AU2001249411B9 - Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides,and diagnostic and therapeutic methods - Google Patents

Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides,and diagnostic and therapeutic methods Download PDF

Info

Publication number
AU2001249411B9
AU2001249411B9 AU2001249411A AU2001249411A AU2001249411B9 AU 2001249411 B9 AU2001249411 B9 AU 2001249411B9 AU 2001249411 A AU2001249411 A AU 2001249411A AU 2001249411 A AU2001249411 A AU 2001249411A AU 2001249411 B9 AU2001249411 B9 AU 2001249411B9
Authority
AU
Australia
Prior art keywords
specific
prostate
testis
nucleic acid
polypeptide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Ceased
Application number
AU2001249411A
Other versions
AU2001249411B2 (en
AU2001249411A1 (en
Inventor
Fahri Saatcioglu
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Application granted granted Critical
Publication of AU2001249411B9 publication Critical patent/AU2001249411B9/en
Publication of AU2001249411A1 publication Critical patent/AU2001249411A1/en
Publication of AU2001249411B2 publication Critical patent/AU2001249411B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Description

16/01 2007 17:11 FAX 61 3 92438333 GRIFFITH HACK IPAUSTRALIA Ia008/02B 0 NOVEL PROSTATE-SPECIFIC OR TESTIS-SPECIFIC NUCLEIC ACID MOLECULES, POLYPEPTIDES. AND DIAGNOSTIC AND THERAPEUTIC METHODS Field of The Invention The invention generally relates to the treatment of disorders associated with Oprostate and testis dysfunction and cell proliferation, and specifically relates to the Cl identification and use of novel genes for diagnosis and treatment of such disorders.
oBackground of The Invention All references, including any patents or patent applications, cited in this specification are hereby incorporated by reference. No admission is made that any reference constitutes prior art. The discussion of the references states what their authors assert, and the applicants reserve the right to challenge the accuracy and pertinency of the cited documents. It will be clearly understood that, although a number of prior art is publications are referred to herein, this reference does not constitute an admission that any of these documents forms part of the common general knowledge in the art, in Australia or in any other country.
Genitourinary disorders are often difficult to diagnose and treat effectively because they are present non-specifically. Two causes of genitourinary disorders are disorders of the prostate gland and the testis.
The prostate is a variable sized gland located in the male pelvis, and is made up of several different cell types, including epithelial cells and stromal cells. Prostateassociated disorders include prostate cancer, benign prostatic hyperplasia, and prostatitis. The male hormone testosterone and other androgen related hormones have major roles in the growth and function of the prostate. The testis is also subject to many defects, including developmental anomalies, inflammation, and cancer.
In men, prostate cancer is the most commonly diagnosed cancer and the second leading cause of cancer mortality following skin cancer. [n the initial stages, prostate cancer is dependent on androgens for growth, and this dependence is the basis for androgen ablation therapy. In most cases, however, prostate cancer progresses to an androgen-independent phenotype, for which there is no effective therapy available at present.
M 'lA.e¢ gda' Paunt 70tI 7 I 71 ea.,J AP4it 4.AU SpoGilisim 2007-l -1o dc IfrOIO7 COMS ID No: SBMI-05942216 Received by IP Australia: Time 17:21 Date 2007-01-16 WO 01/72962 PCT/USU1/09410 Currently, there is limited information regarding the molecular details of prostate cancer progression. Several independent approaches resulted in the identification of a few highly prostate-enriched genes that may have unique roles in this process. The first such gene discovered was Prostate Specific Antigen (PSA), the detection of which is currently used as a diagnostic tool and also as a marker for the progression of prostate cancer, albeit with significant limitations. More recently, several additional prostate-enriched genes were identified including prostate-specific membrane antigen (PSMA), prostate carcinoma tumor antigen 1 (PCTA-1), NKX3.1, prostate stem cell antigen (PSCA), DD3, and PCGEM1.
It would be beneficial to provide reagents useful for the diagnosis and therapy of disorders associated with the prostate and the testis, as well as other tissues.
Summary of the Invention The invention provides, in general, a novel prostate-specific or testis-specific nucleic acid molecules, polypeptides, antibodies, and modulatory compounds for use in methods of diagnosing, treating, and preventing diseases and conditions of the prostate and testis, such as cancer.
In a first aspect the invention provides a substantially pure prostatespecific or testis-specific polypeptide, including a sequence substantially identical to the sequence of any of SEQ ID NOS: 14, 29, 32, 34, 36, 41, or 53. In a preferred embodiment of the first aspect, the substantially pure prostate-specific or testis-specific polypeptide includes the sequence of any of SEQ ID NOS: 14, 29, 32, 34, 36, 41, or 53. In another preferred embodiment, the invention provides an isolated nucleic acid molecule encoding a polypeptide of the first aspect, for example a nucleic acid WO 01/72962 PCT/USU1/09410 molecule including the sequence of any of SEQ ID NOS: 23, 28, 31, 33, or 52. Preferably, the polypeptide is derived from a mammal, a human.
In a second aspect, the invention provides an isolated prostatespecific or testis-specific nucleic acid molecule including a sequence substantially identical to SEQ ID NOS: 1-12, 22, 27, 30, and 51.
In a third aspect, the invention provides an isolated prostate-specific or testis-specific nucleic acid molecule consisting essentially of SEQ ID NOS: 15-21, 24-26, 42-50, and 54-70.
In preferred embodiments of some of the above aspects, the invention provides a vector, a cell, a cell including the vector, and a nonhuman transgenic animal including the isolated nucleic acid molecules.
In a fourth aspect, the invention provides an isolated nucleic acid molecule that hybridizes under high stringency conditions to the complement of any of the sequences set forth in SEQ ID NOS: 1-12, 15-28, 31, 33, 35, 40, 42-50, 51, 52, or 54-70, where the isolated nucleic acid molecule encodes a prostate-specific or testis-specific polypeptide.
In a fifth aspect, the invention provides an isolated nucleic acid molecule, where the nucleic acid molecule includes a sequence that is antisense to the coding strand of any of the prostate-specific or testisspecific nucleic acid molecules set forth in SEQ ID NOS: 1-12, 15-28, 31, 33, 35, 40, 42-50, 51, 52, or 54-70, or a fragment thereof.
In a sixth aspect, the invention provides a probe for analyzing a prostate-specific or testis-specific gene or homolog or fragment thereof, the probe having greater than 55% nucleotide sequence identity to a sequence encoding any of SEQ ID NOS: 1-12, 15-28, 30, 31, 33, 35, 40, 42-50, 51, 52, or 54-70, or fragment thereof, where the fragment includes at least six amino acids, and the probe hybridizes under high stringency conditions to at least a portion of a prostate-specific or testis-specific nucleic acid WO 01/72962 PCT/USU1/09410 molecule. In a preferred embodiment of this aspect, the probe has 100% complementarity to a nucleic acid molecule encoding any of SEQ ID NOS: 1-12, 15-28, 30, 31, 33, 35, 40, 42-50, 51, 52, or 54-70, or fragment thereof, where the fragment comprises at least six amino acids, and said probe hybridizes under high stringency conditions to at least a portion of a prostate-specific or testis-specific nucleic acid molecule.
In a seventh aspect, the invention provides an antibody that specifically binds to a prostate-specific or testis-specific polypeptide that includes an amino acid sequence that is substantially identical to the amino acid sequence of any of SEQ ID NOS: 14, 29, 32, 34, 36, 41, or 53.
In an eighth aspect, the invention provides a method of detecting a prostate-specific or testis-specific gene or fragment thereof in a cell, the method including contacting the nucleic acid molecule of any of SEQ ID NOS: 1-12, 15-28, 30, 31, 33, 35, 40, 42-50, 51, 52, or 54-70, or a fragment thereof, where the fragment is greater than about 18 nucleotides in length, with a preparation of genomic DNA from the cell, under high stringency hybridization conditions, and detecting DNA sequences having about or greater nucleotide sequence identity to any of SEQ ID NOS: 1-12, 15-28, 31, 33, 35, 40, 42-50, 51, 52, or 54-70, thus identifying a prostatespecific or testis-specific gene or fragment thereof. Nucleotides encoding the polypeptides of SEQ ID NOS: 38, 39, or 71-73 can also be used in an embodiment of this aspect. In a preferred embodiment of this aspect, the method includes detecting a neoplastic or cancer cell in a patient predisposed to or at risk for cancer, for example, for prostate cancer.
In a ninth aspect, the invention provides a method for identifying a test compound that modulates the expression or activity of a prostatespecific or testis-specific polypeptide, the method including contacting the prostate-specific or testis-specific polypeptide with the test compound, and determining the effect of the test compound on the prostate-specific or WO 01/72962 PCT/USU1/09410 testis-specific polypeptide expression or activity. In a preferred embodiments of this aspect, the prostate-specific or testis-specific polypeptide includes an amino acid sequence substantially identical to the amino acid sequence of SEQ ID NOS: 14, 29, 32, 34, 36, 38, 39, 41, 53, or 71-73, and fragments and analogs thereof.
In a tenth aspect, the invention provides a method of treating a mammal having a disorder of the prostate or testis, the method including administering to the mammal a therapeutically effective amount of a compound that modulates the activity or expression of a prostate-specific or testis-specific polypeptide, where the compound has a beneficial effect on the disorder in the mammal. In preferred embodiments of this aspect, the disorder is prostate cancer, the mammal is a human, or the prostate-specific or testis-specific polypeptide includes an amino acid sequence substantially identical to the amino acid sequence of SEQ ID NOS: 14, 29, 32, 34, 36, 38, 39, 41, 53, or 71-73, and fragments and analogs thereof.
In an eleventh aspect, the invention provides a pharmaceutical composition including at least one dose of a therapeutically effective amount of a prostate-specific or testis-specific polypeptide or fragment thereof, in a pharmaceutically acceptable carrier, the composition being formulated for the treatment of a disorder of the prostate or testis.
In a twelfth aspect, the invention provides a kit for the analysis of a prostate-specific or testis-specific nucleic acid molecule, the kit including a nucleic acid molecule probe for analyzing a prostate-specific or testisspecific nucleic acid molecule present in a test subject.
In a thirteenth aspect, the invention provides a kit for the analysis of a prostate-specific or testis-specific polypeptide, the kit including an antibody for analyzing a prostate-specific or testis-specific polypeptide present in a test subject.
WO 01/72962 PCT/USU1/09410 As used herein, by "polypeptide," "protein," or "polypeptide fragment" is meant a chain of two or more amino acids, regardless of any post-translational modification glycosylation or phosphorylation), constituting all or part of a naturally or non-naturally occurring polypeptide. By "post-translational modification" is meant any change to a polypeptide or polypeptide fragment during or after synthesis. Posttranslational modifications can be produced naturally (such as during synthesis within a cell) or generated artificially (such as by recombinant or chemical means). A protein can be made up of one or more polypeptides.
By "substantially pure polypeptide" or "substantially pure and isolated polypeptide" is meant a polypeptide (or a fragment thereof) that has been separated from components that naturally accompany it.
Typically, the polypeptide is substantially pure when it is at least 60%, by weight, free from the proteins and naturally occurring organic molecules with which it is naturally associated. Preferably, the polypeptide is a prostate-specific or a testis-specific polypeptide that is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight, pure.
A substantially pure prostate-specific or a testis-specific polypeptide may be obtained by standard techniques, for example, by extraction from a natural source prostate or testis tissue or cell lines), by expression of a recombinant nucleic acid encoding a prostate-specific or a testis-specific polypeptide, or by chemically synthesizing the polypeptide. Purity can be measured by any appropriate method, by column chromatography, polyacrylamide gel electrophoresis, or HPLC analysis.
A protein or polypeptide is substantially free of naturally associated components when it is separated from those contaminants that accompany it in its natural state. Thus, a protein that is chemically synthesized or produced in a cellular system different from the cell from which it naturally originates will be substantially free from its naturally associated WO 01/72962 PCT/USU1/09410 components. Accordingly, substantially pure polypeptides not only include those derived from eukaryotic organisms but also those synthesized in E.
coli or other prokaryotes.
The term "identity" is used herein to describe the relationship of the sequence of a particular nucleic acid molecule or polypeptide to the sequence of a reference molecule of the same type. For example, if a polypeptide or nucleic acid molecule has the same amino acid or nucleotide residue at a given position, compared to a reference molecule to which it is aligned, there is said to be "identity" at that position.
The level of sequence identity of a nucleic acid molecule or a polypeptide to a reference molecule is typically measured using sequence analysis software with the default parameters specified therein, such as the introduction of gaps to achieve an optimal alignment. The "identity" of two or more nucleic acid or polypeptide sequences can therefore be readily calculated by known methods, including but not limited to those described in Computational Molecular Biology, Lesk, ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, and Griffin, eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, Academic Press, 1987; and Sequence Analysis Primer, Gribskov, and Devereux, eds., M. Stockton Press, New York, 1991; and Carillo and Lipman, SIAM J. Applied Math. 48:1073, 1988.
Methods to determine identity are available in publicly available computer programs. Computer program methods to determine identity between two sequences include, but are not limited to, the GCG program package (Devereux et al., Nucleic Acids Research 12(1): 387, 1984), BLASTP, BLASTN, and FASTA (Altschul et al., J. Mol. Biol. 215: 403 (1990). The well known Smith Waterman algorithm may also be used to WO 01/72962 PCT/USU1/09410 determine identity. The BLAST program is publicly available from NCBI and other sources (BLAST Manual, Altschul, et al., NCBI NLM NIH Bethesda, MD 20894). Searches can be performed in URLs such as the following http://www.ncbi.nlm.nih.gov/BLAST/unfinishedgenome.html; or http://www.tigr.org/cgi-bin/BlastSearch/blast.cgi. These software programs match similar sequences by assigning degrees of homology to various substitutions, deletions, and other modifications. Conservative substitutions typically include substitutions within the following groups: glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid, asparagine, glutamine; serine, threonine; lysine, arginine; and phenylalanine, tyrosine.
A nucleic acid molecule or polypeptide is said to be "substantially identical" to a reference molecule if it exhibits, over its entire length, at least 50%, 60%, or 70%, preferably at least 80% or 90%, more preferably at least 95%, and most preferably at least 99% identity to the sequence of the reference molecule. For polypeptides, the length of comparison sequences is at least 16 amino acids, preferably at least 20 amino acids or at least amino acids, more preferably at least 35 amino acids, and most preferably, the full-length polypeptide. For nucleic acid molecules, the length of comparison sequences is at least 50 nucleotides, preferably at least 60 nucleotides, more preferably at least 75 nucleotides or at least 110 nucleotides, and most preferably, the full-length nucleic acid molecule.
Alternatively, or additionally, two nucleic acid sequences are "substantially identical" if they hybridize under high stringency conditions.
By "isolated nucleic acid molecule," "substantially pure nucleic acid molecule," or "substantially pure and isolated nucleic acid molecule" is meant a nucleic acid molecule (for example, DNA) that is free of the genes that, in the naturally occurring genome of the organism from which the nucleic acid molecule of the invention is derived, flank the nucleic acid.
The term includes, for example, a recombinant DNA that is incorporated WO 01/72962 PCT/USU1/09410 into a vector; into an autonomously replicating plasmid or virus; or into the genomic DNA of a prokaryote or eukaryote; or that exists as a separate molecule a cDNA or a genomic or cDNA fragment produced by PCR or restriction endonuclease digestion) independent of other sequences. It also includes a recombinant DNA that is part of a hybrid gene encoding additional polypeptide sequence.
By "antisense," as used herein in reference to nucleic acid molecules, is meant a molecule having a nucleic acid sequence, regardless of length, that is complementary to at least 75 nucleotides, and preferably at least 100, 150, or 200 nucleotides, of the coding strand of a nucleic acid molecule encoding a prostate-specific or a testis-specific polypeptide, as described herein. An antisense molecule may also include regulatory sequences such as transcription enhancers, hormone responsive elements, ribosomal- and RNA polymerase binding sites, etc., which may be located upstream or downstream of the coding region, and may have a distance of several ten base pairs to several ten thousand base pairs. An antisense nucleic acid molecule can be, for example, capable of preferentially lowering the production or expression of a prostate-specific or a testisspecific polypeptide encoded by a prostate-specific or a testis-specific nucleic acid molecule.
By "prostate-specific" or "testis-specific" nucleic acid molecule is meant a nucleic acid molecule, such as a genomic DNA, cDNA, or RNA mRNA) molecule, having at least 50, 60, or 75%, more preferably at least 80, 85, or 95%, and most preferably at least 99% amino acid identity to the nucleic acid molecules described herein, for example, in Figures 4, 11, and 14. In addition, a nucleic acid molecule having at least 50, 60, or more preferably at least 80, 85, or 95%, and most preferably at least 99% nucleotide identity to a nucleotide sequence encoding amino acids 1- 200 ofSTMP1 (SEQ ID NO: 14), preferably encoding amino acids 40-150 WO 01/72962 PCT/USU1/09410 of STMP1, can be considered a prostate-specific or testis-specific nucleic acid molecule. Specifically excluded from this definition is STEAP (AF186249) (Hubert, R. S. et al., Proc NatlAcad Sci US A 96, 14523- 14528, 1999) and nucleic acid molecule sequences set forth in or encoding ESTs AF132025, AF177862, BAB23615, BAA91839, BAB15559, and NP 032190.
A preferred prostate-specific nucleic acid molecule may be preferentially expressed in prostate tissue at a level that is at least higher, preferably at least 10-fold higher, more preferably at least higher, and most preferably at least 20-fold higher than the level of the same nucleic acid molecule in at least one non-prostate tissue, preferably in all other non-prostate tissues. A prostate-specific nucleic acid molecule can also be expressed at high levels in a non-prostate tissue although, generally, the level of expression will be the highest in the prostate. Occasionally, as described herein, a prostate-specific nucleic acid molecule will be expressed at higher levels in non-prostate tissue placenta, lung, or liver) than in the prostate.
A preferred testis-specific nucleic acid molecule may be preferentially expressed in testis tissue at a level that is at least higher, preferably at least 10-fold higher, more preferably at least higher, and most preferably at least 20-fold higher than the level of the same nucleic acid molecule in at least one non- testis tissue, preferably in all other non- testis tissues. A testis -specific nucleic acid molecule can also be expressed at high levels in a non- testis tissue although, generally, the level of expression will be the highest in the testis. Occasionally, as described herein, a testis -specific nucleic acid molecule will be expressed at higher levels in non- testis tissue placenta, lung, or liver) than in the testis.
