone allows insertion of the PS213 cDNA insert downstream of the rabbit immunoglobulin heavy chain signal sequence which directs the expressed proteins into the secretory pathway of the cell. The PS213 cDNA fragment is generated by PCR using standard procedures. Encoded in the sense PCR primer sequence is an Xbal site, immediately followed by a 12 nucleotide sequence that encodes the amino acid sequence Ser-Asn-Glu-Leu ("SNEL") to promote signal protease processing, efficient secretion and final product stability in culture fluids. Immediately following this 12 nucleotide sequence the primer contains nucleotides complementary to template sequences encoding amino acids of the PS213 gene . The antisense primer incorporates a sequence encoding the following eight amino acids just before the stop codons: Asp-Tyr-Lys- Asp-Asp- Asp- Asp-Lys (SEQUENCE ID NO 11). Within this sequence is incorporated a recognition site to aid in analysis and purification of the PS213 protein product. A recognition site (termed "FLAG") that is recognized by a commercially available monoclonal antibody designated anti-FLAG M2 (Eastman Kodak, Co., New Haven, CT) can be utilized, as well as other comparable sequences and their corresponding antibodies. For example, PCR is performed using GeneAmp® reagents obtained from Perkin-Elmer-Cetus, as directed by the supplier's instructions. PCR primers are used at a final concentration of 0.5 μM. PCR is performed on the PS213 plasmid template in a 100 μl reaction for 35 cycles (94°C, 30 seconds; 55°C, 30 seconds; 72°C, 90 seconds) followed by an extension cycle of 72°C for 10 min.
B. Transfection of Dihydrofolate Reductase Deficient Chinese Hamster Ovary Cells. The plasmid described supra is transfected into CHO/dhfr- cells [DXB-111, Uriacio et al.. Proc. Natl. Acad. Sci. USA 77:4451-4466 (1980)]. These cells are available from the A.T.C.C, 12301 Parklawn Drive, Rockville, MD 20852, under Accession No. CRL 9096. Transfection is carried out using the cationic liposome-mediated procedure described by P. L. Feigner et al., Proc. Natl. Acad. Sci. USA 84:7413-7417 (1987). Particularly, CHO/dhfr- cells are cultured in Ham's F-12 media supplemented with 10% fetal calf serum, L- glutamine (1 mM) and freshly seeded into a flask at a density of 5 - 8 x 105 cells per flask. The cells are grown to a confluency of between 60 and 80% for transfection. Twenty micrograms (20μg) of plasmid DNA are added to 1.5 ml of Opti-MEM I medium and 100 μl of Lipofectin Reagent (Gibco-BRL; Grand Island, NY) are added to a second 1.5 ml portion of Opti-MEM I media. The two solutions are mixed and incubated at room temperature for 20 min. After the culture medium is removed from the cells, the cells are rinsed 3 times with 5 ml of Opti-MEM I medium. The Opti-MEM I-Lipofection-plasmid DNA solution then is overlaid onto the cells. The cells are incubated for 3 hr at 37°C, after which time the Opti-MEM I-Lipofectin-DNA solution is replaced with culture medium for an additional 24 hr prior to selection.
C. Selection and Amplification. One day after transfection, cells are passaged 1:3 and incubated with dhfr/G418 selection medium (hereafter, "F-12 minus medium G"). Selection medium is Ham's F-12 with L-glutamine and without hypoxanthine, thymidine and glycine (JRH Biosciences, Lenexa, Kansas) and 300 μg per ml G418 (Gibco-BRL; Grand Island, NY). Media volume-to-
2 surface area ratios of 5 ml per 25 cm are maintained. After approximately two weeks, DHFR/G418 cells are expanded to allow passage and continuous maintenance in F-12 minus medium G.
Amplification of each of the transfected PS213 cDNA sequences is achieved by stepwise selection of DHFR , G418 cells with methotrexate (reviewed by R. Schimke, Cell 37:705-713 [1984]). Cells are incubated with F-12 minus medium G containing 150 nM methotrexate (MTX) (Sigma, St. Louis, MO) for approximately two weeks until resistant colonies appear. Further gene amplification is achieved by selection of 150 nM adapted cells with 5 μM MTX. P. Antigen Production. F-12 minus medium G supplemented with 5 μM MTX is overlaid onto just confluent monolayers for 12 to 24 hr at 37°C in 5% CO2. The growth medium is removed and the cells are rinsed 3 times with Dulbecco's phosphate buffered saline (PBS) (with calcium and magnesium) (Gibco-BRL; Grand Island, NY) to remove the remaining media/serum which may be present. Cells then are incubated with VAS custom medium (VAS custom formulation with L-glutamine with HEPES without phenol red, available from JRH Bioscience; Lenexa, KS, product number 52-08678P), for 1 hr at 37°C in 5% CO2. Cells then are overlaid with VAS for production at 5 ml per T flask. Medium is removed after seven days of incubation, retained, and then frozen to await purification with harvests 2, 3 and 4. The monolayers are overlaid with VAS for 3 more seven day harvests. E. Analysis of Prostrate Tissue Gene PS213 Antigen Expression.
Aliquots of VAS supernatants from the cells expressing the PS213 protein construct are analyzed, either by SDS-polyacrylamide gel electrophoresis (SDS- PAGE) using standard methods and reagents known in the art (Laemmli discontinuous gels), or by mass spectrometry. F. Purification. Purification of the PS213 protein containing the FLAG sequence is performed by immunoaffinity chromatography using an affinity matrix comprising anti-FLAG M2 monoclonal antibody covalently attached to agarose by hydrazide linkage (Eastman Kodak Co., New Haven, CT). Prior to affinity purification, protein in pooled VAS medium harvests from roller bottles is exchanged into 50 mM Tris-HCl (pH 7.5), 150 mM NaCl buffer using a Sephadex G-25 (Pharmacia Biotech Inc., Uppsala, Sweden) column. Protein in this buffer is applied to the anti-ET-AG M2 antibody affinity column. Non-binding protein is eluted by washing the column with 50 mM Tris-HCl (pH 7.5), 150 mM NaCl buffer. Bound protein is eluted using an excess of FLAG peptide in 50 mM Tris- HCl (pH 7.5), 150 mM NaCl. The excess FLAG peptide can be removed from the purified PS213 protein by gel electrophoresis or HPLC. Although plasmid 577 is utilized in this example, it is known to those skilled in the art that other comparable expression systems, such as CMV, can be utilized herein with appropriate modifications in reagent and/or techniques and are within the skill of the ordinary artisan.
The largest cloned insert containing the coding region of the PS213 gene is then sub-cloned into either (i) a eukaryotic expression vector which may contain, for example, a cytomegalovirus (CMV) promoter and/or protein fusible sequences which aid in protein expression and detection, or (ii) a bacterial expression vector containing a superoxide-dismutase (SOD) and CMP-KDO synthetase (CKS) or other protein fusion gene for expression of the protein sequence. Methods and vectors which are useful for the production of polypeptides which contain fusion sequences of SOD are described in EPO 0196056, published October 1, 1986, and those containing fusion sequences of CKS are described in EPO Publication No. 0331961, published September 13, 1989. This so-purified protein can be used in a variety of techniques, including, but not limited to animal immunization studies, solid phase immunoassays, etc.
Example 1 lb: Expression of Protein in a Cell Line Using pcDNA3.1/Myc-His A. Construction of a PS213 Expression Plasmid. Plasmid pcDNA3.1/Myc-His (Cat.# V855-20, Invitrogen, Carlsbad, CA) has been constructed, in the past, for the expression of secreted antigens by most mammalian cell lines. Expressed protein inserts are fused to a myc-his peptide tag. The myc-his tag (SEQUENCE ED NO 12) comprises a c-myc oncoprotein epitope and a polyhistidine sequence which are useful for the purification of an expressed fusion protein by using either anti-myc or anti-his affinity columns, or metalloprotein binding columns.
Plasmids for the expression of secretable PS213 proteins are constructed by inserting a PS213 polynucleotide sequence, selected fragments or complements thereof, of the PS213 polynucleotide (SEQUENCE ID NO 1). Prior to construction of a PS213 expression plasmid, the PS213 cDNA sequence is first cloned into a pCR®-Blunt vector as follows:
The PS213 cDNA fragment is generated by PCR using standard procedures. For example, PCR is performed using procedures and reagents from Stratagene®, Inc. (La Jolla, CA), as directed by the manufacturer. PCR primers are used at a final concentration of 0.5 μM. PCR using 5 U of pfu polymerase (Stratagene, La Jolla, CA) is performed on the PS213 plasmid template (see Example 2) in a 50 μl reaction for 30 cycles (94°C, 1 min; 65°C, 1.5 min; 72°C, 3 min) followed by an extension cycle of 72°C for 8 min. (The sense PCR primer sequence comprises nucleotides which are either complementary to the pINCY vector directly upstream of the PS213 gene insert or which incorporate a 5' EcoRI restriction site, an adjacent downstream protein translation consensus initiator, and a 3' nucleic acid sequence which is the same sense as the 5'-most end of the PS213 cDNA insert. The antisense PCR primer incorporates a 5' NotI restriction sequence and a sequence complementary to the 3' end of the PS213 cDNA insert just upstream of the 3'-most, in-frame stop codon.) Five microliters (5 μl) of the resulting blunted-ended PCR product are ligated into 25 ng of linearized pCR®- Blunt vector (Invitrogen, Carlsbad, CA) interrupting the lethal ccdB gene of the vector. The resulting ligated vector is transformed into TOP10 E_. coH (Invitrogen, Carlsbad, CA) using a One Shot™ Transformation Kit (Invitrogen, Carlsbad, CA) following manufacturer's instructions. The transformed cells are grown on LB- Kan (50 μg/ml kanamycin) selection plates at 37°C. Only cells containing a plasmid with an interrupted ccdB gene will grow after transformation [Grant, S.G.N., Proc. Natl. Acad. Sci. USA 87:4645-4649 (1990)1. Transformed colonies are picked and grown up in 3 ml of LB-Kan broth at 37°C. Plasmid DNA is isolated by using a QIAprep® (Qiagen Inc., Santa Clarita, CA) procedure, as directed by the manufacturer. The DNA is cut with EcoRI or SnaBI, and NotI restriction enzymes to release the PS213 insert fragment. The fragment is run on 1% Seakem® LE agarose/0.5 μg/ml ethidium bromide/TE gel, visualized by UV irradiation, excised and purified using QIAquick™ (Qiagen Inc., Santa Clarita, CA) procedures, as directed by the supplier's instructions.
The pcDNA3.1/Myc-His plasmid DNA is linearized by digestion with EcoRI or SnaBI, and NotI in the polylinker region of the plasmid DNA. The resulting plasmid DNA backbone allows insertion of the PS213 purified cDNA fragment, supra, downstream of a CMV promoter which directs expression of the proteins in mammalian cells. The ligated plasmid is transformed into DH5 alpha™ cells (GibcoBRL Grand Island, NY), as directed by the manufacturer. Briefly, 10 ng of pcDNA3.1/Myc-His containing a PS213 insert are added to 50 μl of competent DH5 alpha cells, and the contents are mixed gently. The mixture is incubated on ice for 30 min, heat shocked for 20 sec at 37°C, and placed on ice for an additional 2 min. Upon addition of 0.95 ml of LB medium, the mixture is incubated for 1 hr at 37°C while shaking at 225 rpm. The transformed cells then are plated onto 100 mm LB/Amp (50μg/ml ampicillin) plates and grown at 37°C. Colonies are picked and grown in 3 ml of LB/Amp broth. Plasmid DNA is purified using a QIAprep Kit. The presence of the insert is confirmed using techniques known to those skilled in the art, including, but not limited to restriction digestion and gel analysis. (J. Sambrook et al., supra. )
B. Transfection of Human Embryonic Kidney Cell 293 Cells. The PS213 expression plasmid described in section A, supra, is retransformed into DH5 alpha cells, plated onto LB/ampicillin agar, and grown up in 10 ml of LB/ampicillin broth, as described hereinabove. The plasmid is purified using a QIAfilter™ Maxi Kit (Qiagen, Chatsworth, CA) and is transfected into HEK293 cells [F.L. Graham et al., J. Gen. Vir. 36:59-72 (1977)1. These cells are available from the A.T.C.C, 12301 Parklawn Drive, Rockville, MD 20852, under Accession No. CRL 1573. Transfection is carried out using the cationic lipofectamine-mediated procedure described by P. Hawley-Nelson et al., Focus 15.73 (1993). Particularly, HEK293 cells are cultured in 10 ml DMEM media supplemented with 10% fetal bovine serum (FBS), L-glutamine (2 mM) and freshly seeded into 100 mm culture plates at a density of 9 x 106 cells per plate. The cells are grown at 37 °C to a confluency of between 70% and 80% for transfection. Eight micrograms (8 μg) of plasmid DNA are added to 800 μl of Opti-MEM I® medium (Gibco-BRL, Grand Island, NY), and 48-96 μl of Lipofectamine™ Reagent (Gibco-BRL, Grand
Island, NY) are added to a second 800 μl portion of Opti-MEM I media. The two solutions are mixed and incubated at room temperature for 15-30 min. After the culture medium is removed from the cells, the cells are washed once with 10 ml of serum-free DMEM. The Opti-MEM I-Lipofectamine-plasmid DNA solution is diluted with 6.4 ml of serum-free DMEM and then overlaid onto the cells. The cells are incubated for 5 hr at 37°C, after which time, an additional 8 ml of DMEM with 20% FBS are added. After 18-24 hr, the old medium is aspirated, and the cells are overlaid with 5 ml of fresh DMEM with 5% FBS. Supernatants and cell extracts are analyzed for PS213 gene activity 72 hr after transfection.
C. Analysis of Prostrate Tissue Gene PS213 Antigen Expression. The culture supernatant, supra, is transferred to cryotubes and stored on ice. HEK293 cells are harvested by washing twice with 10 ml of cold Dulbecco's PBS and lysing by addition of 1.5 ml of CAT lysis buffer (Boehringer Mannheim, Indianapolis, IN), followed by incubation for 30 min at room temperature. Lysate is transferred to 1.7 ml polypropylene microfuge tubes and centrifuged at 1000 x g for 10 min. The supernatant is transferred to new cryotubes and stored on ice. Aliquots of supernatants from the cells and the lysate of the cells expressing the PS213 protein construct are analyzed for the presence of PS213 recombinant protein. The aliquots can be run on SDS-polyacrylamide gel electrophoresis (SDS- PAGE) using standard methods and reagents known in the art. (J. Sambrook et al., supra) These gels can then be blotted onto a solid medium such as nitrocellulose, nytran, etc., and the PS213 protein band can be visualized using Western blotting techniques with anti-myc epitope or anti-histidine monoclonal antibodies (Invitrogen, Carlsbad, CA) or anti-PS213 polyclonal serum (see Example 14). Alternatively, the expressed PS213 recombinant protein can be analyzed by mass spectrometry (see Example 12).
D. Purification. Purification of the PS213 recombinant protein containing the myc-his sequence is performed using the Xpress® affinity chromatography system (Invitrogen, Carlsbad, CA) containing a nickel-charged agarose resin which specifically binds polyhistidine residues. Supernatants from 10 x 100 mm plates, prepared as described supra, are pooled and passed over the nickel-charged column. Non-binding protein is eluted by washing the column with 50 mM Tris- HCl (pH 7.5V150 mM NaCl buffer, leaving only the myc-his fusion proteins. Bound PS213 recombinant protein then is eluted from the column using either an excess of imidazole or histidine, or a low pH buffer. Alternatively, the recombinant protein can also be purified by binding at the myc-his sequence to an affinity column consisting of either anti-myc or anti-histidine monoclonal antibodies conjugated through a hydrazide or other linkage to an agarose resin and eluting with an excess of myc peptide or histidine, respectively.
The purified recombinant protein can then be covalently cross-linked to a solid phase, such as N-hydroxysuccinimide-activated sepharose columns
(Pharmacia Biotech, Piscataway, NJ), as directed by supplier's instructions. These columns containing covalently linked PS213 recombinant protein, can then be used to purify anti-PS213 antibodies from rabbit or mouse sera (see Examples 13 and 14).
E. Coating Microtiter Plates with PS213 Expressed Proteins. Supernatant from a 100 mm plate, as described supra, is diluted in an appropriate volume of PBS. Then, 100 μl of the resulting mixture is placed into each well of a Reacti- Bind™ metal chelate microtiter plate (Pierce, Rockford, IL), incubated at room temperature while shaking, and followed by three washes with 200 μl each of PBS with 0.05% Tween® 20. The prepared microtiter plate can then be used to screen polyclonal antisera for the presence of PS213 antibodies (see Example 17). Although pcDNA3.1/Myc-His is utilized in this example, it is known to those skilled in the art that other comparable expression systems can be utilized herein with appropriate modifications in reagent and/or techniques and are within the skill of one of ordinary skill in the art. The largest cloned insert containing the coding region of the PS213 gene is sub-cloned into either (i) a eukaryotic expression vector which may contain, for example, a cytomegalovirus (CMV) promoter and/or protein fusible sequences which aid in protein expression and detection, or (ii) a bacterial expression vector containing a superoxide-dismutase (SOD) and CMP-KDO synthetase (CKS) or other protein fusion gene for expression of the protein sequence. Methods and vectors which are useful for the production of polypeptides which contain fusion sequences of SOD are described in published EPO application No. EP 0 196 056, published October 1, 1986, and vectors containing fusion sequences of CKS are described in published EPO application No. EP 0 331 961, published September 13, 1989. The purified protein can be used in a variety of techniques, including, but not limited to animal immunization studies, solid phase immunoassays, etc.
Example 12: Chemical Analysis of Prostrate Tissue Proteins A. Analysis of Trvptic Peptide Fragments Using MS. Sera from patients with prostrate disease, such as prostrate cancer, sera from patients with no prostrate disease, extracts of prostrate tissues or cells from patients with prostrate disease, such as prostrate cancer, extracts of prostrate tissues or cells from patients with no prostrate disease, and extracts of tissues or cells from other non-diseased or diseased organs of patients are run on a polyacrylamide gel using standard procedures and stained with Coomassie Blue. Sections of the gel suspected of containing the unknown polypeptide are excised and subjected to an in-gel reduction, acetamidation and tryptic digestion. P. Jeno et al., Anal. Bio. 224:451- 455 (1995) and J. Rosenfeld et al., Anal. Bio. 203:173-179 (1992). The gel sections are washed with 100 mM NH4HCO3 and acetonitrile. The shrunken gel pieces are swollen in digestion buffer (50 mM NH4HCO3, 5 mM CaCl2 and 12.5 μg/ml trypsin) at 4°C for 45 min. The supernatant is aspirated and replaced with 5 to 10 μl of digestion buffer without trypsin and allowed to incubate overnight at 37°C. Peptides are extracted with 3 changes of 5% formic acid and acetonitrile and evaporated to dryness. The peptides are adsorbed to approximately 0.1 μl of POROS R2 sorbent (Perseptive Biosystems, Framingham, Massachusetts) trapped in the tip of a drawn gas chromatography capillary tube by dissolving them in 10 μl of 5% formic acid and passing it through the capillary. The adsorbed peptides are washed with water and eluted with 5% formic acid in 60% methanol. The eluant is passed directly into the spraying capillary of an API III mass spectrometer (Perkin- Elmer Sciex, Thornhill, Ontario, Canada) for analysis by nano-electrospray mass spectrometry. M. Wilm et al., Int. J. Mass Spectrom. Ion Process 136:167-180 (1994) and M. Wilm et al., Anal. Chem. 66: 1-8 (1994). The masses of the tryptic peptides are determined from the mass spectrum obtained off the first quadrupole. Masses corresponding to predicted peptides can be further analyzed in MS/MS mode to give the amino acid sequence of the peptide.
B. Peptide Fragment Analysis Using LC/MS. The presence of polypeptides predicted from mRNA sequences found in hyperplastic disease tissues also can be confirmed using liquid chromatography /tandem mass spectrometry (LC/MS/MS). D. Hess et al., METHODS. A Companion to Methods in Enzymology 6:227-238 (1994). The serum specimen or tumor extract from the patient is denatured with SDS and reduced with dithiothreitol (1.5 mg/ml) for 30 min at 90°C followed by alkylation with iodoacetamide (4 mg/ml) for 15 min at 25°C. Following acrylamide electrophoresis, the polypeptides are electroblotted to a cationic membrane and stained with Coomassie Blue. Following staining, the membranes are washed and sections thought to contain the unknown polypeptides are cut out and dissected into small pieces. The membranes are placed in 500 μl microcentrifuge tubes and immersed in 10 to 20 μl of proteolytic digestion buffer (100 mM Tris-HCl, pH 8.2, containing 0.1 M NaCl, 10% acetonitrile, 2 mM
CaCl2 and 5 μg/ml trypsin) (Sigma, St. Louis, MO). After 15 hr at 37°C, 3 μl of saturated urea and 1 μl of 100 μg/ml trypsin are added and incubated for an additional 5 hr at 37°C. The digestion mixture is acidified with 3 μl of 10% trifluoroacetic acid and centrifuged to separate supernatant from membrane. The supernatant is injected directly onto a microbore, reverse phase HPLC column and eluted with a linear gradient of acetonitrile in 0.05% trifluoroacetic acid. The eluate is fed directly into an electrospray mass spectrometer, after passing though a stream splitter if necessary to adjust the volume of material. The data is analyzed following the procedures set forth in Example 12, Section A.
Example 13: Gene Immunization Protocol A. In Vivo Antigen Expression. Gene immunization circumvents protein purification steps by directly expressing an antigen jn vivo after inoculation of the appropriate expression vector. Also, production of antigen by this method may allow correct protein folding and glycosylation since the protein is produced in mammalian tissue. The method utilizes insertion of the gene sequence into a plasmid which contains a CMV promoter, expansion and purification of the plasmid and injection of the plasmid DNA into the muscle tissue of an animal. Preferred animals include mice and rabbits. See, for example, H. Davis et al., Human Molecular Genetics 2:1847-1851 (1993). After one or two booster immunizations, the animal can then be bled, ascites fluid collected, or the animal's spleen can be harvested for production of hybridomas. B. Plasmid Preparation and Purification. PS213 cDN A sequences are generated from the PS213 cDNA-containing vector using appropriate PCR primers containing suitable 5' restriction sites following the procedures described in Example 11. The PCR product is cut with appropriate restriction enzymes and inserted into a vector which contains the CMV promoter (for example, pRc/CMV or pcDNA3 vectors from Invitrogen, San Diego, CA). This plasmid then is expanded in the appropriate bacterial strain and purified from the cell lysate using a CsCl gradient or a Qiagen plasmid DNA purification column. All these techniques are familiar to one of ordinary skill in the art of molecular biology.
C. Immunization Protocol. Anesthetized animals are immunized intramuscularly with 0.1 - 100 μg of the purified plasmid diluted in PBS or other DNA uptake enhancers (Cardiotoxin, 25% sucrose). See, for example, H. Davis et al., Human Gene Therapy 4:733-740 (1993); and P. W. Wolff et al, Biotechniques 11:474-485 (1991). One to two booster injections are given at monthly intervals. D. Testing and Use of Antiserum. Animals are bled and the resultant sera tested for antibody using peptides synthesized from the known gene sequence (see Example 16) using techniques known in the art, such as Western blotting or ELA techniques. Antisera produced by this method can then be used to detect the presence of the antigen in a patient's tissue or cell extract or in a patient's serum by ELISA or Western blotting techniques, such as those described in Examples 15 through 18.
Example 14: Production of Antibodies Against PS213 A. Production of Polyclonal Antisera. Antiserum against PS213 was prepared by injecting rabbits with peptides whose sequences were derived from that of the predicted amino acid sequence of the PS213 published nucleotide sequence (SEQUENCE ID NO 1). The synthesis of peptides (SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10) is described in Example 10. Peptides used as immunogens were not conjugated to a carrier such as keyhole limpet hemocyanine, KLH, (i.e., they were unconjugated.).