WO 01/72962 PCT/USU1/09410 By "prostate-specific" or a "testis-specific" polypeptide or "prostatespecific" or a "testis-specific" protein is meant a polypeptide that is encoded by a prostate-specific or a testis-specific nucleic acid molecule. A prostate-specific or testis-specific polypeptide may also be defined as a polypeptide having at least 50, 60, or 75%, more preferably at least 80, or 95%, and most preferably at least 99% amino acid identity to the polypeptides described herein, for example, in Figures 4, 11, and 14.
Specifically excluded from this definition is STEAP (AF186249) (Hubert, R. S. et al., Proc NatlAcad Sci USA 96, 14523-14528, 1999) and polypeptide sequences set forth in or encoded by ESTs AF132025, AF177862, BAB23615, BAA91839, BAB15559, and NP_032190. In addition, a polypeptide having at least 50, 60, or 75%, more preferably at least 80, 85, or 95%, and most preferably at least 99% amino acid identity to amino acids 1-200 of STMP1 (SEQ ID NO: 14), preferably amino acids 40-150 of STMP1, can be considered a prostate-specific or testis-specific polypeptide.
A preferred prostate-specific polypeptide is preferentially expressed in prostate tissue at a level that is at least 5-fold higher, preferably at least higher, more preferably at least 15-fold higher, and most preferably at least 20-fold higher than the level of the same polypeptide in at least one non-prostate tissue, preferably in all other non-prostate tissues. A prostatespecific polypeptide can also be expressed at high levels in a non-prostate tissue although, generally, the level of expression will be the highest in the prostate. Occasionally, as described herein, a prostate-specific polypeptide will be expressed at higher levels in non-prostate placenta, lung, liver) than in the prostate.
A preferred testis-specific polypeptide is preferentially expressed in testis tissue at a level that is at least 5-fold higher, preferably at least fold higher, more preferably at least 15-fold higher, and most preferably at WO 01/72962 PCT/USU1/09410 least 20-fold higher than the level of the same polypeptide in at least one non- testis tissue, preferably in all other non-testis tissues. A testis-specific polypeptide can also be expressed at high levels in a non- testis tissue although, generally, the level of expression will be the highest in the testis.
Occasionally, as described herein, a testis-specific polypeptide will be expressed at higher levels in non- testis placenta, lung, liver) than in the testis.
The term prostate-specific or testis-specific polypeptide includes homologs, analogs, fragments, and isoforms, alternatively spliced isoforms, of the sequences described herein. By "biologically active fragment" is meant a polypeptide fragment of a prostate-specific or testisspecific polypeptide that exhibits, for example, extracellular trafficking, cell signaling, or other properties that are at least 30%, preferably at least more preferably at least 75%, and most preferably at least 100%, compared with the properties of a full length prostate-specific or testis-specific polypeptide. By "analog" is meant any substitution, addition, or deletion in the amino acid sequence of a prostate-specific or testis-specific polypeptide that exhibits properties that are at least 30%, preferably at least 50%, more preferably at least 75%, and most preferably at least 100%, compared with the extracellular trafficking or cell signaling properties of the polypeptide from which it is derived. Fragments, homologs, and analogs can be generated using standard techniques, for example, solid phase peptide synthesis or polymerase chain reaction. For example, point mutations may arise at any position of the sequence from an apurinic, apyrimidinic, or otherwise structurally impaired site within the cDNA. Alternatively, point mutations may be introduced by random or site-directed mutagenesis procedures oligonucleotide assisted or by error prone PCR).
Likewise, deletions and/or insertions may be introduced into the sequences, and preferred insertions comprise and/or 3'-fusions with a WO 01/72962 PCT/USU1/09410 polynucleotide that encodes a reporter moiety or an affinity moiety. Other preferred insertions comprise a nucleic acid that further includes functional elements such as a promoter, enhancer, hormone responsive element, origin of replication, transcription and translation initiation sites, etc. It should be appreciated that where insertions with one or more functional elements are present, the resulting nucleic acid may be linear or circular transcription or expression cassettes, plasmids, etc.).
For use in the methods of the invention, the terms "prostate-specific" or "testis-specific" polypeptide further include the polypeptide sequences set forth in or encoded by ESTs AF132025, AF177862, BAB23615, BAA91839, BAB15559, and NP_032190, but does not include STEAP, and a prostate-specific or testis-specific nucleic acid molecule includes the nucleotide sequences set forth in or encoding ESTs AF132025, AF177862, BAB23615, BAA91839, BAB15559, and NP_032190, but does not include
STEAP.
By "prostate-specific or a testis-specific gene or homolog or fragment thereof' is meant a gene, or homolog of a gene, that encodes a prostate-specific or testis-specific polypeptide.
By "specifically binds" is meant a compound, an antibody, that recognizes and binds a protein or polypeptide, for example, a prostatespecific or a testis-specific polypeptide, and that when detectably labeled can be competed away for binding to that protein or polypeptide by an excess of compound that is not detectably labeled. A compound that nonspecifically binds is not competed away by excess detectably labeled compound. A preferred antibody binds to any prostate-specific or a testisspecific polypeptide sequence that is substantially identical to any of the polypeptide sequences set forth in Figures 4, 11, and 14, or encoded by any of the nucleotide sequences set forth in Figures 3, 4, 11, and 14, or portions thereof.
WO 01/72962 PCT/USU1/09410 By a "compound," "test compound," or "candidate compound" is meant a molecule, be it naturally-occurring or artificially-derived, and includes, for example, peptides, proteins, synthetic organic molecules, naturally-occurring organic molecules, nucleic acid molecules, and components thereof.
By "high stringency conditions" is meant conditions that allow hybridization comparable with the hybridization that occurs using a DNA probe of at least 500 nucleotides in length, in a buffer containing 0.5 M NaHPO 4 pH 7.2, 7% SDS, 1 mM EDTA, and 1% BSA (fraction at a temperature of 65 0 C, or a buffer containing 48% formamide, 4.8X SSC, 0.2 M Tris-C1, pH 7.6, 1X Denhardt's solution, 10% dextran sulfate, and 0.1% SDS, at a temperature of 42°C (these are typical conditions for high stringency Northern or Southern hybridizations). High stringency hybridization is also relied upon for the success of numerous techniques routinely performed by molecular biologists, such as high stringency PCR, DNA sequencing, single strand conformational polymorphism analysis, and in situ hybridization. In contrast to Northern and Southern hybridizations, these techniques are usually performed with relatively short probes usually 16 nucleotides or longer for PCR or sequencing, and 40 nucleotides or longer for in situ hybridization). The high stringency conditions used in these techniques are well known to those skilled in the art of molecular biology, and may be found, for example, in Ausubel et al., Current Protocols in Molecular Biology, John Wiley Sons, New York, NY, 1998, hereby incorporated by reference.
By "probe" or "primer" is meant a single-stranded DNA or RNA molecule of defined sequence that can base pair to a second DNA or RNA molecule that contains a complementary sequence ("target"). The stability of the resulting hybrid depends upon the extent of the base pairing that occurs. This stability is affected by parameters such as the degree of WO 01/72962 PCT/USU1/09410 complementarity between the probe and target molecule, and the degree of stringency of the hybridization conditions. The degree of hybridization stringency is affected by parameters such as the temperature, salt concentration, and concentration of organic molecules, such as formamide, and is determined by methods that are well known to those skilled in the art. Probes or primers specific for prostate-specific or a testis-specific nucleic acid molecules, preferably, have greater than 45% sequence identity, more preferably at least 55-75% sequence identity, still more preferably at least 75-85% sequence identity, yet more preferably at least 85-99% sequence identity, and most preferably 100% sequence identity to the nucleic acid sequences encoding the amino acid sequences described herein. Probes can be detectably-labeled, either radioactively or nonradioactively, by methods that are well-known to those skilled in the art.
Probes can be used for methods involving nucleic acid hybridization, such as nucleic acid sequencing, nucleic acid amplification by the polymerase chain reaction, single stranded conformational polymorphism (SSCP) analysis, restriction fragment polymorphism (RFLP) analysis, Southern hybridization, northern hybridization, in situ hybridization, electrophoretic mobility shift assay (EMSA), and other methods that are well known to those skilled in the art.
A molecule, an oligonucleotide probe or primer, a gene or fragment thereof, a cDNA molecule, a polypeptide, or an antibody, can be said to be "detectably-labeled" if it is marked in such a way that its presence can be directly identified in a sample. Methods for detectablylabeling molecules are well known in the art and include, without limitation, radioactive labeling with an isotope, such as 32 P or 3 S) and nonradioactive labeling with a fluorescent label, such as fluorescein, or by generating a construct containing green fluorescent protein (GFP)).
WO 01/72962 PCT/USU1/09410 By "transgenic" is meant any cell that includes a DNA sequence or transgene that is inserted by artifice into a cell and becomes part of the genome of the organism that develops from that cell. As used herein, the transgenic organisms are generally transgenic mammals mice, rats, and goats) and the DNA (transgene) is inserted by artifice into the nuclear genome. By "transgene" is meant any piece of DNA that is inserted by artifice into a cell, and becomes part of the genome of the organism that develops from that cell. Such a transgene may include a gene that is partly or entirely heterologous foreign) to the transgenic organism, or may represent a gene homologous to an endogenous gene of the organism. By "knockout mutation" is meant an artificially induced alteration in the nucleic acid sequence (created via recombinant DNA technology or deliberate exposure to a mutagen) that reduces the biological activity of the polypeptide normally encoded therefrom by at least 80% relative to the unmutated gene. The mutation may, without limitation, be an insertion, deletion, frameshift mutation, or a missense mutation. The knockout mutation can be in a cell ex vivo a tissue culture cell or a primary cell) or in vivo. A "knockout animal" is a mammal, preferably, a mouse, containing a knockout mutation as defined above.
By "sample" is meait a tissue biopsy, cells, blood, serum, urine, stool, or other specimen obtained from a patient or test subject. The sample is analyzed to detect a mutation in a gene encoding a prostate-specific or a testis-specific polypeptide, or expression levels of a gene encoding a prostate-specific or a testis-specific polypeptide, as for example, an indication of the progression of cancer, by methods that are known in the art or described herein. For example, methods such as sequencing, singlestrand conformational polymorphism (SSCP) analysis, or restriction fragment length polymorphism (RFLP) analysis of PCR products derived from a patient sample may be used to detect a mutation in a gene encoding WO 01/72962 PCT/US01/09410 a prostate-specific or a testis-specific polypeptide; ELISA may be used to measure levels of a prostate-specific or a testis-specific polypeptide; and PCR may be used to measure the level of nucleic acids encoding a prostatespecific or a testis-specific polypeptide.
By "pharmaceutically acceptable carrier" is meant a carrier that is physiologically acceptable to the treated mammal while retaining the therapeutic properties of the compound with which it is administered. One exemplary pharmaceutically acceptable carrier is physiological saline solution. Other physiologically acceptable carriers and their formulations are known to one skilled in the art and described, for example, in Remington: The Science and Practice of Pharmacy, 19 th edition), ed. A.
Gennaro, 1995, Mack Publishing Company, Easton, PA.
"Therapeutically effective amount" as used herein in reference to dosage of a medication, refers to the administration of a specific amount of a pharmacologically active agent a prostate-specific or a testisspecific polypeptide, nucleic acid molecule, or modulatory compound) tailored to each individual patient manifesting symptoms characteristic of a specific disorder. For example, a patient receiving the treatment of the present invention might have prostate cancer. A person skilled in the art will recognize that the optimal dose of a pharmaceutical agent to be administered will vary from one individual to another. Dosage in individual patients should take into account the patients height, weight, rate of absorption and metabolism of the medication in question, the stage of the disorder to be treated, and what other pharmacological agents are administered concurrently.
By "treating or "treatment" is meant the medical management of a patient with the intent to cure, ameliorate, stabilize, or prevent a disease, pathological condition, or disorder. This term includes active treatment, that is, treatment directed specifically toward the improvement or WO 01/72962 PCT/US01/09410 associated with the cure of a disease, pathological condition, or disorder, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, or disorder. In addition, this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, or disorder; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, or disorder; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease, pathological condition, or disorder. The phrase "treatment" also includes symptomatic treatment, that is, treatment directed toward constitutional symptoms of the associated disease, pathological condition, or disorder.
By "disorder of the prostate or testis" is meant a disturbance of function and/or structure of the prostate or testis in a living organism, resulting from an external source, a genetic predisposition, a physical or chemical trauma, or a combination of the above. Such disorders include the proliferation of prostate or testicular cells. By "cell proliferation" is meant the growth or reproduction of similar cells, and the invention provides reagents for inhibiting proliferation and stimulating proliferation. By "inhibiting proliferation" is meant the decrease in the number of similar cells by at least 10%, more preferably by at least 20%, and most preferably by at least 50%. By "stimulating proliferation" is meant an increase in the number of similar cells by at least 10%; more preferably by at least and most preferably by at least The reagents described herein, for example, vectors expressing antisense, antagonists, or inhibitors of prostate-specific or testis-specific polypeptides or nucleic acid molecules may be used, for example, to WO 01/72962 PCT/USU1/09410 suppress the excessive proliferation of prostate or testicular cells. Blocking prostate-specific or testis-specific polypeptide or nucleic acid molecule expression or activity in prostate or testicular cells can alter molecular pathways within cancerous cells and thus trigger apoptosis, the process of cell death where a dying cell displays a set of well-characterized biochemical hallmarks which include cytolemmal blebbing, cell soma shrinkage, chromatin condensation, and DNA laddering.
Disorders of the prostate or testis include prostate cancer, benign prostatic hyperplasia, acute prostatitis, testicular cancer, developmental defects of the prostate or testis (such as cryptorchidism or undescended testis, and retractile, ascending, or vanished testis).
By "proliferative disease" is meant a disease that is caused by or results in inappropriately high levels of cell division, inappropriately low levels of apoptosis, or both. For example, cancers such as prostate cancer, testicular cancer, lymphoma, leukemia, melanoma, ovarian cancer, breast cancer, pancreatic cancer, liver cancer, and lung cancer are all examples of proliferative disease.
By "modulate" or "modulating" is meant changing, either by decrease or increase, the expression or biological activity of a prostatespecific or testis-specific nucleic acid molecule or polypeptide, as described herein. It will be appreciated that the degree of modulation provided by a modulating compound in a given assay will vary, but that one skilled in the art can determine the statistically significant change in the level of biological activity that identifies a compound that modulates a prostatespecific or testis-specific nucleic acid molecule or polypeptide.
The invention provides several advantages. For example, it provides methods and reagents that can be used in the diagnosis and treatment of prostate and testis associated diseases, as well as other disorders and conditions that are sensitive to the bioactivities of the reagents WO 01/72962 PCT/USU1/09410 polypeptides, nucleic acid molecules, antibodies) described herein. Since the prostate-specific or testis-specific polypeptides of the invention have been found to be highly expressed in the prostate and testis, these polypeptides can also be used in screens for therapeutics to treat disorders associated with the prostate and testis. These polypeptides are also expressed in other tissues, and can be used as therapeutics and diagnostics for cell proliferative disorders.
Other features and advantages of the invention will be apparent from the detailed description of the invention, the drawings, and the claims.
Brief Description of The Drawings Figure 1 shows an exemplary reverse northern analysis of several clones from a prostate specific cDNA library.
Figure 2 shows an exemplary multiple tissue northern blot.
Figure 3 is a table showing the nucleotide sequences of twelve clones (SEQ ID NOs: 1-12) isolated from prostate tissue and LNCaP cells.
Figure 4A is a schematic diagram showing the STMP1 gene structure.
Figure 4B shows the nucleotide sequence, including the intron junction sequences (SEQ ID NO: 13), and predicted amino acid sequence (SEQ ID NO: 14) of STMP1.
Figure 4C shows the nucleotide sequences of the exons and 3' UTR of STMP1 (SEQ ID NOs: 15-21).
Figure 4D shows the nucleotide sequence of the ORF of STMP1 (SEQ ID NO: 22).
Figure 4E shows the shows the cDNA sequence (SEQ ID NO: 23), and predicted amino acid sequence (SEQ ID NO:14) of STMP1.
Figure 4F shows the nucleotide sequences of the exons and 3' UTR of STMP1 ORF2 (SEQ ID NOs: 17-20 and 24-26).
WO 01/72962 PCT/USU1/09410 Figure 4G shows the nucleotide sequence of the ORF of STMP1 ORF2 (SEQ ID NO: 27).
Figure 4H shows the cDNA sequence (SEQ ID NO: 28), and predicted amino acid sequence (SEQ ID NO: 29) of STMP1 ORF2.
Figure 41 shows the nucleotide sequences of the exons and 3' UTR of STMP1 ORF3 (SEQ ID NOs: 17-19 and 24-26).
Figure 4J shows the nucleotide sequence of the ORF of STMPI ORF3 (SEQ ID NO: Figure 4K shows the cDNA sequence (SEQ ID NO: 31), and predicted amino acid sequence (SEQ ID NO: 32) of STMP1 ORF3.
Figure 4L shows the cDNA sequence (SEQ ID NO: 33), and predicted amino acid sequence (SEQ ID NO:34) of STMP2.
Figure 4M shows the cDNA sequence (SEQ ID NO: 35), and predicted amino acid sequence (SEQ ID NO: 36) of STMP3.
Figure 5 shows a sequence alignment of STMP1 (SEQ ID NO: 14), with STEAP (SEQ ID NO: 37, Accession No. AF186249), and two ESTs (Accession No. BAA91839 and Accession No. BAB15559; SEQ ID NOs: 38 and 39, respectively).
Figure 6A shows a multiple tissue Northern blot probed with STMP1 or G3PDHcDNA.
Figure 6B is a Northern blot probed with STMP1 and PSA in the androgen-responsive prostate cancer cell line LNCaP and in the CWR22 human prostate cancer xenograft model.
Figure 6C is a Northern blot probed with STMP1 and NKX3A in LNCaP, PC-3, and DU-145 cell lines and in the CWR22R human prostate cancer xenograft model.
Figure 7A shows fluorescence microscopy images of COS-1 cells transiently transfected with GFP-STMP1.
WO 01/72962 PCT/USU1/09410 Figure 7B shows fluorescence microscopy images of COS-1 cells transiently transfected with GFP-STMP 1 and labeled with antibodies against Golgi markers.
Figure 8 shows fluorescence microscopy images of COS-1 cells transiently transfected with GFP-STMP 1 and observed by live-cell confocal microscopy.
Figure 9 shows fluorescence microscopy images of COS-1 cells transiently transfected with GFP-STMP1 and labeled with an antibody against an early endosomal marker.
Figure 10 is a schematic diagram showing the SSH9 gene structure and two mRNA species transcribed from the SSH9 gene.
Figure 11A shows the cDNA (SEQ ID NO: 40) and predicted amino acid sequence (SEQ ID NO: 41) for SSH9.
Figure 11B shows the predicted promoter sequence for SSH9 (SEQ ID NO: 42).
Figure 11C shows the predicted intron-exon boundaries for SSH9 (SEQ ID NOs: 43-50).
Figure 12A is a Northern blot probed with SSH9 in the androgenresponsive prostate cancer cell line LNCaP cells and in the CWR22 human prostate cancer xenograft model.
Figure 12B is a Northern blot probed with SSH9 in LNCaP, PC-3, and DU-145 cell lines, and CWR22R human prostate cancer xenograft model.
Figure 12C is a multiple tissue Northern blot probed with SSH9 or GAPDH cDNA.
Figure 13 is a schematic diagram showing the PSL22 gene structure.
Figure 14A shows the nucleotide sequence of the ORF of PSL22 (SEQ ID NO: 51).
16/01 2007 17:11 FAX 61 3 92438333 GRIFFITH HACK IPAUSTRALIA @009/026 0 23- Figure 14B shows the cDNA sequence (SEQ ID NO 52), and predicted amino acid sequence (SEQ ID NO: 53)ofPSL22.
Figure 14C shows the nucleotide sequences of the TATA promoter and transcription start site, exons, and 5' and 3' UTRs of PSL22 (SEQ ID NOs: 54-70).
s Figure 15 shows a sequence alignment of PSL22 (RhoBP) (SEQ ID NO: 53), with ESTs NP032190 (mRhoph), AF132025 (dRhoph), and BAB23615 (SEQ ID Nos: ON 71-73).
CNi Figure 16A is a Northern blot probed with PSL22 in LNCaP, PC-3, and DU-145 0 cell lines, and in the CWR22R human prostate cancer xenograft model.
C'i 10 Figure 16B is a multiple tissue Northern blot probed with PSL22 cDNA.
Detailed Description of the Invention In the claims which follow and in the description of the invention, except where the context requires otherwise due to express language or necessary implication, the is word "comprise" or variations such as "comprises" or "comprising" is used in an inclusive sense, i.e. to specify the presence of the stated features but not to preclude the presence or addition of further features in various embodiments of the invention.
The basic biology of the normal prostate and testis, as well as prostate and testicular cancer initiation and progression is still poorly understood. It is therefore necessary to delineate the molecular events that are at the basis of these processes. To achieve this goal, we have identified, cloned, and characterized highly prostate-and testis-enriched genes whose gene products have important roles for both the normal physiology and the pathophysiology of the prostate and the testis. These gene products also have important roles in other disorders, for example, heart, brain, liver, pancreas, kidney, and colon, which are the tissues where variable low expression, and occasionally, very high expression of specific gene products, can be detected by Northern analysis, The invention provides prostate-specific or testis-specific polypeptides and nucleic acid molecules (see below), and diagnostic and therapeutic methods employing these polypeptides and nucleic acid N;MeliumF\CiulPmdilld7000.070 MP AU'7I I.AU\SpeaPd7Ib.AU SpcInik.1 2007-1-.I.d ic I lo7 COMS ID No: SBMI-05942216 Received by IP Australia: Time 17:21 Date 2007-01-16 WO 01/72962 PCT/USU1/09410 molecules. The invention also provides methods for identifying compounds that modulate the biological activities of prostate-specific or testis-specific polypeptides and nucleic acid molecules, and therapeutic methods employing these compounds. The diagnostic, therapeutic, and screening methods of the invention are first described, followed by general approaches that can be used in carrying out these methods. Finally, experimental results supporting the methods of the invention are described.
Bioassays Prostate-specific and testis-specific polypeptides are expressed in the prostate and testis, and also in other tissues such as kidney, pancreas, liver, lung, and colon. The expression patterns of prostate-specific and testisspecific polypeptides in specific cells and tissues are used to identify cellular targets of prostate-specific and testis-specific polypeptide actions, and to identify bioactivities that are relevant to specific prostate- and testisrelated diseases, such as prostate cancer, testicular cancer, benign prostatic hyperplasia, acute prostatitis, and developmental testis defects.
Therapeutic and diagnostic utilities for prostate-specific and testisspecific polypeptides are identified by, for example, conducting bioassays in vitro. Culture systems that reflect prostate-specific and testis-specific polypeptide expression patterns, along with the distribution of particular receptors, such as the androgen receptor, are selected. For example, LNCaP cells express androgen receptors, and respond to one or more isoforms of prostate-specific and testis-specific polypeptides in a variety of bioassays. The activities of prostate-specific and testis-specific polypeptides STMP1, SSH9, PSL22) are compared, using sister cultures, in various dose-response assays, including but not limited to, inhibition of proliferation, apoptosis, signaling events changes in kinase activity), changes in transcription factor activity (such as that of the androgen receptor), intracellular trafficking, or cell signaling. The relative WO 01/72962 PCT/US01/09410 potencies of the prostate-specific and testis-specific polypeptides are determined on the basis of, for example, protein concentration.
Diagnostic Methods Employing Prostate-Specific Or Testis-Specific Nucleic Acid Molecules, Polypeptides, and Antibodies Prostate-specific or testis-specific nucleic acid molecules, polypeptides, and antibodies are used in methods to diagnose or monitor a variety of diseases and conditions, including those involving mutations in, or inappropriate expression of, prostate-specific or testis-specific genes.
Prostate-specific or testis-specific expression has been documented in a variety of tissues, as discussed above. Thus, detection of abnormalities in prostate-specific or testis-specific genes or their expression is used in methods to diagnose, or to monitor treatment or development of diseases of these tissues.
The diagnostic methods of the invention are used, for example, with patients that have a prostate-related or testis-related disease, for example, prostate or testicular cancer, in an effort to determine its etiology, and thus, to facilitate selection of an appropriate course of treatment The diagnostic methods are also used with patients that have not yet developed a prostaterelated or testis-related disease, but who may be at risk of developing such a disease, or with patients that are at an early stage of developing such a disease. Many prostate-related or testis-related diseases occur during development, and thus, the diagnostic methods of the invention are also carried out on a fetus or embryo during development. Also, the diagnostic methods of the invention are used in prenatal genetic screening, for example, to identify parents who may be carriers of a recessive prostaterelated or testis-related mutation.
Prostate-specific or testis-specific abnormalities that are detected using the diagnostic methods of the invention include those characterized WO 01/72962 PCT/USU1/09410 by, for example, abnormal prostate-specific or testis-specific polypeptides, (ii) prostate-specific or testis-specific genes containing mutations that result in the production of such polypeptides, and (iii) mutations that result in production of abnormal amounts of prostatespecific or testis-specific polypeptides.
Levels of prostate-specific or testis-specific expression in a patient sample are determined by using any of a number of standard techniques that are well known in the art. For example, prostate-specific or testisspecific expression in a biological sample a blood, prostate or testis tissue sample, or amniotic fluid) from a patient is monitored by standard northern blot analysis or by quantitative PCR (see, Ausubel et al., Current Protocols in Molecular Biology, John Wiley Sons, New York, NY, 1998; PCR Technology: Principles and Applications for DNA Amplification, H.A. Ehrlich, Ed., Stockton Press, NY; Yap et al. Nucl.
Acids. Res. 19:4294, 1991).
A biological sample obtained from a patient can be analyzed for one or more mutations in prostate-specific or testis-specific nucleic acid molecules using a mismatch detection approach. Generally, this approach involves PCR amplification of nucleic acid molecules from a patient sample, followed by identification of a mutation a mismatch) by detection of altered hybridization, aberrant electrophoretic gel migration, binding, or cleavage mediated by mismatch binding proteins, or by direct nucleic acid molecule sequencing. Any of these techniques can be used to facilitate detection of mutant prostate-specific or testis-specific genes, and each is well known in the art. Examples of these techniques are described, for example, by Orita et al. (Proc. Natl. Acad. Sci. USA 86:2766-2770, 1989) and Sheffield et al. (Proc. Natl. Acad. Sci. USA 86:232-236, 1989).
Mismatch detection assays also provide an opportunity to diagnose a prostate-specific or testis-specific gene-mediated predisposition to a disease WO 01/72962 PCT/US01/09410 before the onset of symptoms. For example, a patient heterozygous for a prostate-specific or testis-specific mutation that suppresses normal prostatespecific or testis-specific biological activity or expression may show no clinical symptoms of a prostate-specific or testis-specific gene-related disease, and yet possess a higher than normal probability of developing a prostate or testicular disease. Given such a diagnosis, patients can take precautions to minimize their exposure their exposure to adverse environmental factors and to carefully monitor their medical condition (for example, through frequent physical examinations). As mentioned above, this type of diagnostic approach can also be used to detect prostate-specific or testis-specific mutations in prenatal screens.
The prostate-specific or testis-specific diagnostic assays described above can be carried out using any biological sample (for example, a blood, prostate, or testis tissue sample, or amniotic fluid) in which a prostatespecific or testis-specific polypeptide or nucleic acid molecule is normally expressed. A mutant prostate-specific or testis-specific gene can also be identified using these sources as test samples. Alternatively, a prostatespecific or testis-specific mutation, as part of a diagnosis for predisposition to a prostate-specific or testis-specific gene-associated disease, can be tested for using a DNA sample from any cell, for example, by mismatch detection techniques. Preferably, the DNA sample is subjected to PCR amplification prior to analysis.
In yet another diagnostic approach of the invention, an immunoassay is used to detect or monitor prostate-specific or testis-specific protein expression in a biological sample. Anti-prostate-specific or testis-specificpolypeptide polyclonal or monoclonal antibodies (as described below) can be used in any standard immunoassay format ELISA, Western blot, or RIA; see, Ausubel et al., supra) to measure prostate-specific or testisspecific polypeptide levels. These levels are compared to wild-type WO 01/72962 PCT/USU1/09410 prostate-specific or testis-spcific levels. For example, an increase in prostate-specific or testis-specific polypeptide production may be indicative of a condition or a predisposition to a condition involving overexpression of prostate-specific or testis-specific polypeptide biological activity, such as late stage prostate cancer.
Immunohistochemical techniques can also be utilized for prostatespecific or testis-specific polypeptide detection. For example, a tissue sample can be obtained from a patient, sectioned, and stained for the presence of prostate-specific or testis-specific polypeptide using an antiprostate-specific or testis-specific antibody (see below) and any standard detection system one that includes a secondary antibody conjugated to horseradish peroxidase). General guidance regarding such techniques can be found in, Bancroft et al., Theory and Practice ofHistological Techniques, Churchill Livingstone, 1982, and Ausubel et al., supra.
In a preferred example, a combined diagnostic method can be employed that includes an evaluation of prostate-specific or testis-specific protein production (for example, by immunological techniques or the protein truncation test (Hogerrorst et al., Nature Genetics 10:208-212, 1995), and a nucleic acid molecule-based detection technique designed to identify more subtle prostate-specific or testis-specific mutations (for example, point mutations). As described above, a number of mismatch detection assays are available to those skilled in the art, and any preferred technique can be used. Mutations in prostate-specific or testis-specific genes can be detected that either result in loss or gain of prostate-specific or testis-specific polypeptide or nucleic acid molecule expression or loss or gain of normal prostate-specific or testis-specific polypeptide or nucleic acid molecule biological activity.
Prostate-specific or testis-specific polypeptides or nucleic acid molecules can be used to correlate the course of prostate cancer to a marker WO 01/72962 PCT/USU1/09410 other than PSA, to monitor the course of an anticancer therapy, or to detect a neoplastic cell in a system. For example, a predetermined quantity of an RNA encoding a prostate-specific or testis-specific polypeptide is correlated with the presence of a neoplastic cell, for example, from a biopsy. The total RNA is extracted from the biopsy specimen, and a real time quantitative rt-PCR employing individual reactions with primer pairs specific to prostate-specific or testis-specific sequences is performed in parallel with a biopsy specimen known to be free of cancer cells. Biopsy specimens are determined to have a cancer cell, where the detected prostate-specific or testis-specific mRNA quantity is at least 5 times higher than in the control specimen. An exemplary extraction of total RNA utilizes the Quiagen BioRobot kit in conjunction with the BioRobot 9600 system, and the real time rtPCR is performed in a Perkin Elmer ABI Prism 7700.
In alternative aspects of the inventive subject matter, the method of detecting a neoplastic cell need not be limited to biopsy tissues from prostate or testis tissue, but may employ various alternative tissues, including lymphoma tumor cells, and various solid tumor cells, so long as such tumor cells overproduce mRNA of prostate-specific or testis-specific polypeptides. Appropriate alternative tumor cells can readily be identified by the above described method. Likewise, the system need not be restricted to a mammal, but may also include cell-, and tissue cultures grown in vitro, and tumor cells and specimens from animals other than mammals. For example, tumor cell and tissue grown in vitro may advantageously be utilized to investigate drug action on such cells, and sequences encoding prostate-specific or testis-specific polypeptides may conveniently be employed as tumor marker. Alternatively, body fluids serum, saliva, etc.) that may or may not contain tumor cells are also contemplated a suitable substrate for the method presented herein, so long as they contain WO 01/72962 PCT/USU1/09410 to at least some extent mRNA encoding a prostate-specific or testis-specific polypeptide.
In still other aspects of contemplated methods, the polypeptide quantity need not necessarily be limited to at least 5 times more than the control specimen in order to establish that the tissue has a cancer cell. For example, where the concentration of the polypeptide is hormone dependent, amounts between 3-8 fold and more may be appropriate. In contrast, where the concentration of cancer cells in the biopsy specimen is relatively low, amounts of less than 5-fold, including 1.5 to 4.9-fold and less are contemplated.
The detection process may include fluorescence detection, luminescence detection, scintigraphy, autoradiography, and formation of a dye. For example, for microscopic analysis of biopsy specimens, luciferase labeled probes are particularly advantageous in conjunction with a luminescence substrate luciferin). Luminescence quantification may then be performed utilizing a CCD-camera and image analysis system.
Similarly, radioactivity may be detected via autoradiographic or scintigraphic procedures on a tissue section, in a fluid or on a solid support.
Where the probe is a natural or synthetic ligand of a prostate-specific or testis-specific polypeptide, the ligand may include molecules with a chemical modification that increase the affinity to the polypeptide and/or induce irreversible binding to the polypeptide. For example, transition state analogs or suicide inhibitors for a particular reaction catalyzed by the polypeptide are especially contemplated. Labeling of antibodies, antibody fragments, small molecules, and binding of the labeled entity is a technique that is well known in the art, and all known methods are generally suitable for use in conjunction with methods contemplated herein. Furthermore, the probe need not be limited to a fluorescein labeled antibody, and alternative probes include antibody fragments Fab, Fab', scFab, etc.).
WO 01/72962 PCT/USU1/09410 Still further contemplated variations include substitution of one or more atoms or chemical groups in the sequence with a radioactive atom or group. For example, where cDNAs are employed as a hybridizationspecific probes, a fluorophor or enzyme P-galactosidase for generation of a dye, or luciferase for generation of luminescence) may be coupled to the sequence to identify position and/or quantity of a complementary sequence. Alternatively, where contemplated cDNA molecules are utilized for affiity isolation procedures, the cDNA may be coupled to a molecule that is known to have a high-affinity Kd<10 4 mor 1 partner, such as biotin, or an oligo-histidyl tag. In another example, one or more phosphate groups may be exchanged for a radioactive phosphate group with a 32P or 3 3 P isotope to assist in detection and quantification, where the radiolabeled cDNA is employed as a hybridization probe.
Therapeutic Methods Employing Prostate-Specific Or Testis-Specific Nucleic Acid Molecules, Polypeptides, and Antibodies The invention includes methods of treating or preventing prostatespecific or testis-specific diseases. Therapies are designed to circumvent or overcome a prostate-specific or testis-specific gene defect, or inadequate or excessive prostate-specific or testis-specific gene expression, and thus modulate and possibly alleviate conditions involving defects in prostatespecific or testis-specific genes or proteins. In considering various therapies, it is understood that such therapies are, preferably, targeted to the affected or potentially affected organs, for example, the prostate or the testis. Reagents that are used to modulate prostate-specific or testisspecific biological activity can include, without limitation, full length prostate-specific or testis-specific polypeptides; prostate-specific or testisspecific cDNA, mRNA, or antisense RNA; prostate-specific or testisspecific antibodies; and any compound that modulates prostate-specific or WO 01/72962 PCT/US01/09410 testis-specific polypeptide or nucleic acid molecule biological activity, expression, or stability.
Treatment or prevention of diseases resulting from a mutated prostate-specific or testis-specific gene is accomplished, for example, by replacing a mutant prostate-specific or testis-specific gene with a normal prostate-specific or testis-specific gene, administering a normal prostatespecific or testis-specific gene, modulating the function of a mutant prostate-specific or testis-specific protein, delivering normal prostatespecific or testis-specific protein to the appropriate cells, or altering the levels of normal or mutant. prostate-specific or testis-specific protein. It is also possible to correct a prostate-specific or testis-specific gene defect to modify the physiological pathway an intracellular trafficking pathway) in which the prostate-specific or testis-specific protein participates.
To replace a mutant protein with normal protein, or to add protein to cells that do not express sufficient or normal prostate-specific or testisspecific protein, it may be necessary to obtain large amounts of pure prostate-specific or testis-specific protein from cultured cell systems in which the protein is expressed (see, below). Delivery of the protein to the affected tissue can then be accomplished using appropriate packaging or administrating systems. Alternatively, small molecule analogs that act as prostate-specific or testis-specific molecule agonists or antagonists can be administered to produce a desired physiological effect (see below).
Gene therapy is another therapeutic approach for preventing or ameliorating diseases caused by prostate-specific or testis-specific gene defects. Nucleic acid molecules encoding wild type prostate-specific or testis-specific proteins can be delivered to cells that lack sufficient, normal prostate-specific or testis-specific biological activity cells carrying mutations in prostate-specific or testis-specific genes). The nucleic acid WO 01/72962 PCT/USU1/09410 molecules must be delivered to those cells in a form in which they can be taken up by the cells and so that sufficient levels of protein, to provide effective prostate-specific or testis-specific function, can be produced.