Animal Immunization. Female white New Zealand rabbits weighing 2 kg or more were used for raising polyclonal antiserum. One animal was immunized per unconjugated peptide (SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10). One week prior to the first immunization, 5 to 10 ml of blood were obtained from the animal to serve as a non-immune prebleed sample. Unconjugated peptides, SEQUENCE ID NO 7, SEQUENCE ID NO 8,
SEQUENCE ID NO 9, and SEQUENCE ID NO 10, were used to prepare the primary immunogen by emulsifying 0.5 ml of the peptide at a concentration of 2 mg/ml in PBS (pH 7.2) which contained 0.5 ml of complete Freund's adjuvant (CFA) (Difco, Detroit, MI). The immunogen was injected into several sites of the animal via subcutaneous, intraperitoneal, and intramuscular routes of administration. Four weeks following the primary immunization, a booster immunization was administered. The immunogen used for the booster immunization dose was prepared by emulsifying 0.5 ml of the same unconjugated peptide used for the primary immunogen, except that the peptide now was diluted to 1 mg/ml with 0.5 ml of incomplete Freund's adjuvant (IF A) (Difco, Detroit, MI). Again, the booster dose was administered into several sites via subcutaneous, intraperitoneal and intramuscular types of injections. The animals were bled (5 ml) two weeks after the booster immunizations and each serum was tested for immunoreactivity to the peptide as described below. The booster and bleed schedule was repeated at 4 week intervals until an adequate titer was obtained. The titer or concentration of antiserum was determined by using unconjugated peptides in a microtiter EIA as described in Example 17, below. An antibody titer of 1 :500 or greater was considered an adequate titer for further use and study.
Table 2. Titer of rabbit anti-PS213 peptide antisera (12 week bleed)
Peptide Immunogen Titer
SEQUENCE ID NO 7 52,000
SEQUENCE ID NO 8 50,000
SEQUENCE ID NO 9 46,500
SEQUENCE ID NO 10 >62,500
B. Production of Monoclonal Antibody.
1. Immunization Protocol. Mice are immunized using peptides which can either be conjugated to a carrier such as KLH [prepared as described hereinbelow, or unconjugated (i.e., not conjugated to a carrier such as KLH)] except that the amount of the unconjugated or conjugated peptide for monoclonal antibody production in mice is one-tenth the amount used to produce polyclonal antisera in rabbits. Thus, the primary immunogen consists of 100 μg of unconjugated or conjugated peptide in 0.1 ml of CFA emulsion while the immunogen used for booster immunizations consists of 50 μg of unconjugated or conjugated peptide in 0.1 ml of FA. Hybridomas for the generation of monoclonal antibodies are prepared and screened using standard techniques. The methods used for monoclonal antibody development follow procedures known in the art such as those detailed in Kohler and Milstein, Nature 256:494 (1975) and reviewed in J.G.R. Hurrel, ed., Monoclonal Hybridoma Antibodies: Techniques and Applications. CRC Press, Inc., Boca Raton, FL (1982). Another method of monoclonal antibody development which is based on the Kohler and Milstein method is that of L.T. Mimms et al., Virology 176:604-619 (1990).
The immunization regimen (per mouse) consists of a primary immunization with additional booster immunizations. The primary immunogen used for the primary immunization consists of 100 μg of unconjugated or conjugated peptide in 50 μl of PBS (pH 7.2) previously emulsified in 50 μl of CFA. Booster immunizations performed at approximately two weeks and four weeks post primary immunization consist of 50 μg of unconjugated or conjugated peptide in 50 μl of PBS (pH 7.2) emulsified with 50 μl IFA. A total of 100 μl of this immunogen are inoculated intraperitoneally and subcutaneously into each mouse. Individual mice are screened for immune response by microtiter plate enzyme immunoassay (EIA) as described in Example 17 approximately four weeks after the third immunization. Mice are inoculated either intravenously, intrasplenically or intraperitoneally with 50 μg of unconjugated or conjugated peptide in PBS (pH 7.2) approximately fifteen weeks after the third immunization..
Three days after this intravenous boost, splenocytes are fused with, for example, Sp2/0-Agl4 myeloma cells (Milstein Laboratories, England) using the polyethylene glycol (PEG) method. The fusions are cultured in Iscove's Modified Dulbecco's Medium (IMDM) containing 10% fetal calf serum (FCS), plus 1 % hypoxan thine, aminopterin and thymidine (HAT). Bulk cultures were screened by microtiter plate EIA following the protocol in Example 17. Clones reactive with the peptide used an immunogen and non-reactive with other peptides (i.e., peptides of PS213 not used as the immunogen) are selected for final expansion. Clones thus selected are expanded, aliquoted and frozen in IMDM containing 10% FCS and 10% dimethyl sulfoxide, (DMSO).
2. Peptide Conjugation. Peptide is conjugated to maleimide activated KLH (commercially available as Imject®, available from Pierce Chemical Company, Rockford, IL). Imject® contains about 250 moles of reactive maleimide groups per mole of hemocyanine. The activated KLH is dissolved in phosphate buffered saline (PBS, pH 8.4) at a concentration of about 7.7 mg/ml. The peptide is conjugated through cysteines occurring in the peptide sequence, or to a cysteine previously added to the synthesized peptide in order to provide a point of attachment. The peptide is dissolved in DMSO (Sigma Chemical Company, St. Louis, MO) and reacted with the activated KLH at a mole ratio of about 1.5 moles of peptide per mole of reactive maleimide attached to the KLH. A procedure for the conjugation of peptide is provided hereinbelow. It is known to the ordinary artisan that the amounts, times and conditions of such a procedure can be varied to optimize peptide conjugation.
The conjugation reaction described hereinbelow is based on obtaining 3 mg of KLH peptide conjugate ("conjugated peptide"), which contains about 0.77 μmoles of reactive maleimide groups. This quantity of peptide conjugate usually is adequate for one primary injection and four booster injections for production of polyclonal antisera in a rabbit. Briefly, peptide is dissolved in DMSO at a concentration of 1.16 μmoles/100 μl of DMSO. One hundred microliters (100 μl) of the DMSO solution are added to 380 μl of the activated KLH solution prepared as described hereinabove, and 20 μl of PBS (pH 8.4) are added to bring the volume to 500 μl. The reaction is incubated overnight at room temperature with stirring. The extent of reaction is determined by measuring the amount of unreacted thiol in the reaction mixture. The difference between the starting concentration of thiol and the final concentration is assumed to be the concentration of peptide which has coupled to the activated KLH. The amount of remaining thiol is measured using Ellman's reagent (5,5'-dithiobis(2-nitrobenzoic acid), Pierce Chemical Company, Rockford, IL). Cysteine standards are made at a concentration of 0, 0.1, 0.5, 2, 5 and 20 mM by dissolving 35 mg of cysteine HC1 (Pierce Chemical Company, Rockford, IL) in 10 ml of PBS (pH 7.2) and diluting the stock solution to the desired concentration(s). The photometric determination of the concentration of thiol is accomplished by placing 200 μl of PBS (pH 8.4) in each well of an Immulon 2® microwell plate (Dynex Technologies, Chantilly, VA). Next, 10 μl of standard or reaction mixture are added to each well. Finally, 20 μl of Ellman's reagent at a concentration of 1 mg/ml in PBS (pH 8.4) are added to each well. The wells are incubated for 10 minutes at room temperature, and the absorbance of all wells is read at 415 nm with a microplate reader (such as the
BioRad Model 3550, BioRad, Richmond, CA). The absorbance of the standards is used to construct a standard curve and the thiol concentration of the reaction mixture is determined from the standard curve. A decrease in the concentration of free thiol is indicative of a successful conjugation reaction. Unreacted peptide is removed by dialysis against PBS (pH 7.2) at room temperature for 6 hours. The conjugate is stored at 2-8°C if it is to be used immediately; otherwise, it is stored at -20°C or colder. 3. Production of Ascites Fluid Containing Monoclonal Antibodies.