Alternatively, for some prostate-specific or testis-specific mutations, it may be possible slow the progression of the resulting disease or to modulate prostate-specific or testis-specific activity by introducing another copy of a homologous gene bearing a second mutation in that gene, to alter the mutation, or to use another gene to block any negative effect.
Transducing retroviral, adenoviral, and adeno-associated viral vectors can be used for somatic cell gene therapy, especially because of their high efficiency of infection and stable integration and expression (see, Cayouette et al., Human Gene Therapy 8:423-430, 1997; Kido et al., Current Eye Research 15:833-844, 1996; Bloomer et al., Journal of Virology 71:6641-6649, 1997; Naldini et al., Science 272:263-267, 1996; and Miyoshi et al., Proc. Natl. Acad. Sci., USA 94:10319-1032, 1997). For example, the full length prostate-specific or testis-specific gene, or a portion thereof, can be cloned into a retroviral vector and expression can be driven from its endogenous promoter, from the retroviral long terminal repeat, or from a promoter specific for a target cell type of interest (such as aortic or other vascular cells). Other viral vectors that can be used include, for example, vaccinia virus, bovine papilloma virus, or a herpes virus, such as Epstein-Barr Virus (also see, for example, the vectors of Miller, Human Gene Therapy 15-14, 1990; Friedman, Science 244:1275-1281, 1989; Eglitis et al., BioTechniques 6:608-614, 1988; Tolstoshev et al., Current Opinion in Biotechnology 1:55-61, 1990; Sharp, The Lancet 337:1277- 1278, 1991; Cornetta et al., Nucleic Acid Research and Molecular Biology 36:311-322, 1987; Anderson, Science 226:401-409, 1984; Moen, Blood Cells 17:407-416, 1991; or Miller et al., Biotechnology 7:980-990, 1989).
Retroviral vectors are particularly well developed and have been used in WO 01/72962 PCT/USU1/09410 clinical settings (Rosenberg et al., N. Engl. J. Med 323:370, 1990; Anderson et al., U.S. Patent No. 5,399,346).
Gene transfer can also be achieved using non-viral means involving transfection in vitro, by means of any standard technique, including but not limited to, calcium phosphate, DEAE dextran, electroporation, protoplast fusion, and liposomes. Transplantation of normal genes into the affected tissues of a patient can also be accomplished by transferring a normal prostate-specific or testis-specific gene into a cultivatable cell type ex vivo, after which the cell (or its descendants) is injected into a targeted tissue.
Another strategy for inhibiting prostate-specific or testis-specific function using gene therapy involves intracellular expression of an anti-prostatespecific or testis-specific antibody or a portion of an prostate-specific or testis-specific antibody. For example, the gene (or gene fragment) encoding a monoclonal antibody that specifically binds to prostate-specific or testis-specific polypeptide and inhibits its biological activity is placed under the transcriptional control of a tissue-specific gene regulatory sequence. Another therapeutic approach involves administration of recombinant prostate-specific or testis-specific polypeptide, either directly to the site of a potential or actual disease-affected tissue (for example, by injection) or systemically (for example, by any conventional recombinant protein administration technique). The dosage of a prostate-specific or testis-specific polypeptide depends on a number of factors, including the size and health of the individual patient but, generally, between about 0.006 mg/kg to about 0.6 mg/kg, inclusive, is administered per day to an adult in any pharmaceutically acceptable formulation.
Non-viral approaches can also be employed for the introduction of therapeutic DNA into cells predicted to be subject to diseases involving a prostate-specific or testis-specific disorder. For example, a prostatespecific or testis-specific nucleic acid molecule or an antisense nucleic acid WO 01/72962 PCT/USU1/09410 molecule can be introduced into a cell by lipofection (Felgner et al., Proc.
Natl. Acad. Sci. USA 84:7413, 1987; Ono et al., Neuroscience Letters 17:259, 1990; Brigham et al., Am. J. Med. Sci. 298:278, 1989; Staubinger et al., Methods in Enzymology 101:512, 1983), asialoorosomucoid-polylysine conjugation (Wu et al., Journal ofBiological Chemistry 263:14621, 1988; Wu et al., Journal of Biological Chemistry 264:16985, 1989), or, less preferably, micro-injection under surgical conditions (Wolff et al., Science 247:1465, 1990).
Prostate-specific or testis-specific cDNA expression for use in gene therapy methods can be directed from any suitable promoter the human cytomegalovirus (CMV), simian virus 40 (SV40), or metallothionein promoters), and regulated by any appropriate mammalian regulatory element. For example, if desired, enhancers known to preferentially direct gene expression in specific cell types can be used to direct prostate-specific or testis-specific expression. The enhancers used can include, without limitation, those that are characterized as tissue- or cell-specific enhancers. Alternatively, if a prostate-specific or testisspecific genomic clone is used as a therapeutic construct (such clones can be identified by hybridization with prostate-specific or testis-specific cDNA, described above), regulation can be mediated by the cognate regulatory sequences, or, if desired, by regulatory sequences derived from a heterologous source, including any of the promoters or regulatory elements described above.
Antisense-based strategies can be employed to explore prostatespecific or testis-specific gene function and as a basis for therapeutic drug design. These strategies are based on the principle that sequence-specific suppression of gene expression (via transcription or translation) can be achieved by intracellular hybridization between genomic DNA or mRNA and a complementary antisense species. The formation of a hybrid RNA WO 01/72962 PCT/USU1/09410 duplex interferes with transcription of the target prostate-specific or testisspecific-encoding genomic DNA molecule, or processing, transport, translation, or stability of the target prostate-specific or testis-specific mRNA molecule.
Antisense strategies can be delivered by a variety of approaches.
For example, antisense oligonucleotides or antisense RNA can be directly administered by intravenous injection) to a subject in a form that allows uptake into cells. Alternatively, viral or plasmid vectors that encode antisense RNA (or antisense RNA fragments) can be introduced into a cell in vivo or ex vivo. Antisense effects can be induced by control (sense) sequences; however, the extent of phenotypic changes are highly variable.
Phenotypic effects induced by antisense effects are based on changes in criteria such as protein levels, protein activity measurement, and target mRNA levels.
For example, prostate-specific or testis-specific gene therapy can also be accomplished by direct administration of antisense prostate-specific or testis-specific mRNA to a cell that is expected to be adversely affected by the expression of wild-type or mutant prostate-specific or testis-specific polypeptides. The antisense prostate-specific or testis-specific mRNA can be produced and isolated by any standard technique, but is most readily produced by in vitro transcription using an antisense prostate-specific or testis-specific cDNA under the control of a high efficiency promoter the T7 promoter). Administration of antisense prostate-specific or testisspecific mRNA to cells can be carried out by any of the methods for direct nucleic acid molecule administration described above.
An alternative strategy for inhibiting prostate-specific or testisspecific function using gene therapy involves intracellular expression of an anti-prostate-specific or testis-specific antibody or a portion of an anti- WO 01/72962 PCT/US01/09410 prostate-specific or testis-specific antibody. For example, the gene (or gene fragment) encoding a monoclonal antibody that specifically binds to prostate-specific or testis-specific and inhibits its biological activity can be placed under the transcriptional control of a tissue-specific gene regulatory sequence.
Another therapeutic approach within the invention involves administration of recombinant prostate-specific or testis-specific polypeptide, either directly to the site of a potential or actual diseaseaffected tissue (for example, by injection) or systemically (for example, by any conventional recombinant protein administration technique). The dosage of prostate-specific or testis-specific depends on a number of factors, including the size and health of the individual patient, but, generally, between 0.1 mg and 100 mg, inclusive, are administered per day to an adult in any pharmaceutically acceptable formulation.
In a patient diagnosed as having a prostate-specific or testis-specific mutation gene or a prostate-specific or testis-specific disease, or as susceptible to prostate-specific or testis-specific gene mutations, aberrant prostate-specific or testis-specific polypeptide or nucleic acid molecule expression (even if those mutations or expression patterns do not yet result in alterations in prostate-specific or testis-specific expression or biological activity), or to a prostate-specific or testis-specific disease, any of the above-described therapies are administered before the occurrence of the disease phenotype. Also, compounds shown to modulate prostate-specific or testis-specific polypeptide or nucleic acid molecule expression or prostate-specific or testis-specific polypeptide or nucleic acid molecule biological activity are administered to patients diagnosed with potential or actual diseases by any standard dosage and route of administration.
Alternatively, gene therapy using an antisense prostate-specific or testis- WO 01/72962 PCT/US01/09410 specific mRNA expression construct is undertaken to reverse or prevent the gene defect prior to the development of the full course of the disease.
The therapeutic methods of the invention are, in some cases, targeted to prenatal treatment. For example, a fetus found to have a prostatespecific or testis-specific mutation is administered a gene therapy vector including a normal prostate-specific or testis-specific gene, or administered a normal prostate-specific or testis-specific protein. Such treatment may be required only for a short period of time, or may, in some form, be required throughout such a patient's lifetime. Any continued need for treatment, however, is determined using, for example, the diagnostic methods described above. Also as discussed above, prostate-specific or testisspecific polypeptide or nucleic acid molecule abnormalities may be associated with diseases in adults, and thus, adults are subject to the therapeutic methods of the invention as well.
Additionally, prostate-specific or testis-specific polypeptides may be used to stimulate an immune system to assist in generating immunity against, for example, prostate cancer cells.
The methods of the present invention can be used to diagnose or treat the disorders described herein in any mammal, for example, humans, domestic pets, or livestock. Where a non-human mammal is treated or diagnosed, the prostate-specific or testis-specific polypeptide, nucleic acid molecule, or antibody employed is preferably specific for that species.
WO 01/72962 PCT/US01/09410 Identification of Molecules that Modulate Prostate-Specific Or Testis- Specific Polypeptide or Nucleic Acid Molecule Biological Activity or Whose Biological Activity is Modulated by Prostate-Specific Or Testis- Specific Polypeptides or Nucleic Acid Molecules Isolation of prostate-specific or testis-specific cDNAs (as described herein) also facilitates the identification of molecules that increase or decrease prostate-specific or testis-specific polypeptide or nucleic acid molecule biological activity. Similarly, molecules whose activity is modulated by prostate-specific or testis-specific polypeptide or nucleic acid molecule biological activity can be identified. According to one approach, candidate molecules are added at varying concentrations to the culture medium of cells expressing prostate-specific or testis-specific mRNA.
Prostate-specific or testis-specific biological activity is then measured using standard techniques. The measurement of biological activity can include, without limitation, the measurement of prostate-specific or testis-specific protein and nucleic acid molecule expression levels, response to androgens, or intracellular localization and trafficking.
If desired, the effect of candidate modulators on expression can also be measured at the level of prostate-specific or testis-specific protein production using the same general approach and standard immunological detection techniques, such as western blotting or immunoprecipitation with a prostate-specific or testis-specific-specific antibody (see below).
A test compound that is screened in the methods described above can be a chemical, be it naturally-occurring or artificially-derived. Such compounds can include, for example, polypeptides, synthesized organic molecules, naturally occurring organic molecules, nucleic acid molecules, and components thereof. Candidate prostate-specific or testis-specific modulators include peptide as well as non-peptide molecules peptide WO 01/72962 PCT/USU1/09410 or non-peptide molecules found, in a cell extract, manmmalian serum, or growth medium in which mammalian cells have been cultured).
Administration of Prostate-Specific Or Testis-Specific Polypeptides, Prostate-Specific Or Testis-Specific Nucleic Acid Molecules, and Modulators of Prostate-Specific Or Testis-Specific Polypeptide or Nucleic Acid Molecule Synthesis or Function A prostate-specific or testis-specific protein, nucleic acid molecule, or modulator is administered within a pharmaceutically-acceptable diluent, carrier, or excipient, in unit dosage form to patients or experimental animals. Also, conventional pharmaceutical practice is employed to provide suitable formulations or compositions in which to administer neutralizing prostate-specific or testis-specific antibodies or prostatespecific or testis-specific-inhibiting compounds a prostate-specific or testis-specific antisense molecule or a prostate-specific or testis-specific dominant negative mutant) to patients suffering from a prostate-specific or testis-specific disease, such as prostate cancer, testicular cancer, benign hyperplasia of the prostate, or developmental defects of the prostate or testis. Administration can begin before or after the patient is symptomatic.
Any appropriate route of administration can be employed, for example, administration can be parenteral, intravenous, intra-arterial, subcutaneous, intramuscular, intracranial, intraorbital, ophthalmic, intraventricular, intracapsular, intraspinal, intracisternal, intraperitoneal, intranasal, inhalation to deep lung, aerosol, by suppositories, oral, or topical by applying an adhesive patch carrying a formulation capable of crossing the dermis and entering the bloodstream). Preferably, the administration is local to the afflicted tissue, such as prostate or testis tissue. Therapeutic formulations can be in the form of liquid solutions or suspensions; for oral administration, formulations can be in the form of WO 01/72962 PCT/USU1/09410 tablets or capsules; and for intranasal formulations, in the form of powders, nasal drops, or aerosols. Any of the above formulations may be a sustained-release formulation.
Methods that are well known in the art for making formulations are found, for example, in Remington's Pharmaceutical Sciences, (1 8 th edition), ed. A. Gennaro, 1990, Mack Publishing Company, Easton, PA.
Formulations for parenteral administration can, for example, contain excipients; sterile water; or saline; polyalkylene glycols, such as polyethylene glycol; oils of vegetable origin; or hydrogenated napthalenes.
Sustained-release, biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers can be used to control the release of the compounds. Other potentially useful parenteral delivery systems for prostate-specific or testisspecific modulatory compounds include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes.
Formulations for inhalation can contain excipients, for example, lactose, or can be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate, and deoxycholate, or can be oily solutions for administration in the form of nasal drops, or as a gel.
Prostate-Specific Or Testis-Specific Fragments Polypeptide fragments that include various portions of prostatespecific or testis-specific proteins are useful in identifying the domains important for their biological activities, such as protein-protein interactions and transcription. Methods for generating such fragments are well known in the art (see, for example, Ausubel et al., supra), using the nucleotide sequences provided herein. For example, a prostate-specific or testisspecific protein fragment can be generated by PCR amplifying a desired prostate-specific or testis-specific nucleic acid molecule fragment using WO 01/72962 PCT/USU1/09410 oligonucleotide primers designed based upon the prostate-specific or testisspecific nucleic acid sequences. Preferably, the oligonucleotide primers include unique restriction enzyme sites that facilitate insertion of the amplified fragment into the cloning site of an expression vector a mammalian expression vecoor, see above). This vector can then be introduced into a cell a mammalian cell; see above) by artifice, using any of the various techniques known in the art such as those described herein, resulting in the production of a prostate-specific or testis-specific polypeptide fragment in the cell containing the expression vector.
Prostate-specific or testis-specific polypeptide fragments chimeric fusion proteins) can also be used to raise antibodies specific for various regions of prostate-specific or testis-specific polypeptides.
Preferred prostate-specific or testis-specific fragments include, without limitation, fragments including the N-terminal domain of STMP 1 (amino acids 1-200), the P5CR domain, and fragments thereof.
Synthesis of Prostate-Specific Or Testis-Specific Proteins. Polypeptides, and Polyp eptide Fragments Those skilled in the art of molecular biology will understand that a wide variety of expression systems can be used to produce recombinant prostate-specific or testis-specific proteins. The precise host cell used is not critical to the invention. The prostate-specific or testis-specific proteins can be produced in a prokaryotic host E. coli) or in a eukaryotic host S. cerevisiae, insect cells such as Sf9 cells, or mammalian cells such as COS, NIH 3T3, CHO, or HeLa cells). These cells are commercially available from, for example, the American Type Culture Collection, Rockville, MD (see also Ausubel et al., Current Protocols in Molecular Biology, John Wiley Sons, New York, NY, 1998). The method of transformation and the choice of expression vehicle expression WO 01/72962 PCT/USU1/09410 vector) will depend on the host system selected. Transformation and transfection methods are described, in Ausubel et al., Current Protocols in Molecular Biology, John Wiley Sons, New York, NY, 1998, and expression vehicles can be chosen from those provided, e.g. in Pouwels et al., Cloning Vectors: A Laboratory Manual, 1985, Supp. 1987).
The characteristics of prostate-specific or testis-specific nucleic acid molecules are analyzed by introducing such genes into various cell types or using in vitro extracellular systems. The function ofprostate-specific or testis-specific proteins produced in such cells or systems are then examined under different physiological conditions. Also, cell lines can be produced that over-express the prostate-specific or testis-specific gene product, allowing purification of prostate-specific or testis-specific proteins for biochemical characterization, large-scale production, antibody production, and patient therapy.
The polypeptides of the invention may be produced in vivo or in vitro, and may be chemically and/or enzymatically modified. The polypeptides can be isolated from prostate tissue or prostate cancer cells that may or may not be in a hormone dependent state. Alternatively, and especially where larger amounts >10mg) are desirable, recombinant production in a bacterial, yeast, insect cell, or mammalian cell system) may advantageously be employed to generate significant quantities of prostate-specific or testis-specific polypeptides.
Furthermore, recombinant production not only offers a more economical strategy to produce the polypeptides of the invention, but also allows specific modification in the amino acid sequence and composition to tailor particular biochemical, catalytic and physical properties. For example, where increased solubility of is desirable, one or more hydrophobic amino acids may be replaced with hydrophilic amino acids.
WO 01/72962 PCT/USU1/09410 Alternatively, where reduced or increased catalytic activity is required, one or more amino acids may be replaced or eliminated.
In still another example, the polypeptides of the invention can be synthesized as fusion proteins including, for example, fusions with enzymatically active partners for dye formation or substrate conversion) and fluorescent partners such as GFP, EGFB, BFP, etc.
With respect to chemical and enzymatic modifications of contemplated polypeptides, it is many modifications are appropriate, including addition of mono-, and bifunctional linkers, coupling with protein- and non-protein macromolecules, and glycosylation. For example, mono- and bifunctional linkers are especially advantageous where polypeptides are immobilized to a solid support, or covalently coupled to a molecule that enhances immunogenicity of contemplated polypeptides KLH, or BSA conjugation). Alternatively, the polypeptides may be coupled to antibodies or antibody fragments to allow rapid retrieval of the polypeptide from a mixture of molecules. Further couplings include covalent and non-covalent coupling of polypeptides with molecules that prolong the serum half-life and/or reduce immunogenicity such as cyclodextranes and polyethylene glycols.
Use of Prostate-Specific Or Testis-Specific Antibodies Antibodies to prostate-specific or testis-specific proteins are used to detect prostate-specific or testis-specific proteins or to inhibit the biological activities of prostate-specific or testis-specific proteins. For example, a nucleic acid molecule encoding an antibody or portion of an antibody can be expressed within a cell to inhibit prostate-specific or testis-specific function. In addition, the antibodies can be coupled to compounds, such as radionuclides and liposomes for diagnostic or therapeutic uses. Antibodies that inhibit the activity of a prostate-specific or testis-specific polypeptide WO 01/72962 PCT/US01/09410 can also be useful in preventing or slowing the development of a disease caused by inappropriate expression of a wild type or mutant prostatespecific or testis-specific gene. For example, the antibodies of the invention may be utilized to localize and locally quantify disease-specific markers in prostate or testis tissue sections, e.g, in prostate or testicular cancer.
Detection Of Prostate-Specific Or Testis-Specific Gene Expression As noted, the antibodies described above can be used to monitor prostate-specific or testis-specific protein expression. In situ hybridization of RNA can be used to detect the expression of prostate-specific or testisspecific genes. RNA in situ hybridization techniques rely upon the hybridization of a specifically labeled nucleic acid probe to the cellular RNA in individual cells or tissues. Therefore, RNA in situ hybridization is a powerful approach for studying tissue- and temporal-specific gene expression. In this method, oligonucleotides, cloned DNA fragments, or antisense RNA transcripts of cloned DNA fragments corresponding to unique portions of prostate-specific or testis-specific genes are used to detect specific mRNA species, in the tissues of animals, such as mice, at various developmental stages, or to monitor tumnor progression. Other gene expression detection techniques are known to those of skill in the art and can be employed for detection of prostate-specific or testis-specific gene expression.
Identification of Additional Prostate-Specific Or Testis-Specific Genes Standard techniques, such as the polymerase chain reaction (PCR) and DNA hybridization, as well as the SSH and other techniques described herein, can be used to clone prostate-specific or testis-specific homologues in other species and other prostate-specific or testis-specific genes in WO 01/72962 PCT/USU1/09410 humans. Prostate-specific or testis-specific genes and homologues can be readily identified using low-stringency DNA hybridization or lowstringency PCR with human prostate-specific or testis-specific probes or primers. Degenerate primers encoding human prostate-specific or testisspecific or human prostate-specific or testis-specific amino acid sequences can be used to clone additional prostate-specific or testis-specific genes and homologues by RT-PCR.
Additional prostate-specific or testis-specific genes include genes expressed during various growth and developmental phases of the diseased prostate or tcstis, those involved in prostate cancer, benign prostatic hyperplasia, or testicular cancer, and genes expressed as a result of a drug regimen.
Construction of Transgenic Animals and Knockout Animals Characterization of prostate-specific or testis-specific genes provides information that allows prostate-specific or testis-specific knockout animal models to be developed by homologous recombination. Preferably, a prostate-specific or testis-specific knockout animal is a mammal, most preferably a mouse. Similarly, animal models of prostate-specific or testisspecific overproduction can be generated by integrating one or more prostate-specific or testis-specific sequences into the genome of an animal, according to standard transgenic techniques. Moreover, the effect of prostate-specific or testis-specific gene mutations dominant gene mutations) can be studied using transgenic mice carrying mutated prostatespecific or testis-specific transgenes or by introducing such mutations into the endogenous prostate-specific or testis-specific gene, using standard homologous recombination techniques.
A replacement-type targeting vector, which can be used to create a knockout model, can be constructed using an isogenic genomic clone, for WO 01/72962 PCT/USU1/09410 example, from a mouse strain such as 129/Sv (Stratagene Inc., LaJolla, CA). The targeting vector can be introduced into a suitably-derived line of embryonic stem (ES) cells by electroporation to generate ES cell lines that carry a profoundly truncated form of a prostate-specific or testis-specific gene. To generate chimeric founder mice, the targeted cell lines are injected into a mouse blastula-stage embryo. Heterozygous offspring can be interbred to homozygosity. Prostate-specific or testis-specific knockout mice provide a tool for studying the role of prostate-specific or testisspecific polypeptides and nucleic acid molecules in embryonic development and in disease. Moreover, such mice provide the means, in vivo, for testing therapeutic compounds for amelioration of diseases or conditions involving a prostate-specific or testis-specific polypeptide or nucleic acid molecule-dependent or prostate-specific or testis-specific polypeptide or nucleic acid molecule-affected pathway.
Animal Models The prostate-specific and testis-specific polypeptides, antisense compounds, etc., of the invention can also be used in conjunction with animal models of prostate or testis disorders, to test the therapeutic, diagnostic, and screening methods of the invention. An exemplary prostate cancer model in transgenic mice is called TRAMP, in which the SV40 large T antigen is targeted to the prostate (Greenberg et al., PNAS 92, 3439- 3443, 1995). Another test system is the CWR22 (androgen-dependent) and CWR22R (androgen-independent) xenografts, as known in the art and as described herein. Growth, PSA secretion, metastasis, etc. of these xenografts could be monitored in the presence and absence of the prostatespecific or testis-specific polypeptides, nucleic acid molecules, and other compounds of the invention. Other animal models, for example, animal WO 01/72962 PCT/USU1/09410 models of other forms of cancer, or immunocompromised animals, e.g., nude mice, may also be used.
The following Examples will assist those skilled in the art to better understand the invention and its principles and advantages. It is intended that these Examples be illustrative of the invention and not limit the scope thereof.
EXAMPLE 1 Suppression Subtraction Of Prostate- And Testes-Specific Genes And Subcloning Into Pzero cDNA derived from poly(A)+ RNA of 10 different normal human tissues were subtracted against normal human prostate cDNA using suppression subtraction hybridization (SSH) (Diatchenko, L. et al., Proc.
Natl. Acad. Sci. USA 93, 6025-6030, 1996) and the resulting cDNA fragments were cloned into an appropriate vector. SSH was performed as described (Clontech PCR-Select Cloning Kit) using prostate poly(A)+ RNA against a pool of poly(A)+ RNA obtained from ten normal human tissues (heart, brain, placenta, lung, liver, skeletal muscle, kidney, spleen, thymus, and ovary). Upon secondary PCR amplification (12 cycles), the reactions were extracted with phenol/chloroform, the DNA with ethanol, and the pellets washed once with 70% ethanol. After drying, the DNA pellet was dissolved in 0.2XTE or MQ dH 2 0 and cut with Rsal in a 20 tl reaction for 2 hrs at 37 0 C to excise adaptors. After digestion, the reactions were run on a 1.5% agarose gel, with molecular size markers on one side, at 5 V/cm, min. Care was taken not to expose the gel to short wavelength UV light.
The adapter bands were excised, and the gel was run at 5 V/cm for 15 min in a reversed electric field to concentrate the cDNA bands.
WO 01/72962 PCT/USU1/09410 The gel was visualized (long wave UV light) and the amplified cDNAs, ranging in size between 100 bp-lkB, were excised. The DNA was purified using the QAIEX gel DNA purification kit. The purified DNA was cloned into EcoRV-cut, dephosphorylated pZERO (Invitrogen).
Ligation reactions were performed in 10 gl final volume in the presence of PEG, 1X T4 Ligase buffer at 37 0 C overnight and a 1/5 dilution of 11 of the ligation mix (PSL) was transformed into DH10B electrocompetent cells (>1010 efficiency) or equivalent. Colonies were picked and the presence of cDNA inserts was confirmed. To that end, PCR was performed with T7 and SP6 primers directly from the colonies. 10% of the reactions were run on a 1.5% agarose gel to visualize amplified products. The colonies with inserts were grown and glycerol stocks were prepared and stored at -80 0
C.
EXAMPLE 2 Reverse Northern Blot And Sequence Analyses To clone androgen-responsive genes represented in the PSL, the reverse northern technique was used (Hedrick, S.M. et al., Nature 308, 149- 153, 1984; Sakaguchi, N. et al., EMBO J5: 2139-2147, 1986). In this procedure, RNA made from two populations of cells that are to be compared is used to make cDNA probes that are then hybridized to two identical arrays of clones. To that end, PSL clones were amplified by PCR and spotted on nylon filters in 96-well format to generate two identical blots for each set of 92 clones (the remaining four spots were used for positive and negative controls). To make the probes, the androgenresponsive prostate cancer cell line LNCaP was used (Horoszewicz, J.S. et al., Cancer Res. 43, 1809-1818, 1983) and was either left untreated (the probe) or treated with the synthetic androgen R1881 for 24 hours (the probe). Poly(A)+ RNA was isolated from these cells and was used to make WO 01/72962 PCT/USU1/09410 the 3 2 P-labeled probes. After hybridization with the and probes, clones that showed differential hybridization were selected for further analysis, confirmation by a secondary reverse northern blot, and northern blotting.
Reverse northern screening on the cDNA clones was done essentially as described previously (Hedrick, S.M. et al., supra; Sakaguchi, N. et al., supra) with some modifications. DNA (approximately 400 ng) from PCR amplification in step 6 was diluted in 200 pl of 0.4M NaOH, 10 mM EDTA and mixed well by pipetting. After incubation at 95 0 C for 5-10 minutes, the tubes were chilled on ice. Denatured DNA was blotted on two separate pieces of Zeta Probe GT+ membrane (Bio-Rad) using a dot-blot apparatus (Bio-Rad). Positive (Prostate specific antigen (PSA) cDNA) and negative (glyceraldehyde 3-phosphate dehydrogenase (G3PDH) cDNA) controls were included on each blot (bottom right) in duplicate. The membranes were rinsed with 2XSSC, air dried, and then baked at 80 0 C for 30 minutes.
An exemplary reverse northern analysis is shown in Figure 1. Note that there was a substantial increase in PSA hybridization in the blot (probe prepared from cells that have been stimulated by androgens) compared with the blot (probe prepared from unstimulated cells), whereas there was no significant change in hybridization of G3PDH between the two blots.
Arrowheads indicate the positive clones identified in this experiment.
To verify the tissue-specific nature of the isolated sequences, positive clones were tested in a standard northern blot against RNA preparations of multiple non-prostate tissue samples. Figure 2 shows a multiple tissue northern blot using NKX3A as a probe, to show an exemplary tissue expression pattern seen in the positive clones. Lanes 1and 12-16 are RNA preparations from non-prostate tissues, lane 11 is a RNA preparation from prostate, lane 12 is a RNA preparation from testis.
WO 01/72962 PCT/USU1/09410 Twelve clones with no significant homology to known sequences (by BLAST analysis) were isolated from prostate tissue and LNCaP cells. SEQ ID NOs: 1-9 were identified as androgen-responsive differentiallyexpressed genes in the prostate, while SEQ ID NOs: 10-12 were identified as androgen-responsive differentially-expressed genes in LNCaP cells.
EXAMPLE 3 Isolation And Characterization Of The STMP1 Gene And mRNA A normal prostate cDNA library was screened by and 3'-RACE analysis, and resulted in the full-length cDNA for L74. Since computeraided secondary structure prediction of the deduced amino acid sequence of L74 suggested the presence of a six-transmembrane domain in its Cterminal half, L74 was renamed Six-Transmembrane Protein of Prostate 1 (STMP1).
When the full-length STMP1 cDNA was used in BLAST analysis, it was found to match a BAC clone (GenBank accession AC002064) except for a 313 bp repetitive unit in the 3' UTR region, thereby identifying it as the STMP1 gene and localizing it to Chr7q21. The repetitive region is likely to be a cloning or sequencing artifact of the BAC clone. Computational exon/intron junction analysis and alignment of the full-length cDNA sequence with the BAC clone revealed that STMP1 gene is composed of six exons and five introns (Figure 4A). The transcription start site, the location and size of the exons and introns, and the location of the partial cDNA clone L74 (black box) are indicated. The start (atg) and stop codons (tga), as well as the putative polyadenylation signal (pA) are also indicated. The first two exons are short, non-coding exons of 83 and 61 bp, whereas exons 3-6 encode the open reading frame (ORF) and are 525, 528, 165, and 3281 bp long, respectively (Figure 4C). The STMP1 gene spans around 26 kb, which is in part due to the extremely large size of intron 2 (12713 bp).
WO 01/72962 PCT/USU1/09410 There are three different predicted promoters within 4 kb upstream of the STMP1 initiation codon, none of which has any significant TATA or CAAT box consensus sequences, suggesting that STMP1 is transcribed from a TATA-less promoter.
The STMP1 cDNA (GenBank accession AY008445) has a predicted untranslated region (5'UTR) of approximately 1 kb (deduced by RACE analysis) and an unusually long 3'UTR of approximately 4 kb that comprises -77% of the total cDNA sequence. The ORF starts within the 3 rd exon and is predicted to encode a 490 amino-acid protein (Figure 4B).
A search for protein motifs identified six predicted transmembranc domains in the C-terminal half of STMP1 starting at F209 (Figures 4B and 4E).
Only the cDNA sequence surrounding the ORF is indicated. The exonintron junctions are indicated and the location of the predicted transmembrane domains are highlighted (TM 1-6) (Figure 4B). The stop codon is indicated with an asterisk. STMP 1 has two alternatively spliced forms, shown in Figures 4F-4K, which lead to two predicted isoforms of the protein.
EXAMPLE 4 STMP1 Belongs To A New Subfamily Of Six-Transmembrane Domain Proteins BLAST analysis of GenBank with the predicted STMP1 amino acid sequence identified two independent ESTs and STEAP, a recently discovered cell membrane protein enriched in prostate for expression. An alignment of these sequences, obtained by Clustal and GenDoc programs, is shown in Figure 5. Completely conserved residues are shaded in black; residues that are conserved in two or three of the sequences are shaded light and dark gray, respectively. This alignment suggested that while the EST WO 01/72962 PCT/USU1/09410 BAA91839 cDNA may be close to full-length, BAB15559 cDNA may represent a partial sequence.
The sequences of two proteins related to STMP 1 were determined (Figures 4L and 4M, STMP2 and STMP3, respectively). The STMP2 and STMP3 sequences contain the EST sequences. The GFP-fusion of STMP2 gives similar localization as STMP1. Both STMP2 and STMP3 are more widely distributed and have higher levels in some tissues other than the prostate. For example, STMP2 has the highest expression in the placenta and the lung, and is also highly expressed in the heart, liver, prostate, and testis, while STMP3 has the highest expression in the liver, and is also highly expressed in the heart, placenta, lung, kidney, pancreas, prostate, testis, small intestine, and colon.
The sequence similarity between STMP1 and STEAP is limited and not significant before residue 210 of STMP1 where the predicted sixtransmembrane coding domain starts. This suggests that the N-terminal region is structurally and functionally related among STMP proteins, forming a six-transmembrane protein subfamily that is distinct from
STEAP.
EXAMPLE STMP1 Expression Is Highly Enriched In Prostate The expression profile of STMP1 was then determined in various human tissues by Northern analysis, in which a multiple tissue Northern blot was hybridized to the STMP1 probe (see Materials and Methods). As shown in Figure 6A, STMP1 hybridized to a major mRNA species of kb, and three minor mRNA species of 2.2, 4.0, and 4.5 kb in the prostate tissue. The stronger hybridization that is observed with G3PDH in the heart and skeletal muscle samples is due to its higher expression in these tissues. The lanes represent: 1.Heart, 2. Brain, 3. Placenta, 4. Lung, WO 01/72962 PCT/USU1/09410 Liver, 6. Skeletal Muscle, 7. Kidney, 8. Pancreas, 9. Spleen, 10. Thymus, 11. Prostate, 12. Testis, 13. Ovary, 14. Small Intestine, 15. Colon, 16.
Peripheral blood leukocyte. The location of the full-length 6.5 kb mRNA, as well as the lower molecular weight STMP1 species are indicated by arrows to the left of the figure. There was 15-20-fold lower mRNA expression of the 6.5 kb band in the heart, brain, kidney, pancreas, and ovary, compared to prostate, and no detectable expression in other tissues.
In-contrast, the three lower molecular weight species, encoded by alternatively spliced forms of STMP1, were only detectable in the prostate.
Hybridization with a glyceraldehyde 3-phosphate dehydrogenase (G3PDH) cDNA probe resulted in approximately similar signals in all lanes, except for the heart and skeletal muscle where G3PDH is known to be more abundant compared with other tissues. These data show that STMP1 expression is high in the prostate, although expression can be seen in other tissues, and that STMP1 has isoforms that are restricted to the prostate for expression.
EXAMPLE 6 Characterization Of STMP1 Expression Since androgen is a major hormonal stimulus for the normal prostate gland and for early stage prostate cancer, the possible androgen regulation of STMP1 was assessed by Northern analysis in the androgen-responsive prostate cancer cell line LNCaP. Cells were either left untreated or treated with the synthetic androgen R1881 (10- 8 M) with increasing amounts of time (hours) as indicated (Figure 6B), harvested, and total RNA isolated and used in Northern analysis with STMP1 cDNA as probe. The same membrane was also probed for the androgen-dependent gene PSA.
Relative induction of mRNA accumulation is indicated at the bottom of the lanes, as determined by phosphorimager analysis (Molecular Dynamics).
WO 01/72962 PCT/USU1/09410 The CWR22 xenograft was grown in nude mice and tumor samples were collected either before or 1, 2, or 4 weeks after castration. Total RNA was isolated and was then used in Northern analysis with the same probes.
Ethidium bromide-stained 18S RNA is shown as a control for RNA integrity and loading. At 6 h, there was an approximately 25% increase in STMP1 expression, which was lost by 24 h, with a final 20% decrease observed at 48 h compared with basal levels. In contrast, the mRNA accumulation of the androgen-regulated gene PSA dramatically increased upon androgen stimulation in a time-dependent manner, as expected, reaching approximately 22-fold higher lcvcls by 48 hours. Relative induction of STMPJ mRNA accumulation is indicated at the bottom of the lanes determined by phosphorimager analysis. As is shown in Figure 6B, STMP1 displayed similar expression levels in untreated and R1881-treated LNCaP cells, indicating that STMP1 expression is not significantly regulated by androgens in LNCaP cells.