Frozen hybridoma cells prepared as described hereinabove are thawed and placed into expansion culture. Viable hybridoma cells are inoculated intraperitoneally into Pristane treated mice. Ascitic fluid is removed from the mice, pooled, filtered through a 0.2 μ filter and subjected to an immunoglobulin class G (IgG) analysis to determine the volume of the Protein A column required for the purification. 4. Purification of Monoclonal Antibodies From Ascites Fluid. Briefly, filtered and thawed ascites fluid is mixed with an equal volume of Protein A sepharose binding buffer (1.5 M glycine, 3.0 M NaCl, pH 8.9) and refiltered through a 0.2 μ filter. The volume of the Protein A column is determined by the quantity of IgG present in the ascites fluid. The eluate then is dialyzed against PBS (pH 7.2) overnight at 2-8°C. The dialyzed monoclonal antibody is sterile filtered and dispensed in aliquots. The immunoreactivity of the purified monoclonal antibody is confirmed by determining its ability to specifically bind to the peptide used as the immunogen by use of the EIA microtiter plate assay procedure of Example 17. The specificity of the purified monoclonal antibody is confirmed by determining its lack of binding to irrelevant peptides such as peptides of PS213 not used as the immunogen. The purified anti-PS213 monoclonal thus prepared and characterized is placed at either 2-8°C for short term storage or at -80°C for long term storage. 5. Further Characterization of Monoclonal Antibody. The isotype and subtype of the monoclonal antibody produced as described hereinabove can be determined using commercially available kits (available from Amersham. Inc., Arlington Heights, IL). Stability testing also can be performed on the monoclonal antibody by placing an aliquot of the monoclonal antibody in continuous storage at 2-8°C and assaying optical density (OD) readings throughout the course of a given period of time.
C. Use of Recombinant Proteins as Immunogens. It is within the scope of the present invention that recombinant proteins made as described herein can be utilized as immunogens in the production of polyclonal and monoclonal antibodies, with corresponding changes in reagents and techniques known to those skilled in the art. Example 15: Purification of Serum Antibodies Which Specifically
Bind to PS213 Peptides Immune sera, obtained as described hereinabove in Examples 13 and/or 14, is affinity purified using immobilized synthetic peptides prepared as described in Example 10, or recombinant proteins prepared as described in Example 11. An IgG fraction of the antiserum is obtained by passing the diluted, crude antiserum over a Protein A column (Affi-Gel protein A, Bio-Rad, Hercules, CA). Elution with a buffer (Binding Buffer, supplied by the manufacturer) removes substantially all proteins that are not immunoglobulins. Elution with 0.1M buffered glycine (pH 3) gives an immunoglobulin preparation that is substantially free of albumin and other serum proteins.
Immunoaffinity chromatography is performed to obtain a preparation with a higher fraction of specific antigen-binding antibody. The peptide used to raise the antiserum is immobilized on a chromatography resin, and the specific antibodies directed against its epitopes are adsorbed to the resin. After washing away non- binding components, the specific antibodies are eluted with 0.1 M glycine buffer, pH 2.3. Antibody fractions are immediately neutralized with 1.0M Tris buffer (pH 8.0) to preserve immunoreactivity. The chromatography resin chosen depends on the reactive groups present in the peptide. If the peptide has an amino group, a resin such as Affi-Gel 10 or Affi-Gel 15 is used (Bio-Rad, Hercules, CA). If coupling through a carboxy group on the peptide is desired, Affi-Gel 102 can be used (Bio-Rad, Hercules, CA). If the peptide has a free sulfhydryl group, an organomercurial resin such as Affi-Gel 501 can be used (Bio-Rad, Hercules, CA). Alternatively, spleens can be harvested and used in the production of hybridomas to produce monoclonal antibodies following routine methods known in the art as described hereinabove.
Example 16: Western Blotting of Tissue Samples Protein extracts were prepared by homogenizing tissue samples in 0.1 M Tris-HCl (pH 7.5), 15% (w/v) glycerol, 0.2 mM EDTA, 1.0 mM 1 ,4- dithiothreitol, 10 μg/ml leupeptin, 0.1% Triton X-100 , and 1.0 mM phenylmethylsulfonylfluoride [Kain et al., Biotechniques. 17:982 (1994)]. Following homogenization, the homogenates were centrifuged at 4°C for 5 minutes to separate supernatant from debris. For protein quantitation, 3-10 μl of supernatant were added to 1.5 ml of bicinchoninic acid reagent (Sigma, St. Louis, MO), and the resulting absorbance at 562 nm were measured.
For SDS-PAGE, samples were adjusted to desired protein concentration with Tricine Buffer (Novex, San Diego, CA), mixed with an equal volume of 2X Tricine sample buffer (Novex, San Diego, CA), and heated for 5 minutes at 100°C in a thermal cycler. Samples were then applied to a Novex 10-20% Precast Tricine Gel for electrophoresis. Following electrophoresis samples were transferred from the gels to nitrocellulose membranes in Novex Tris-Glycine Transfer buffer. Membranes were then probed with specific anti-peptide antibodies and the reagents and procedures provided in the Western Lights Plus or Western Lights (Tropix, Bedford, MA) chemiluminesence detection kits. Chemiluminesent bands were visualized by exposing the developed membranes to Hyperfilm ECL (Amersham, Arlington Heights, IL). The bands were also visualized directly on the membranes by the addition and development of chromogenic substrate 5-bromo-4-chloro-3-indolyl phosphate (BCIP). This chromogenic solution contains 0.016% BCIP, 100 mM NaCl, 5 mM MgCl2, and 100 mM Tris-HCl, pH 9.5. The filter was incubated in the chromogenic solution at room temperature until the bands developed to the desired intensity. Molecular mass determination was made based upon the mobility of pre- stained molecular weight standards (Novex, San Diego, CA) and biotinylated molecular weight standards (Tropix, Bedford, MA).
Figure 1 shows the results of the Western blot performed on a panel of tissue extracts using antiserum against PS213 synthetic peptide, SEQUENCE ID NO 10 (see Example 14). Each lane of Figure 1 represents a different tissue protein extract: 1, endometrial cancer; 2, ovarian cancer; 3, normal bladder; 4, lung cancer; 5, normal colon; 6, BPH; 7, BPH; 8, prostate cancer; 9, blank; 10 molecular weight markers (kD). A strong band appears between 148 kD and 60 kD in the prostate cancer (lane 8, indicated by an arrow) and one of the two BPH extracts (lane 6). A faint band is seen at the same position in the other BPH sample (lane 7) and also in the lung cancer (lane 4).
Example 17: EIA Microtiter Plate Assay The immunoreactivity of antiserum preferably obtained from rabbits as described in Example 14 was determined by means of a microtiter plate EIA, as follows. Briefly, synthetic peptides, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, and SEQUENCE ID NO 10, prepared as described in Example 10, were dissolved in carbonate buffer (50 mM, pH 9.6) to a final concentration of 2 μg/ml. Next, 100 μl of the peptide or protein solution were placed in each well of an Immulon 2® microtiter plate (Dynex Technologies, Chantilly, VA). The plate was incubated overnight at room temperature and then washed four times with deionized water. The wells were blocked by adding 125 μl of a suitable protein blocking agent, such as Superblock® (Pierce Chemical Company, Rockford, IL), to each well and then immediately discarding the solution. This blocking procedure was performed three times. Antiserum obtained from immunized rabbits or mice, prepared as previously described, was diluted in a protein blocking agent (e.g., a 3% Superblock® solution) in PBS containing 0.05% Tween-20® (monolaurate polyoxyethylene ether) (Sigma Chemical Company, St. Louis, MO) and 0.05% sodium azide at dilutions of 1:100, 1:500, 1:2500, 1:12,500, and 1:62,500 and placed in each well of the coated microtiter plate. The wells then were incubated for three hours at room temperature. Each well was washed four times with deionized water. One hundred microliters of alkaline phosphatase-conjugated goat anti-rabbit IgG or goat anti-mouse IgG antiserum (Southern Biotech, Birmingham, AB) diluted 1:2000 in 3% Superblock® solution in phosphate buffered saline containing 0.05% Tween 20® and 0.05% sodium azide, were added to each well. The wells were incubated for two hours at room temperature. Next, each well was washed four times with deionized water. One hundred microliters of paranitrophenyl phosphate substrate (I irkegaard and Perry Laboratories, Gaithersburg, MD) then were added to each well. The wells were incubated for thirty minutes at room temperature. The absorbance at 405 nm was read in each well. Positive reactions were identified by an increase in absorbance at 405 nm in the test well above that absorbance given by a non- immune serum (negative control). A positive reaction was indicative of the presence of detectable anti-PS213 antibodies. Titers of the anti-peptide antisera were calculated from the previously described dilutions of antisera and defined as the calculated dilution, where A405nm=0.5 OD.