To determine the possible androgenic regulation of STMP1 expression in an in vivo setting, the androgen-dependent xenograft model CWR22, which is derived from a primary human prostate tumor was used (Wainstein, M. A. et al., Cancer Res 54, 6049-6052, 1994). Since they are androgen-dependent for growth, the CWR22 tumors in nude mice display marked regression upon castration and may regress completely. CWR22 xenografts were grown in nude mice in the presence of a sustained release testosterone pellet. After the tumors had grown, the mice were castrated, the testosterone pellets were removed, and the regressing tumors were collected at 1, 2, or 4 weeks post-castration. Total RNA was prepared from these tumor samples and used in Northern analysis. As shown in Figure 6B, similar to the obsevations in LNCaP cells, STMPI mRNA accumulation in the CWR22 tumors showed no significant change upon castration and was not affected by the presence of androgens (note that WO 01/72962 PCT/USU1/09410 there is underloading of RNA for CWR22 2wk sample). In contrast, the mRNA accumulation of the androgen-regulated gene PSA was dramatically decreased upon castration, dropping to approximately 16% of pre-castrate levels by two weeks post-castration. These results are consistent with the findings in LNCaP cells and suggest that STMP1 expression is not significantly regulated by androgens in prostate cancer cells. STMP1 expression was substantially lower in the CWR22 tumors compared with LNCaP cells.
The expression profile of STMP1 was also analyzed in the androgenindependent prostate cancer cell lines PC3 and DU145, as well as in four independent, relapsed derivatives of CWR22 tumors, named CWR22R (Nagabhushan, M. et al., Cancer Res 56, 3042-3046, 1996), representative of advanced prostate cancer (Figure 6C). LNCaP (in the presence or absence ofR1881 (10 8 PC-3, or DU-145 cells were grown and total RNA was isolated. Four independent lines of the androgen independent human prostate cancer xenograft CWR22R, were grown in nude mice, tumors were collected, and total RNA was isolated and used in Northern analysis with STMP1 or the androgen target gene NKX3.1 cDNAs as probes. Ethidium bromide-stained 18S RNA is shown as a control for RNA integrity and loading. The relative induction of STMP1 and NKX3.1 mRNA accumulation is indicated at the bottom of the lanes determined by phosphorimager analysis (Molecular Dynamics). As is shown in Figure 6C, STMP1 expression was high in LNCaP cells and did not significantly change in response to R1881 treatment compared with a ~9-fold induction of the androgen target gene NKX3.1. There was no STMP1 expression in the androgen-independent prostate cancer cell lines PC-3 or DU-145, as was the case for NKX3.1. In contrast, there was significant STMP1 expression in tumors from all four independent CWR22R xenograft lines tested, ranging between -30-60% of that observed in LNCaP cells. A WO 01/72962 PCT/USU1/09410 similar overexpression pattern was also observed for NKX3.1 (Figure 6C) consistent with previous findings (Korkmaz, K. S. et al., Gene 260, 25-36, 2000).
An interesting property of STMP1 expression profile is that even though it is expressed at low levels in the androgen dependent CWR22 xenograft, it is highly expressed in the relapsed CWR22R which is androgen receptor (AR) positive, but is not responsive to androgens. This indicates that STMP1 expression is deregulated once the prostate tumor progresses from an androgen-dependent to an androgen-independent phase.
In addition, STMP1 is not expressed in the AR-negative prostate cancer cell lines PC-3 and DU-145, but is expressed at high levels in the AR-positive cell line LNCaP and the CWR22 and CWR22R xenografts. Thus, expression of STMP1 is correlated with the presence of a functional AR in the cell.
It has been known for over 50 years that androgens play a key role both in the development and maintenance of the normal prostate and the initiation and progression of prostate cancer. Androgen withdrawal results in involution of both the normal prostate gland as well as a prostate tumor in the early stages of the disease that is still androgen dependent.
Consequently, androgen withdrawal is commonly used as treatment to reverse tumor growth. However, in the case of the prostate tumor, after a few months or years, the tumor recurs in almost all cases in an androgenindependent state. At this point there is no effective therapy and prognosis for survival is extremely poor. Since STMP1 is overexpressed during this later androgen-insensitive state, it will be a useful tool in diagnostic and therapeutic applications for prostate cancer.
These data indicate that STMP1 expression is deregulated once prostate cancer progresses from an androgen-dependent to an androgenindependent state.
WO 01/72962 PCT/USU1/09410 EXAMPLE 7 Intracellular Localization Of STMP1 To gain insight into the intracellular localization pattern ofSTMP1, a green fluorescent protein (GFP)-STMP1 fusion protein was generated. The use of such GFP chimeric proteins has recently become a standard method to assess intracellular localization and dynamics of proteins. COS-1 cells were transiently transfected with GFP-STMP1, fixed and processed for confocal microscopy as described in Materials and Methods.
A series of 11 confocal sections along the z-axis were collected through a single cell at nominal 100 nm intervals. Three of the consecutive sections and the projection of all 11 sections are shown in Figure 7A.
Arrows indicate tubular-vesicular structures (VTS) in different sizes, shapes, and locations (Bar=5utm). In all 11 z-plane sections, GFP-STMP1 showed bright juxtanuclear distribution pattern, characteristic of the Golgi complex. Additionally, GFP-STMP1 was dispersed in spots of variable size throughout the cytoplasm and at the cell periphery projection). Some of these bright fluorescent spots were tubular arrow and Figure 8) or vesicular arrow) in morphology.
To determine more directly whether GFP-STMP 1 was localized to the Golgi complex, we compared its intracellular distribution with those of two well characterized Golgi markers, the medial Golgi enzyme mannosidase II (ManII) (Rabouille, C. et al., JCell Sci 108, 1617-1627, 1995) and the coat protein P-COP (Pepperkok, R. et al., Cell 74, 71-82, 1993). COS-1 cells were transfected with GFP-STMP1, fixed, labeled with the appropriate primary and secondary antibodies and imaged by confocal laser scanning microscopy. Green GFP-STMP1 fluorescence and red (Texas Red-labeled secondary antisera) 3-COP and ManII fluorescence were detected by confocal laser microscopy. Panels to the right show the WO 01/72962 PCT/USU1/09410 overlay images with yellow/orange staining indicating the regions of colocalization. Bars=5pm. As shown in Figure 7B, the distribution of GFP-STMP 1 extended throughout the Golgi complex, as evidenced by significant colocalization with both ManII and p-COP. However, some areas of non-overlap between the GFP-STMP1 and both Golgi markers were observed suggesting that STMP1, at least in part, is differentially localized within the Golgi complex compared with these two markers.
Since GFP-STMP1 was associated with VTS (Figure 7A and Figure 8), more specific localization of GFP-STMP 1 to the trans-Golgi network (TGN), an important site for the sorting of proteins destined to the plasma membrane, secretory vesicles, or lysosomes (Farquhar, M. G. Palade, G.
E. Trends CellBiol 8, 2-10, 1998; Mellman, I. Warren, Cell 100, 99- 112, 2000; Lemmon, S. K. Traub, L. Curr Opin Cell Biol 12, 457- 466, 2000) was assessed. An antibody against TGN46, a TGN resident protein that shuttles between the TGN and the plasma membrane (Prescott AR, et al., Eur J Cell Biol 72, 238-246, 1997; Ponnambalam, S. et al., J Cell Sci. 109, 675-685, 1996), was used in immunoflourescence microscopy experiments as above. As shown in Figure 7B, GFP-STMP1 extensively colocalized with TGN46, greater than that observed with ManII and P-COP, suggesting that in the Golgi complex, STMP1 is primarily localized to.the TGN. Note that the images with TGN46 were obtained with lower objective power.
EXAMPLE 8 STMP1 Shuttles Between The Golgi And The Plasma Membrane And Colocalizes To The Early Endosomes The dynamic properties and intracellular trafficking of GFP-STMP1 were studied using confocal time-lapse imaging in living cells. COS-1 cells were transiently transfected with GFP-STMP 1 and, 16 h after transfection, WO 01/72962 PCT/USU1/09410 12 consecutive images were collected from live cells every 20s at 37 0 C by confocal laser scanning microscope (Figure The upper panel shows a VTS extending out and retracting back to the Golgi body (white arrows).
In the middle panel and the first image in the lower panel (160s), red arrows indicate the translocation of a VTS from the Golgi body to the cell periphery. In the lower panel, yellow arrows point to the movement of a VTS from the edge of the cell towards the Golgi body. Note that the results shown are representative of multiple time-lapse analyses and the changes in the images are not due to movement from the plain of focus. Bar=5 pm.
As shown in Figure 8, some VTS were found to be detaching and some to be associating with the Golgi complex. The VTS were highly dynamic and pleiomorphic in size. Some of the VTS followed straight or curvilinear paths, some moved in a stop-and-go fashion, and some showed saltatory movements. The VTS indicated at the top panel (white arrows) extended away from and then retracted back to the Golgi. The VTS in the middle panel and the first image in the lower panel (red arrows) detached from the Golgi complex, paused, and then moved towards the cell periphery until it disappeared at the cell edge suggesting that STMP1 is associated with the secretory pathway. The VTS in the lower panel (yellow arrow) moved from the cell periphery towards the Golgi body suggesting that STMP 1 is localized to the endocytic pathway.
EXAMPLE 9 Colocalization Of GFP-STMP1 With The Early Endosomal Marker EEA1 To probe whether GFP-STMP1 was associated with the endocytic pathway, the intracellular distribution of GFP-STMP 1 was compared with that of the early endosome protein EEA1 (Stenmark, H. et al., JBiol Chem 271, 204048-204054, 1996). COS-1 cells were transfected with GFP- STMP1, fixed, immunostained with EEA1 antibodies and observed by WO 01/72962 PCT/USU1/09410 confocal laser scanning microscopy. Green GFP-STMP1 fluorescence and red (Texas Red-labeled secondary antiserum) EEA1 fluorescence were detected by confocal laser microscopy. The panel to the right shows the overlay images with yellow/orange staining indicating the regions of colocalization. Arrows indicate examples of the VTS in the cell periphery which contain both EEA1 and STMP1. Bar=5prm. As shown in Figure 9, EEA1 manifested a similar intracellular distribution in both transfected and untransfected cells. Furthermore, GFP-STMP1 significantly colocalized with EEA1 both in the cell periphery and also in the perinuclear area (Figure 9, arrows) suggesting that STMP1 is associated with early endosomes and the endocytic pathway.
EXAMPLE Isolation And Characterization of the SSH9 Gene And mRNA The SSH9 gene was identified and mapped (Figure 10). The predicted promoter site, the transcription start site, and the location and size of the exons and introns are indicated. The start and stop codons, as well as two polyadenylation signals, leading to two alternatively spliced transcripts, are also indicated. Figures 11A-C show the nucleotide and predicted amino acid sequence of SSH9, as well as the predicted promoter sequence and exon-intron boundaries.
The expression profile of SSH9, determined in various human tissues by Northern analysis (Figure 12C), revealed that the 0.7 kb splice variant of SSH9 was highly testis-specific, while the 1.4 kb transcript was expressed in both prostate and testis.
The androgen regulation of SSH9 was examined in LNCaP cells and in CWR22 xenografts (Figure 12A) revealed that SSH9 is not regulated in LNCaP cells, but is regulated in CWR22 xenografts. The expression WO 01/72962 PCT/USU1/09410 profile of SSH9 was also examined in the androgen-independent prostate cancer cell lines PC3 and DU145, and in CWR22R cells (Figure 12B).
EXAMPLE 11 Isolation And Characterization of the PSL22 Gene And mRNA The PSL22 gene was identified and mapped (Figure 13). The location and size of the exons and introns, the location of the partial cDNA clone (black box), as well as the alignment of the full-length cDNA clone with GenBank Accession Nos. AC008551 and AC011449, are indicated.
Figures 14A-C show the nucleotide sequence of the ORF, cDNA and predicted amino acid sequence, as well as the predicted promoter, exon, and UTR sequences of PSL22.
BLAST analysis of GenBank with the predicted PSL22 amino acid sequence identified PSL22 as a Rho binding protein. Figure 15 shows a multiple sequence alignment of PSL22 with related proteins. Completely conserved residues are shown in black; residues found in three sequences are shaded.
The expression profile ofPSL22, determined in various human tissues by Northern analysis (Figure 116B), revealed that while the highest expression was seen in the prostate, high expression was seen in the kidney, pancreas, and colon.
The androgen regulation of PSL22 was examined in LNCaP cells, in the androgen-independent prostate cancer cell lines PC3 and DU145, and in CWR22R cells (Figure 16A). The results showed that PSL22 is androgen regulated in LNCaP cells, where it is highly expressed, but is not androgen regulated in the PC3 and DU145 cells.
WO 01/72962 PCT/USU1/09410 EXAMPLE 12 Materials And Methods The following materials and methods were used in performing the exemplary experiments shown herein. It is understood that these materials and methods are subject to modifications that do not change the nature of the invention, as will be understood by those of ordinary skill in the art.
Probes Poly RNA 1 gg or Random primer (N7) 200 ng RNAse-free sterile H20 to 20 ul Heat at 70°C for 10 min, and chill on ice.
While heating the RNA samples, the following solution was prepared: 5X 1st strand buffer 10 gl 0.1 mM DTT 5 gl mM each dTTP+dGTP 2 gl 32 P alpha dATP 5 gl 32 P alpha dCTP 5 tl Superscript II (200 U l, BRL) 2pl The solution was mixed by pipetting, spun briefly, incubated at 250C for 5 min, and then for an additional 1 hour at 37°C. 2 tl of 10mM dCTP dATP was added and the mixture was incubated for 30 min at 37°C and then heat inactivated at 70°C for 10 min. Unincorporated nucleotides were removed using prespun G25 columns (Bio-Rad). Specific activity (which should be over 5xl08cpm/gg) was calculated.
WO 01/72962 PCT/USU1/09410 Hybridization Freshly prepared 25 ml Hybridization mix SDS, 0.5 M NaHPO 4 ImM EDTA) was pre-warmed at 65 0 C and 12.5 ml was used for prehybridization of each membrane, 5-10 min at 65 The probe was heat denatured at 95 0 C for 3-5 min and transferred to the prehybridization mix at 65 0 C. Hybridization was carried out at 65 0 C overnight.
Washing Wash solution I (2xSSC and 1% SDS) and II (0.lxSSC and SDS) were prewarmed, and the membrane were washed once with Solution I and then with Solution II for 30 min at 65 0 C. The membranes were covered with plastic wrap and exposed to a phoshorimager screen.
Selection Clones that showed differences between the and blots were picked (usually 1-8 on each blot pair). A secondary round of reverse northern analysis for confirmation was performed, this time spotting each clone in duplicate on each blot. After phosphorimager analysis, the blots were stripped in 0.lxSSC and 0.5% SDS for 2x15 min at 95 0 C and hybridized with a PSA probe (or depending on the hormone that is being used, with a probe for any abundant target genes in the tissue under study).
For the clones that were confirmed to be different from PSA, for differential expression in the secondary reverse northern, northern analysis was performed using established protocols. A time course ofR1881 induction of LNCaP cells, as well as the CWR22 xenograft model upon androgen ablation (Wainstein, M. A. et al., Cancer Res. 54, 6049-6052, 1994) and the androgen-independent CWR22R relapsed xenograft (Nagabhushan, M. et al., Cancer Res. 56, 3042-6, 1996), was used.
WO 01/72962 PCT/USU1/09410 Sequence analysis Sequence analysis was perfomed by the dideoxy chain termination methods using an ABI automated sequencer. Homology search was done using a basic BLAST algorithm. Figure 3 shows a table of results obtained from the BLAST analysis of isolated clones and their homology to known genes (The cutoff for significant homology was 50% identity).
Isolation of prostate cancer related genes from LNCaP cells The prostate cancer cell line LNCaP was cultured in two batches in culture conditions similar to those previously described (Horoszewicz JS et al., Cancer Res. 43: 1809-1818, 1983). The first batch was left untreated, while the second batch was treated with the synthetic androgen R1881 for 24 hrs. Cells from both batches were harvested and total RNA was then isolated from each batch. From the total RNA, polyA RNA was obtained using standard procedures, and was used in the Suppression Subtraction Hybridization (SSH; Diatchenko et al., supra) procedure to identify hormone regulated genes. The tester in the SSH procedure was cDNA from untreated cells and the driver was cDNA from R1881-treated cells. The suppression subtraction protocol was performed according to the original description of the method (Diatchenko et al., supra).
Cell culture LNCaP, PC-3 and DU-145 cells were routinely maintained and treated as described previously (Korklnaz, K. S. et al., DNA Cell Biol 19, 499-506, 2000; Korkmaz, K. S. et al., Gene 260, 25-36, 2000).
WO 01/72962 PCT/USU1/09410 Xenograft studies Transplantation, growth, and harvesting of tumors from mice bearing the CWR22 and CWR22R xenografts were as previously described (Wainstein, M. supra; Nagabhushan, supra).
Cloning and plasmid construction A 262 bp cDNA fragment was originally obtained from a screen of a prostate specific library (Ausubel, F. et al. (1997) Current Protocols in Molecular Biology (John Wiley and Sons, New York) and termed L74. Rapid Amplification of cDNA Ends (RACE) was performed (oligonucleotide sequences available upon request) using the Marathon- Ready cDNA that was prepared from normal prostate tissue (Clontech) and/or SMART-RACE LNCaP cDNA library (Clontech) that was generated according to the manufacturer's recommendations. RACE products were cloned into pCRII-TOPO (Invitrogen), positive clones were confirmed by Southern analysis, and sequenced. In parallel, a Xgtl0 cDNA library made from a pool of normal human prostates (Clontech) was screened by established procedures to obtain additional clones. Overlapping clones were used to deduce the full-length STMP1 cDNA sequence.
The full-length STMP1 ORF was amplified by using primers centered around the start and stop codons (sequences available upon request) and fused in frame to the C-terminus of green flourescent protein (GFP) using the vector pcDNA3.1-NT-GFP-TOPO (Invitrogen) to generate GFP- STMP1.
Northern analysis Total RNA was prepared by the single step guanidine thiocyanate procedure and used in Northern analysis 15 |tg of total RNA was used per lane. Probes were generated by random priming and had a specific WO 01/72962 PCT/USU1/09410 activity of >3x10 8 dpm/4g. A cDNA fragment of STMP1 spanning residues 145-2202 bp was used as probe. Bands were visualized and quantitated by phosphorimager analysis (Molecular Dynamics).
Confocal microscopy COS-1 cells were transiently transfected by electroporation using a BTX square-wave pulser at 150 V, 1 ms duration. Cells were grown either on cover slips placed in 6-well tissue culture plates for indirect immunofluorescence or on Lab-Tek Chambered Coverglass (Nalge Nunc International) for live-cell microscopy. Transiently transfected cells were observed 16 h after transfection by Leica TCS-SP confocal microscope. All live-cell experiments were done at 37C.
Indirect immunofluorescence The indirect immunofluorescence was carried out as previously described (Misteli, T. Spector, D. L. Mol Cell 3, 697-705, 1999). The following antibodies were used: anti-p-coat protein (p-COP) antiserum (kindly provided by J. Lippincott-Schwartz), anti-mannosidase II (kindly provided by T. Misteli), anti-TGN46 (Serotec, kindly provided by J.S.
Bonifacino), and anti-EEA1 (Affinity Biotechnologies). Texas Redconjugated secondary antibodies specific for mouse and rabbit were purchased from ICN Biomedicals (Costa Mesa, CA).
Other Embodiments All publications and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each independent publication or patent application was specifically and individually indicated to be incorporated by reference.
WO 01/72962 PCT/USU1/09410 While the invention has been described in connection with specific embodiments thereof, it will be understood that it is capable of further modifications and this application is intended to cover any variations, uses, or adaptations of the invention following, in general, the principles of the invention and including such departures from the present disclosure that come within known or customary practice within the art to which the invention pertains and may be applied to the essential features hereinbefore set forth, and follow in the scope of the appended claims.