Example 18: Coating of Solid Phase Particles A. Coating of Microparticles with Antibodies Which Specifically Bind to PS213 Antigen. Affinity purified antibodies which specifically bind to PS213 protein (see Example 15) are coated onto microparticles of polystyrene, carboxylated polystyrene, polymethylacrylate or similar particles having a radius in the range of about 0.1 to 20 μm. Microparticles may be either passively or actively coated. One coating method comprises coating ED AC (l-(3- dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (Aldrich Chemical Co., Milwaukee, WI) activated carboxylated latex microparticles with antibodies which specifically bind to PS213 protein, as follows. Briefly, a final 0.375% solid suspension of resin washed carboxylated latex microparticles (available from Bangs Laboratories, Carmel, IN or Serodyn, Indianapolis, IN) are mixed in a solution containing 50 mM MES buffer, pH 4.0 and 150 mg/1 of affinity purified anti-PS213 antibody (see Example 14) for 15 min in an appropriate container. ED AC coupling agent is added to a final concentration of 5.5 μg/ml to the mixture and mixed for 2.5 hr at room temperature.
The microparticles then are washed with 8 volumes of a Tween 20®/sodium phosphate wash buffer (pH 7.2) by tangential flow filtration using a 0.2 μm Microgon Filtration module. Washed microparticles are stored in an appropriate buffer which usually contains a dilute surfactant and irrelevant protein as a blocking agent, until needed.
B. Coating of 1/4 Inch Beads. Antibodies which specifically bind to PS213-antigen also may be coated on the surface of 1/4 inch polystyrene beads by routine methods known in the art (Snitman et al., US Patent 5,273,882) and used in competitive binding or EIA sandwich assays.
Polystyrene beads first are cleaned by ultrasonicating them for about 15 seconds in 10 mM NaHCO3 buffer at pH 8.0. The beads then are washed in deionized water until all fines are removed. Beads then are immersed in an antibody solution in 10 mM carbonate buffer, pH 8 to 9.5. The antibody solution can be as dilute as 1 μg/ml in the case of high affinity monoclonal antibodies or as concentrated as about 500 μg/ml for polyclonal antibodies which have not been affinity purified. Beads are coated for at least 12 hours at room temperature, and then they are washed with deionized water. Beads may be air dried or stored wet (in PBS, pH 7.4). They also may be overcoated with protein stabilizers (such as sucrose) or protein blocking agents used as non-specific binding blockers (such as irrelevant proteins, Carnation skim milk, Superblock®, or the like).
Example 19: Microparticle Enzyme Immunoassay (MEIA)
PS213 antigens are detected in patient test samples by performing a standard antigen competition EIA or antibody sandwich EIA and utilizing a solid phase such as microparticles (MEIA). The assay can be performed on an automated analyzer such as the IMx® Analyzer (Abbott Laboratories, Abbott Park, IL).
A. Antibody Sandwich EIA. Briefly, samples suspected of containing PS213 antigen are incubated in the presence of anti-PS213 antibody-coated microparticles (prepared as described in Example 17) in order to form antigen/antibody complexes. The microparticles then are washed and an indicator reagent comprising an antibody conjugated to a signal generating compound (i.e., enzymes such as alkaline phosphatase or horseradish peroxide) is added to the antigen/antibody complexes or the microparticles and incubated. The microparticles are washed and the bound antibody/antigen/antibody complexes are detected by adding a substrate (e.g., 4-methyl umbelliferyl phosphate (MUP), or OPD/peroxide, respectively), that reacts with the signal generating compound to generate a measurable signal. An elevated signal in the test sample, compared to the signal generated by a negative control, detects the presence of PS213 antigen. The presence of PS213 antigen in the test sample is indicative of a diagnosis of a prostrate disease or condition, such as prostrate cancer.
B. Competitive Binding Assay. The competitive binding assay uses a peptide or protein that generates a measurable signal when the labeled peptide is contacted with an anti-peptide antibody coated microparticle. This assay can be performed on the IMx® Analyzer (available from Abbott Laboratories, Abbott Park, IL). The labeled peptide is added to the PS213 antibody -coated microparticles (prepared as described in Example 17) in the presence of a test sample suspected of containing PS213 antigen, and incubated for a time and under conditions sufficient to form labeled PS213 peptide (or labeled protein) / bound antibody complexes and/or patient PS213 antigen / bound antibody complexes. The PS213 antigen in the test sample competes with the labeled PS213 peptide (or PS213 protein) for binding sites on the microparticle. PS213 antigen in the test sample results in a lowered binding of labeled peptide and antibody coated microparticles in the assay since antigen in the test sample and the PS213 peptide or PS213 protein compete for antibody binding sites. A lowered signal (compared to a control) indicates the presence of PS213 antigen in the test sample. The presence of PS213 antigen suggests the diagnosis of a prostrate disease or condition, such as prostrate cancer.
The PS213 polynucleotides and the proteins encoded thereby which are provided and discussed hereinabove are useful as markers of prostrate tissue disease, especially prostrate cancer. Tests based upon the appearance of this marker in a test sample such as blood, plasma or serum can provide low cost, non- invasive, diagnostic information to aid the physician to make a diagnosis of cancer, to help select a therapy protocol, or to monitor the success of a chosen therapy. This marker may appear in readily accessible body fluids such as blood, urine or stool as antigens derived from the diseased tissue which are detectable by immunological methods. This marker may be elevated in a disease state, altered in a disease state, or be a normal protein of the prostrate which appears in an inappropriate body compartment.
Example 20: Immunohistochemical Detection of PS213 Protein Antiserum against a PS213 synthetic peptide derived from the consensus peptide sequence (SEQUENCE ED NO 10) described in Example 14, above, is to immunohistochemically stain a variety of normal and diseased tissues using standard procedures. Briefly, frozen blocks of tissue are cut into 6 micron sections, and placed on microscope slides. After fixation in cold acetone, the sections are dried at room temperature, then washed with phosphate buffered saline and blocked. The slides are incubated with the antiserum against a synthetic peptide derived from the consensus PS213 peptide sequence (SEQUENCE ED NO 10) at a dilution of 1:500, washed, incubated with biotinylated goat anti-rabbit antibody, washed again, and incubated with avidin labeled with horseradish peroxidase. After a final wash, the slides are incubated with 3-amino-9- ethylcarbazole substrate which gives a red stain. The slides are counterstained with hematoxylin, mounted, and examined under a microscope by a pathologist.

Claims

We Claim:
1 . A method of detecting the presence of a target PS213 polynucleotide in a test sample, said method comprising: (a) contacting the test sample with at least one PS213-specific polynucleotide or complement thereof, wherein said PS213-specific polynucleotide has at least 50% identity with (i) SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1, or (iii) complements of (i) or (ii); and
(b) detecting the presence of target PS213 polynucleotides from the test sample which bind to said PS213-specific polynucleotide.
2. The method of claim 1 , wherein said target PS213 polynucleotide is attached to a solid phase prior to performing step (a).
3. The method of claim 1, wherein said PS213-specific polynucleotide is attached to a solid phase prior to performing step (a).
4. A method for detecting PS213 mRNA in a test sample, said method comprising: (a) performing reverse transcription on said sample using at least one primer in order to produce cDNA;
(b) amplifying the cDNA obtained from step (a) using PS213 oligonucleotides as sense and antisense primers to obtain PS213 amplicon; and
(c) detecting the presence of said PS213 amplicon, wherein the PS213 oligonucleotides utilized in steps (a) and (b) have at least 50% identity with (i)
SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1, or (iii) complements of (i) or (ii).
5. The method of claim 4, wherein said test sample is reacted with a solid phase prior to performing one of steps (a), (b), or (c).
6. The method of claim 4, wherein said detection step comprises utilizing a detectable label capable of generating a measurable signal.
7. A method of detecting a target PS213 polynucleotide in a test sample suspected of containing said target polynucleotide, comprising: (a) contacting the test sample with at least one PS213 oligonucleotide as a sense primer and with at least one PS213 oligonucleotide as an anti-sense primer and amplifying to obtain a first stage reaction product;
(b) contacting said first stage reaction product with at least one other PS213 oligonucleotide to obtain a second stage reaction product, with the proviso that the other PS213 oligonucleotide is located 3' to the PS213 oligonucleotides utilized in step (a) and is complementary to said first stage reaction product; and
(c) detecting said second stage reaction product as an indication of the presence of the target PS213 polynucleotide, wherein the PS213 oligonucleotides utilized in steps (a) and (b) have at least 50% identity with (i) SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1, or (iii) complements of (i) or (ii).
8. The method of claim 7, wherein said test sample is reacted with a solid phase prior to performing one of steps (a), (b), or (c).
9. The method of claim 7, wherein said detection step comprises utilizing a detectable label capable of generating a measurable signal.
10. The method of claim 9, wherein said detectable label is reacted to a solid phase.
1 1. A test kit useful for detecting a PS213 polynucleotide in a test sample, said test kit comprising a container containing at least one PS213 polynucleotide having at least 50% identity with (i) SEQUENCE ID NO 1, (ii) fragments of SEQUENCE ID NO 1 , or (iii) complements of (i) or (ii).
12. A purified polynucleotide derived from a PS213 nucleic acid molecule, wherein said polynucleotide encodes a polypeptide which has at least 50% identity with a polypeptide having an amino acid sequence selected from the group consisting of SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof
13. The polynucleotide of claim 12, wherein said polynucleotide hybridizes selectively to a PS213 nucleic acid sequence.
14. The polynucleotide of claim 12, wherein said polynucleotide has an overall length of about 20 to about 50 nucleotides.
15. The polynucleotide of claim 12, wherein said polynucleotide has an overall length of about 10 to about 25 nucleotides.
16. The polynucleotide of claim 12, wherein said polynucleotide is produced by recombinant techniques.
17. The polynucleotide of claim 12, wherein said polynucleotide is produced by synthetic techniques.
18. The polynucleotide of claim 12, wherein said polynucleotide comprises a sequence encoding at least one PS213 epitope.
19. The polynucleotide of claim 12, wherein said polynucleotide is attached to a solid phase.
20. The polynucleotide of claim 19, wherein said solid phase comprises an array of polynucleotide molecules attached thereto.
21 . A recombinant expression system comprising a polynucleotide according to claim 12, operably linked to a control sequence compatible with a desired host.
22. A cell transfected with the recombinant expression system of claim 21.
23. A PS213 polypeptide having at least 50% identity with an amino acid sequence selected from the group consisting of SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
24. The polypeptide of claim 23, wherein said polypeptide is produced by recombinant techniques.
25. The polypeptide of claim 23, wherein said polypeptide is produced by synthetic techniques.
26. A specific binding molecule which binds to at least one PS213 epitope, wherein said PS213 epitope is derived from an amino acid sequence having at least 50% identity with an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
27. The specific binding molecule of claim 26, wherein said molecule is an antibody molecule.
28. A test kit for determining the presence of PS213 antigen or anti- PS213 antibody in a test sample, said kit comprising a container containing a PS213 polypeptide having at least 50% identity with an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
29. The test kit of claim 28, wherein said PS213 polypeptide is attached to a solid phase.
30. A test kit for determining the presence of PS213 antigen in a test sample, said kit comprising a container containing a specific binding molecule which binds to a PS213 antigen having at least one PS213 epitope.
31. The kit of claim 30, wherein said specific binding molecule is attached to a solid phase.
32. A method for producing a polypeptide comprising at least one
PS213 epitope, said method comprising incubating host cells that have been transfected with an expression vector containing a polynucleotide sequence encoding a polypeptide, wherein said polypeptide comprises an amino acid sequence having at least 50% identity with an amino acid sequence selected from the group consisting of SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
33. A method for detecting PS213 antigen in a test sample suspected of containing said PS213 antigen, comprising: (a) contacting the test sample with a specific binding molecule which binds to at least one epitope of a PS213 antigen selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof, wherein said contacting is carried out for a time and under conditions sufficient for the formation of binding molecule/antigen complexes; and
(b) detecting the presence of said complexes as an indication of the presence of said PS213 antigen.
34. The method of claim 33, wherein said specific binding molecule is an antibody molecule or a fragment thereof.
35. The method of claim 33, wherein said specific binding molecule is attached to a solid phase.
36. A method for detecting the presence of antibodies specific for a PS213 antigen in a test sample suspected of containing such antibodies, said method comprising: (a) contacting the test sample with a PS213 polypeptide, wherein said
PS213 polypeptide contains at least one PS213 epitope derived from an amino acid sequence having at least 50% identity with an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof, and further wherein said contacting is carried out for a time and under conditions sufficient to allow antigen/antibody complexes to form; and
(b) detecting the presence of said complexes as an indication of the presence of antibodies specific for a PS213 antigen.
37. The method of claim 36, wherein said PS213 polypeptide is attached to a solid phase.
38. A cell transfected with a polynucleotide according to claim 12, wherein said polynucleotide encodes at least one PS213 epitope.
39. A method for producing antibodies which specifically bind to PS213 antigen, comprising administering to an individual an isolated immunogenic polypeptide or fragment thereof in an amount sufficient to elicit an immune response, wherein said immunogenic polypeptide comprises at least one PS213 epitope and has at least 50% identity with a sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
40. A method for producing antibodies which specifically bind to PS213 antigen, comprising administering to an individual a plasmid comprising a sequence which encodes at least one PS213 epitope derived from a polypeptide having an amino acid sequence selected from the group consisting of SEQUENCE ID NO 6, SEQUENCE ID NO 7, SEQUENCE ID NO 8, SEQUENCE ID NO 9, SEQUENCE ID NO 10, and fragments thereof.
41. The test kit of claim 11 further comprising a container with tools useful for collection of said sample, wherein the tools are selected from the group consisting of lancets, absorbent paper, cloth, swabs and cups.
42. The test kit of claim 28 further comprising a container with tools useful for collection of said sample, wherein the tools are selected from the group consisting of lancets, absorbent paper, cloth, swabs and cups.
43. The test kit of claim 30 further comprising a container with tools useful for collection of said sample, wherein the tools are selected from the group consisting of lancets, absorbent paper, cloth, swabs and cups.
44. The test kit of claim 30, wherein said specific binding molecule is an antibody or fragment thereof.
45. The method of claim 1 , wherein the presence of said target PS213 polynucleotide in the test sample is indicative of prostrate disease.
46. The method of claim 4, wherein the presence of said amplicon is indicative of prostrate disease.
47. The method of claim 7, wherein the presence of said second stage reaction product is indicative of prostrate disease.
48. The method of claim 33, wherein detection of said complexes is indicative of prostrate disease.
49. The method of claim 36, wherein detection of said complexes is indicative of prostrate disease.
PCT/US1998/018983 1997-09-11 1998-09-11 Reagents and methods useful for detecting diseases of the prostate WO1999013111A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CA002303404A CA2303404A1 (en) 1997-09-11 1998-09-11 Reagents and methods useful for detecting diseases of the prostate
EP98948162A EP1012343A1 (en) 1997-09-11 1998-09-11 Reagents and methods useful for detecting diseases of the prostate

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US92797897A 1997-09-11 1997-09-11
US08/927,978 1997-09-11

Publications (1)

Publication Number Publication Date
WO1999013111A1 true WO1999013111A1 (en) 1999-03-18

Family

ID=25455530

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/018983 WO1999013111A1 (en) 1997-09-11 1998-09-11 Reagents and methods useful for detecting diseases of the prostate

Country Status (3)

Country Link
EP (1) EP1012343A1 (en)
CA (1) CA2303404A1 (en)
WO (1) WO1999013111A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000061765A2 (en) * 1999-04-12 2000-10-19 Lexicon Genetics Incorporated Lipoxygenase proteins and polynucleotides encoding the same
WO2001090323A2 (en) * 2000-05-19 2001-11-29 Millennium Pharmaceuticals, Inc. 46638, a putative family member of human lipoxygenase

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994010343A1 (en) * 1992-10-29 1994-05-11 Thomas Jefferson University Methods of detecting micrometastasis of prostate cancer
WO1995025178A1 (en) * 1994-03-14 1995-09-21 Biomide Investment Limited Partnership Methods for detecting 12-lipoxygenase and its encoding dna

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994010343A1 (en) * 1992-10-29 1994-05-11 Thomas Jefferson University Methods of detecting micrometastasis of prostate cancer
WO1995025178A1 (en) * 1994-03-14 1995-09-21 Biomide Investment Limited Partnership Methods for detecting 12-lipoxygenase and its encoding dna

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BRASH A R ET AL: "Discovery of a second 15S-lipoxygenase in humans.", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA, (1997 JUN 10) 94 (12) 6148-52, United States, XP002089902 *
DATABASE EMBL Entry HSU78294; 3 July 1997 (1997-07-03), "Homo sapiens 15S-lipoxygenase mRNA, complete cds.", XP002089906 *
DATABASE EMBL ENTRY R45395; 11 July 1994 (1994-07-11), "Prolyl endopeptidase inhibitor peptide-4", XP002089905 *
SPINDLER, S. A. (1) ET AL: "What is the role of 13-hydroxyoctadecadienoic acid in prostate cancer.", MOLECULAR BIOLOGY OF THE CELL, (1996) VOL. 7, NO. SUPPL., PP. 492A. MEETING INFO.: ANNUAL MEETING OF THE 6TH INTERNATIONAL CONGRESS ON CELL BIOLOGY AND THE 36TH AMERICAN SOCIETY FOR CELL BIOLOGY SAN FRANCISCO, CALIFORNIA, USA DECEMBER 7-11, 1996 ISSN, XP002089903 *
SPINDLER, STEPHEN A. ET AL: "Production of 13-hydroxyoctadecadienoic acid (13-HODE) by prostate tumors and cell lines.", BIOCHEMICAL AND BIOPHYSICAL RESEARCH COMMUNICATIONS, (OCT. 29, 1997) VOL. 239, NO. 3, PP. 775-781, XP002089904 *

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000061765A2 (en) * 1999-04-12 2000-10-19 Lexicon Genetics Incorporated Lipoxygenase proteins and polynucleotides encoding the same
WO2000061765A3 (en) * 1999-04-12 2001-02-22 Lexicon Genetics Inc Lipoxygenase proteins and polynucleotides encoding the same
US6582957B1 (en) 1999-04-12 2003-06-24 Lexicon Genetics Incorporated Lipoxygenase proteins and polynucleotides encoding the same
US7144730B2 (en) 1999-04-12 2006-12-05 Lexicon Genetics Incorporated Lipoxygenase proteins and polynucleotides encoding the same
WO2001090323A2 (en) * 2000-05-19 2001-11-29 Millennium Pharmaceuticals, Inc. 46638, a putative family member of human lipoxygenase
WO2001090323A3 (en) * 2000-05-19 2002-07-18 Millennium Pharm Inc 46638, a putative family member of human lipoxygenase

Also Published As

Publication number Publication date
CA2303404A1 (en) 1999-03-18
EP1012343A1 (en) 2000-06-28

Similar Documents

Publication Publication Date Title
US6890749B2 (en) Reagents and methods useful for detecting diseases of the prostate
US6252047B1 (en) Reagents and methods useful for detecting diseases of the prostate
US6465181B2 (en) Reagents and methods useful for detecting diseases of the prostate
US20060154291A1 (en) Reagents and methods useful for detecting diseases of the breast
EP1032706A1 (en) Reagents and methods useful for detecting diseases of the breast
WO1999002714A1 (en) Reagents and methods useful for detecting diseases of the breast
US20050170406A1 (en) Reagents and methods useful for detecting diseases of the urinary tract
US20020045164A1 (en) Reagents and methods useful for detecting diseases of the lung
EP1003779A1 (en) Reagents and methods useful for detecting diseases of the breast
US20060029963A1 (en) Reagents and methods useful for detecting diseases of the breast
US20030130492A1 (en) Reagents and methods useful for detecting diseases of the breast
US20020037503A1 (en) Reagents and methods useful for detecting diseases of the breast
WO1999022027A1 (en) Reagents and methods useful for detecting diseases of the breast
WO1999031274A2 (en) Reagents and methods useful for detecting diseases of the pancreas
EP1012343A1 (en) Reagents and methods useful for detecting diseases of the prostate
US20020188114A1 (en) Reagents and methods useful for detectings diseases of the lung
US20020188115A1 (en) Reagents and methods useful for detecting diseases of the breast
WO1999034017A1 (en) Reagents and methods useful for detecting diseases of the breast
WO1999014357A9 (en) Reagents and methods useful for detecting diseases of the prostate, breast and ovary

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

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

Ref document number: 1998948162

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2303404

Country of ref document: CA

Ref country code: CA

Ref document number: 2303404

Kind code of ref document: A

Format of ref document f/p: F

WWP Wipo information: published in national office

Ref document number: 1998948162

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 1998948162

Country of ref document: EP