Claims (1)

  1. 69- THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS: ct 1. A substantially pure prostate-specific or testis-specific polypeptide, said polypeptide sequence comprising a sequence at least 80% identical to the sequence of SEQ ID NO 34. 2. A substantially pure prostate-specific or testis-specific polypeptide, said polypeptide sequence comprising a sequence at least 95% identical to the sequence of SSEQ ID NOS: 14 or 32. oo 3. The substantially pure prostate-specific or testis-specific polypeptide according to claim 1, said polypeptide sequence comprising the sequence of SEQ ID NO 34. 4. The substantially pure prostate-specific or testis-specific polypeptide according to claim 2, said polypeptide sequence comprising the sequence of SEQ ID NOS: 14 or 32. An isolated nucleic acid molecule encoding a polypeptide according to any one of claims 1 to 4. 6. The isolated nucleic acid according to claim 5, wherein said nucleic acid molecule comprises the sequence of any of SEQ ID NOS: 13, 23, 31, or 33. 7. An isolated prostate-specific or testis-specific nucleic acid molecule, said nucleic acid molecule comprising a sequence at least 80% identical to SEQ ID NOS: 9. 15-22, 24-27, or 33. 8. An isolated-prostate-specific or testis-specific nucleic acid molecule, said nucleic acid molecule comprising a sequence at least 95% identical to SEQ ID NOS: 13, 23, or i:Malbolllr Cn OICAoa M'700. 4l A P4I CAUSp'=|1P471c. ALJ pctriUlmllkn 2007-.I.- dnt It IM7 COMS ID No: SBMI-05942216 Received by IP Australia: Time 17:21 Date 2007-01-16 16/01 2007 17:11 FAX 61 3 32438333 GRIFFITH HACK IPAUSTRALIA 1011/026 S- 70 9. The isolated prostate-specific or testis-specific nucleic acid molecule according to claim 7, said nucleic acid molecule consisting essentially of SEQ ID NOS: 9, 15-22, 24-27, or 33. 10. The isolated prostate-specific or testis-specific nucleic acid molecule according to claim 8, said nucleic acid molecule consisting essentially of SEQ ID NOS: 13, 23, or O 11. The polypeptide according to any one of claims 1 to 4, wherein said polypeptide C 10 is derived from a mammal, 12. The polypeptide according to claim 11, wherein said mammal is a human. 13. A vector comprising the isolated nucleic acid molecule according to any one of is claims 5 to 14. A cell comprising the isolated nucleic acid molecule according to any one of claims 5 to 15. A cell comprising the vector according to claim 13. 16, A non-human transgenic animal comprising the isolated nucleic acid molecule according to any one of claims 5 to 17. An isolated nucleic acid molecule that hybridizes under high stringency conditions to the complement of a sequence at least 80% identical to SEQ ID NOS: 9, 15-22, 24-27, or 33 wherein said isolated nucleic acid molecule encodes a prostate-specific or testis- specific polypeptide. NAMdltbiinivtCauPalit O.l700d.47 l P47164.A7IpAU C\l P4d7Iod.AU Spolcricaik2007-1.16 la IO 07 COMS ID No: SBMI-05942216 Received by IP Australia: Time 17:21 Date 2007-01-16 16/01 2007 17:12 FAX 61 3 92438333 GRIFFITH HACK IPAUSTRALIA 1a012/026 0 -71- 18. An isolated nucleic acid molecule that hybridizes under high stringency Sconditions to the complement of a sequence at least 95% identical to SEQ ID NOS: 13, 23, or wherein said isolated nucleic acid molecule encodes a prostate-specific or testis- s specific polypeptide. S19. An isolated nucleic acid molecule, wherein said nucleic acid molecule CN comprises a sequence that is antisense to the coding strand of a prostate-specific or O testis-specific nucleic acid molecule of a sequence at least 80% identical to SEQ ID C, 10 NOS: 9, 15-22, 24-27, or 33. An isolated nucleic acid molecule, wherein said nucleic acid molecule comprises a sequence that is antisense to the coding strand ofa prostate-specific or testis-specific nucleic acid molecule ofa sequence at least 95% identical to SEQ ID NOS: 13, 23, or 21. A probe for analyzing a prostate-specific or testis-specific gene or homolog or fragment thereof, said probe having greater than 55% nucleotide sequence identity to a sequence encoding any of SEQ ID NOS: 9, 15-22, 24-27, or 33, or fragment thereof, wherein said fragment comprises at least six amino acids, and said probe hybridizes under high stringency conditions to at least a portion ofa prostate-specific or testis-specific nucleic acid molecule. 22. A probe for analyzing a prostate-specific or testis-specific gene or homolog or fragment thereof, said probe having greater than 95% nucleotide sequence identity to SEQ ID NOS: 13, 23, or wherein said fragment comprises at least six amino acids, and said probe hybridizes under high stringency conditions to at least a portion of a prostate-specific or testis-specific nucleic acid molecule. N:\Mnl lnibo\Clur t\CsPmboni Wl-47O Q 716 AU\%Spoi.\P7164.Al Spdfneial n iO?7--16 ct l,'01)07 COMS ID No: SBMI-05942216 Received by IP Australia: Time 17:21 Date 2007-01-16 16/01 2007 17:12 FAX 61 3 82438333 GRIFFITH HACK IPAUSTRALIA 1a13/026 0 -72- 23. The probe according to claim 21, wherein said probe has 100% complementarity Sto a nucleic acid molecule encoding any ofSEQ ID NOS: 9, 15-22, 24-27, or 33, or a fragment thereof, wherein said fragment comprises at least six amino acids, and said probe s hybridizes under high stringency conditions to at least a portion of a prostate-specific or testis-specific nucleic acid molecule. C' 24. The probe according to claim 22, wherein said probe has 100% complementarity O to a nucleic acid molecule of SEQ ID NOS: 13, 23, or C 1 0 wherein said fragment comprises at least six amino acids, and said probe hybridizes under high stringency conditions to at least a portion of a prostate-specific or testis-specific nucleic acid molecule. An antibody that specifically binds to a prostate-specific or testis-specific is polypeptide, said polypeptide comprising an amino acid sequence that is at least identical to the amino acid sequence of SEQ ID NO 34. 26. An antibody that specifically binds to a prostate-specific or testis-specific polypeptide, said polypeptide comprising an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 14 or 32. 27. A method of detecting a prostate-specific or testis-specific gene or fragment thereof in a cell, said method comprising: contacting the nucleic acid molecule of any of SEQ ID NOS: 9, 15-22, 24-27, 33, or 35, or a fragment thereof, wherein said fragment is greater than about 18 nucleotides in length, with a preparation of genomic DNA from said cell, under high stringency hybridization conditions; and detecting DNA sequences having about 55% or greater nucleotide sequence identity to any of SEQ ID NOS: 9, 15-22, 24-27, 33, or 3o thereby identifying a prostate-specific or testis-specific gene or fragment thereof N:\Mulbotun\Cuauni\anl7O8470 099\ P71 AU3KUshPa7I MI.AU Spl hMion 3007-1 16 doc 1601/07 COMS ID No: SBMI-05942216 Received by IP Australia: Time 17:21 Date 2007-01-16 16/01 2007 17:12 FAX 61 3 92438333 GRIFFITH HACK IPAUSTRLIA la-14/021 -73- 28. A method of detecting a prostate-specific or testis-specific gene or fragment thereof in a cell, said method comprising: contacting the nucleic acid molecule of SEQ ID NOS: 13, 23, or under high stringency hybridization conditions; and s detecting DNA sequences having about 95% or greater nucleotide sequence identity to SEQ ID NOS: 13, 23, or thereby identifying a prostate-specific or testis-specific gene or fragment thereof. 29. A method for identifying a test compound that modulates the expression or activity of a prostate-specific or testis-specific polypeptide, wherein said prostate- specific or said testis-specific polypeptide comprises an amino acid sequence at least identical to the amino acid sequence of SEQ ID NOS: 34 or 36, or fragments or analogs thereof, said method comprising: contacting said prostate-specific or testis-specific polypeptide with said is test compound; and determining the effect of said test compound on said prostate-specific or testis-specific polypeptide expression or activity. A method for identifying a test compound that modulates the expression or activity of a prostate-specific or testis-specific polypeptide, wherein said prostate- specific or said testis-specific polypeptide comprises an amino acid sequence at least identical to the amino acid sequence of SEQ ID NO: 14 or 32, said method comprising: contacting said prostate-specific or testis-specific polypeptide with said test compound; and determining the effect of said test compound on said prostate-specific or testis-specific polypeptide expression or activity. 31. A method of treating a mammal having a disorder of the prostate or testis, said method comprising administering to said mammal a therapeutically effective amount of a compound that modulates the activity or expression of a prostate-specific or testis- specific polypeptide, wherein said prostate-specific or said testis-specific polypeptide r:M;Il burm alall\ Pmlini]M'lOo l79Q\P 71 d.A Spacijm47E .AU icllcgillin ,7-l Io c lW i/o 7 COMS ID No: SBMI-05942216 Received by IP Australia: Time 17:21 Date 2007-01-16 16/01 2007 17:13 FAX 61 3 92438333 GRIFFITH HACK IPAUSTRALIA 1a015/026 8 74 Scomprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID) NOS: 34 or 36, or fragments or analogs thereof, wherein said compound has a beneficial effect on said disorder in said mammal. 32. A method of treating a mammal having a disorder of the prostate or testis, said method comprising administering to said mammal a therapeutically effective amount of 0a compound that modulates the activity or expression of a prostate-specific or testis- C'l specific polypeptide, wherein said prostate-specific or said testis-specific polypeptide 0 comprises an amino acid sequence at least 95% identical to the amino acid sequence of C' .0 SEQ ID NO: 14 or 32, wherein said compound has a beneficial effect on said disorder in said mammal. 33. The method according to claim 31or claim 32, wherein said disorder is prostate cancer. 34. The method according to claim 31 or claim 32, wherein said mammal is a human. Use of a compound that modulates the activity or expression of a prostate- 2 o specific or testis-specific polypeptide, in the manufacture of a medicament for treating a mammal having a disorder of the prostate or testis, wherein said prostate-specific or said testis-specific polypeptide comprises an amino acid sequence at least 80%/ identical to the amino acid sequence of SEQ ID NOS: 34 or 36, or fragments or analogs thereof 36. Use of a compound that modulates the activity or expression of a prostate- specific or testis-specific polypeptide, in the manufacture of a medicament for treating a mammal having a disorder of the prostate or testis, wherein said prostate-specific or said testis-specific polypeptide comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 14 or 32. 37. A pharmaceutical composition comprising at least one dose of a therapeutically effective amount of a prostate-specific or testis-specific polypeptide or fragment NI4itMb unitlCwmpMl'mn l701.i7QMPa7l6a.AU\Spe pn4716,AU SpinFtim 2DI07-1-16 dn I110[107 COMS ID No: SBMI-05942216 Received by IP Australia: Time 17:21 Date 2007-01-16 16/01 2007 17:13 FAX 61 3 92438333 GRIFFITH HACK IPAUSTRLIA IaO11/026 0 75 0 7 thereof, in a pharmaceutically acceptable carrier, wherein said prostate-specific or said c testis-specific polypeptide comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NO: 34, or fragments or analogs thereof, said composition being formulated for the treatment of a disorder of the prostate or testis. 38. A pharmaceutical composition comprising at least one dose of a therapeutically ON effective amount of a prostate-specific or testis-specific polypeptide or fragment Ci thereof, in a pharmaceutically acceptable carrier, wherein said prostate-specific or said 0 testis-specific polypeptide comprises an amino acid sequence at least 95% identical to Co the amino acid sequence of SEQ ID NO: 14 or 32, said composition being formulated for the treatment of a disorder of the prostate or testis. 39. A kit for the analysis of a prostate-specific or testis-specific nucleic acid molecule or fragment thereof, said kit comprising a nucleic acid molecule probe for analyzing a prostate-specific or testis-specific nucleic acid molecule present in a test subject, wherein said probe comprises a nucleotide sequence having greater than nucleotide sequence identity to a sequence encoding any of SEQ ID NOS: 9, 15-22, 24- 27, 33, or 35, or fragment thereof, wherein said fragment comprises at least six amino acids, and said probe hybridizes under high stringency conditions to at least a portion of a prostate-specific or testis-specific nucleic acid molecule. A kit for the analysis of a prostate-specific or testis-specific nucleic acid molecule or fragment thereof, said kit comprising a nucleic acid molecule probe for analyzing a prostate-specific or testis-specific nucleic acid molecule present in a test subject, wherein said probe comprises a nucleotide sequence having greater than nucleotide sequence identity to a sequence encoding SEQ ID NOS: 13, 23, or wherein said fragment comprises at least six amino acids, and said probe hybridizes under high stringency conditions to at least a portion of a prostate-specific or testis-specific nucleic acid molecule. 41. A kit for the analysis of a prostate-specific or testis-specific polypeptide, said kit comprising an antibody for analyzing a prostate-specific or testis-specific polypeptide N;rM lo oCuwtl IHs IPlllIXI7.d799\P47 I4,AlSpecill\P<7164.AU SPdSicri n 1-16 do IW1/01 k7 COMS ID No: SBMI-05942216 Received by IP Australia: Time 17:21 Date 2007-01-16 16/01 2007 17:13 FAX 61 3 92438333 GRIFFITH HACK IPAUSTRALIA 1a017/026 -76 present in a test subject, wherein said prostate-specific or said testis-specific polypeptide comprises an amino acid sequence at least 80% identical to the amino acid sequence of SEQ ID NOS: 34 or 36, or fragments or analogs thereof. s 42. A kit for the analysis of a prostate-specific or testis-specific polypeptide, said kit comprising an antibody for analyzing a prostate-specific or testis-specific polypeptide present in a test subject, wherein said prostate-specific or said testis-specific polypeptide comprises an amino acid sequence at least 95% identical to the amino acid sequence of SEQ ID NO: 14 or 32. 43. A substantially pure prostate-specific or testis-specific polypeptide according to claim I or claim 2, an isolated nucleic acid molecule according to any one of claims 7, 8, and 17 to 20, a vector according to claim 13, a cell according to claim 14, a non- human trnsgenic animal according to claim 16, a probe according to claim 21 or claim is 22, an antibody according to claim 25 or claim 26, a method according to any one of claims 27 to 32, a use according to claim 35 or claim 36, a pharmaceutical composition according to claim 37 or claim 38, or a kit according to any one of claims 40 to 42, substantially as herein described with reference to any one of the examples and/or figures. "rMlcuntcumn Fuwinogu '64Av epyi~r,71g,&.Au lI-SpmiRlmsl 2007,1. 1LX 1w0107 COMS ID No: SBMI-05942216 Received by IP Australia: Time 17:21 Date 2007-01-16
AU2001249411A 2000-03-24 2001-03-23 Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides,and diagnostic and therapeutic methods Ceased AU2001249411B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US19192900P 2000-03-24 2000-03-24
US60/191,929 2000-03-24
PCT/US2001/009410 WO2001072962A2 (en) 2000-03-24 2001-03-23 Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods

Publications (3)

Publication Number Publication Date
AU2001249411B9 true AU2001249411B9 (en) 2001-10-08
AU2001249411A1 AU2001249411A1 (en) 2001-12-20
AU2001249411B2 AU2001249411B2 (en) 2007-02-15

Family

ID=22707491

Family Applications (2)

Application Number Title Priority Date Filing Date
AU2001249411A Ceased AU2001249411B2 (en) 2000-03-24 2001-03-23 Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides,and diagnostic and therapeutic methods
AU4941101A Pending AU4941101A (en) 2000-03-24 2001-03-23 Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides,and diagnostic and therapeutic methods

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU4941101A Pending AU4941101A (en) 2000-03-24 2001-03-23 Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides,and diagnostic and therapeutic methods

Country Status (5)

Country Link
EP (1) EP1268526A4 (en)
JP (1) JP2004521602A (en)
AU (2) AU2001249411B2 (en)
CA (1) CA2403637A1 (en)
WO (1) WO2001072962A2 (en)

Families Citing this family (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030149531A1 (en) 2000-12-06 2003-08-07 Hubert Rene S. Serpentine transmembrane antigens expressed in human cancers and uses thereof
US20060052321A1 (en) 2002-04-05 2006-03-09 Raitano Arthur B Nucleic acid and corresponding protein entitled 98P4B6 useful in treatment and detection of cancer
DK1086223T3 (en) 1998-06-01 2009-11-30 Agensys Inc Novel serpentine transmembrane antigens expressed in human cancers and their applications
US20040048798A1 (en) * 1999-06-01 2004-03-11 Raitano Arthur B. Nucleic acid and corresponding protein entitle 98P4B6 useful in treatment and detection of cancer
US20040141975A1 (en) 1998-06-01 2004-07-22 Raitano Arthur B. Nucleic acid and corresponding protein entitled 98P4B6 useful in treatment and detection of cancer
US6833438B1 (en) 1999-06-01 2004-12-21 Agensys, Inc. Serpentine transmembrane antigens expressed in human cancers and uses thereof
AU769889B2 (en) * 1999-04-29 2004-02-05 University Of Tennessee Research Corporation, The Isolated nucleic acids of the p-Hyde family, P-Hyde proteins, and methods of inducing susceptibility to induction of cell death in cancer
US6835812B1 (en) 1999-04-29 2004-12-28 University Of Tennessee Research Corporation Human p-Hyde proteins
US7611892B2 (en) 2000-03-24 2009-11-03 President And Fellows Of Harvard College Prostate-specific or testis-specific nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods
WO2005114216A2 (en) * 2005-02-22 2005-12-01 Biomedisinsk Innovasjon As Stamp2 nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods
GB2379660A (en) * 2001-01-30 2003-03-19 Aeomica Inc Human GTP-Rho binding protein 2
EP1231216A3 (en) * 2001-01-30 2004-03-17 Aeomica, Inc. Human gtp-rho binding protein 2
EP2287186B1 (en) 2001-09-06 2014-12-31 Agensys, Inc. Nucleic acid and corresponding protein entitled STEAP-1 useful in treatment and detection of cancer
US7494646B2 (en) 2001-09-06 2009-02-24 Agensys, Inc. Antibodies and molecules derived therefrom that bind to STEAP-1 proteins
JP2005534287A (en) * 2002-04-05 2005-11-17 アジェンシス, インコーポレイテッド Nucleic acids named 98P4B6 and corresponding proteins useful in the treatment and detection of cancer
JP5840351B2 (en) * 2002-09-06 2016-01-06 アジェンシス,インコーポレイテッド Nucleic acids and corresponding proteins referred to as 98P4B6 useful in the treatment and detection of cancer
BR122018071808B8 (en) 2003-11-06 2020-06-30 Seattle Genetics Inc conjugate
GB0326834D0 (en) * 2003-11-18 2003-12-24 Celltech R&D Ltd A protein involved in lung cancer
JP4445925B2 (en) 2003-12-25 2010-04-07 富士フイルム株式会社 Organic EL element, organic EL display device, organic EL element manufacturing method, and organic EL element manufacturing apparatus
CN101258166B (en) 2004-04-22 2013-01-30 阿根西斯公司 Antibody combining with STEAP-1 protein and deriving molecule thereof
JP5234734B2 (en) 2004-06-01 2013-07-10 ジェネンテック, インコーポレイテッド Antibody-drug conjugates and methods
US20100111856A1 (en) 2004-09-23 2010-05-06 Herman Gill Zirconium-radiolabeled, cysteine engineered antibody conjugates
EP3088004B1 (en) 2004-09-23 2018-03-28 Genentech, Inc. Cysteine engineered antibodies and conjugates
KR101541550B1 (en) 2006-10-27 2015-08-04 제넨테크, 인크. Antibodies and immunoconjugates and uses therefor
JP5240205B2 (en) * 2008-01-11 2013-07-17 アステラス製薬株式会社 Model animal with testicular pain or discomfort and frequent urination
US8470980B2 (en) 2009-09-09 2013-06-25 Centrose, Llc Extracellular targeted drug conjugates
CA2793890C (en) 2010-04-15 2017-08-15 Spirogen Developments Sarl Pyrrolobenzodiazepines and conjugates thereof
JP2013534520A (en) 2010-06-08 2013-09-05 ジェネンテック, インコーポレイテッド Cysteine engineered antibodies and conjugates
US20120121615A1 (en) 2010-11-17 2012-05-17 Flygare John A Alaninyl maytansinol antibody conjugates
EP2707723B1 (en) 2011-05-12 2016-02-10 Genentech, Inc. Multiple reaction monitoring lc-ms/ms method to detect therapeutic antibodies in animal samples using framework signature pepides
MX350152B (en) 2011-10-14 2017-08-29 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof.
WO2013130093A1 (en) 2012-03-02 2013-09-06 Genentech, Inc. Biomarkers for treatment with anti-tubulin chemotherapeutic compounds
BR112015008173A2 (en) 2012-10-12 2017-11-28 Adc Therapeutics Sarl pyrrolobenzodiazepine-anti-psma antibody conjugates
SI2906251T1 (en) 2012-10-12 2018-01-31 Adc Therapeutics Sa Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
HUE045435T2 (en) 2012-10-12 2019-12-30 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof
EP2906250B1 (en) 2012-10-12 2018-05-30 ADC Therapeutics SA Pyrrolobenzodiazepine-anti-psma antibody conjugates
DK2906296T3 (en) 2012-10-12 2018-05-22 Adc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugates
ES2660029T3 (en) 2012-10-12 2018-03-20 Medimmune Limited Antibody-pyrrolobenzodiazepine conjugates
CN105050661B (en) 2012-10-12 2018-03-30 Adc疗法责任有限公司 Pyrrolobenzodiazepines Zhuo antibody conjugates
CA2894961C (en) 2012-12-21 2020-09-15 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
AU2013366490B9 (en) 2012-12-21 2018-02-01 Medimmune Limited Unsymmetrical pyrrolobenzodiazepines-dimers for use in the treatment of proliferative and autoimmune diseases
WO2014140174A1 (en) 2013-03-13 2014-09-18 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
AU2014244245C1 (en) 2013-03-13 2018-04-19 Genentech, Inc. Pyrrolobenzodiazepines and conjugates thereof
WO2014140862A2 (en) 2013-03-13 2014-09-18 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
TWI636792B (en) 2013-08-12 2018-10-01 建南德克公司 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
GB201317982D0 (en) 2013-10-11 2013-11-27 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2015052532A1 (en) 2013-10-11 2015-04-16 Spirogen Sàrl Pyrrolobenzodiazepine-antibody conjugates
WO2015052535A1 (en) 2013-10-11 2015-04-16 Spirogen Sàrl Pyrrolobenzodiazepine-antibody conjugates
EP3054985B1 (en) 2013-10-11 2018-12-26 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
CN105873614B (en) 2013-12-16 2020-10-30 基因泰克公司 Peptidomimetic compounds and antibody-drug conjugates thereof
EA201691023A1 (en) 2013-12-16 2016-10-31 Дженентек, Инк. PEPTIDOMIMETIC CONNECTIONS AND THEIR CONJUGATES ANTIBODIES WITH MEDICINE
MX2016007578A (en) 2013-12-16 2016-10-03 Genentech Inc 1-(chloromethyl)-2,3-dihydro-1h-benzo[e]indole dimer antibody-drug conjugate compounds, and methods of use and treatment.
JP6531166B2 (en) 2014-09-10 2019-06-12 メドイミューン・リミテッドMedImmune Limited Pyrrolobenzodiazepine and its conjugate
AR101844A1 (en) 2014-09-12 2017-01-18 Genentech Inc ANTIBODIES AND GENETICALLY MODIFIED CONJUGATES WITH CYSTEINE
GB201416112D0 (en) 2014-09-12 2014-10-29 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof
EP3191134B1 (en) 2014-09-12 2019-11-20 Genentech, Inc. Anthracycline disulfide intermediates, antibody-drug conjugates and methods
EP3194400A1 (en) 2014-09-17 2017-07-26 Genentech, Inc. Pyrrolobenzodiazepines and antibody disulfide conjugates thereof
BR112017011111A2 (en) 2014-11-25 2017-12-26 Adc Therapeutics Sa pyrrolobenzodiazepine-antibody conjugates
KR20170086121A (en) 2014-12-03 2017-07-25 제넨테크, 인크. Quaternary amine compounds and antibody-drug conjugates thereof
GB201506411D0 (en) 2015-04-15 2015-05-27 Bergenbio As Humanized anti-axl antibodies
GB201506402D0 (en) 2015-04-15 2015-05-27 Berkel Patricius H C Van And Howard Philip W Site-specific antibody-drug conjugates
MA43345A (en) 2015-10-02 2018-08-08 Hoffmann La Roche PYRROLOBENZODIAZEPINE ANTIBODY-DRUG CONJUGATES AND METHODS OF USE
MA43354A (en) 2015-10-16 2018-08-22 Genentech Inc CONJUGATE DRUG CONJUGATES WITH CLOUDY DISULPHIDE
MA45326A (en) 2015-10-20 2018-08-29 Genentech Inc CALICHEAMICIN-ANTIBODY-DRUG CONJUGATES AND METHODS OF USE
GB201601431D0 (en) 2016-01-26 2016-03-09 Medimmune Ltd Pyrrolobenzodiazepines
GB201602356D0 (en) 2016-02-10 2016-03-23 Medimmune Ltd Pyrrolobenzodiazepine Conjugates
GB201602359D0 (en) 2016-02-10 2016-03-23 Medimmune Ltd Pyrrolobenzodiazepine Conjugates
JP6943872B2 (en) 2016-03-25 2021-10-06 ジェネンテック, インコーポレイテッド Multiple whole antibody and antibody complex drug quantification assay
GB201607478D0 (en) 2016-04-29 2016-06-15 Medimmune Ltd Pyrrolobenzodiazepine Conjugates
PL3458101T3 (en) 2016-05-20 2021-05-31 F. Hoffmann-La Roche Ag Protac antibody conjugates and methods of use
US20170370906A1 (en) 2016-05-27 2017-12-28 Genentech, Inc. Bioanalytical analysis of site-specific antibody drug conjugates
WO2017214024A1 (en) 2016-06-06 2017-12-14 Genentech, Inc. Silvestrol antibody-drug conjugates and methods of use
CN109689111B (en) 2016-08-11 2024-04-05 基因泰克公司 Pyrrolobenzodiazepine prodrugs and antibody conjugates thereof
WO2018065501A1 (en) 2016-10-05 2018-04-12 F. Hoffmann-La Roche Ag Methods for preparing antibody drug conjugates
GB201617466D0 (en) 2016-10-14 2016-11-30 Medimmune Ltd Pyrrolobenzodiazepine conjugates
GB201702031D0 (en) 2017-02-08 2017-03-22 Medlmmune Ltd Pyrrolobenzodiazepine-antibody conjugates
DK3544636T3 (en) 2017-02-08 2021-05-10 Adc Therapeutics Sa Pyrrolobenzodiazepine antibody conjugates
CA3055132A1 (en) 2017-04-03 2018-10-11 F. Hoffmann-La Roche Ag Antibodies binding to steap-1
DK3612537T3 (en) 2017-04-18 2022-08-08 Medimmune Ltd PYRROLOBENZODIAZEPIN CONJUGATES
CA3057748A1 (en) 2017-04-20 2018-10-25 Adc Therapeutics Sa Combination therapy with an anti-axl antibody-drug conjugate
KR102442736B1 (en) 2017-06-14 2022-09-16 에이디씨 테라퓨틱스 에스에이 Dosage regime for administration of anti-CD19 ADCs
KR102312910B1 (en) 2017-08-18 2021-10-15 메디뮨 리미티드 Pyrrolobenzodiazepine conjugates
TW201920192A (en) 2017-09-20 2019-06-01 韓商Ph製藥公司 THAILANSTATIN analogs
GB201803342D0 (en) 2018-03-01 2018-04-18 Medimmune Ltd Methods
GB201806022D0 (en) 2018-04-12 2018-05-30 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof
GB201814281D0 (en) 2018-09-03 2018-10-17 Femtogenix Ltd Cytotoxic agents
CN113056287A (en) 2018-10-24 2021-06-29 豪夫迈·罗氏有限公司 Conjugated chemical degradation inducers and methods of use
WO2020123275A1 (en) 2018-12-10 2020-06-18 Genentech, Inc. Photocrosslinking peptides for site specific conjugation to fc-containing proteins
GB201901197D0 (en) 2019-01-29 2019-03-20 Femtogenix Ltd G-A Crosslinking cytotoxic agents
GB2597532A (en) 2020-07-28 2022-02-02 Femtogenix Ltd Cytotoxic compounds
WO2023147329A1 (en) 2022-01-26 2023-08-03 Genentech, Inc. Antibody-conjugated chemical inducers of degradation and methods thereof
WO2023147328A1 (en) 2022-01-26 2023-08-03 Genentech, Inc. Antibody-conjugated chemical inducers of degradation with hydolysable maleimide linkers and methods thereof

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5599686A (en) * 1994-06-28 1997-02-04 Merck & Co., Inc. Peptides
US5820880A (en) * 1995-06-07 1998-10-13 The United States Of America As Represented By The Secretary Of The Army Liposomal formulation
US6048970A (en) * 1998-05-22 2000-04-11 Incyte Pharmaceuticals, Inc. Prostate growth-associated membrane proteins
DK1086223T3 (en) * 1998-06-01 2009-11-30 Agensys Inc Novel serpentine transmembrane antigens expressed in human cancers and their applications
EP1268758A2 (en) * 1999-06-01 2003-01-02 Incyte Genomics, Inc. Molecules for diagnostics and therapeutics
JP2003505029A (en) * 1999-07-19 2003-02-12 インサイト・ゲノミックス・インコーポレイテッド GTP binding protein

Also Published As

Publication number Publication date
AU2001249411B2 (en) 2007-02-15
CA2403637A1 (en) 2001-10-04
AU4941101A (en) 2001-10-08
WO2001072962A3 (en) 2002-01-17
EP1268526A2 (en) 2003-01-02
WO2001072962A2 (en) 2001-10-04
EP1268526A4 (en) 2004-09-08
JP2004521602A (en) 2004-07-22

Similar Documents

Publication Publication Date Title
AU2001249411B9 (en) Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides,and diagnostic and therapeutic methods
AU2001249411A1 (en) Novel prostate-specific or testis-specific nucleic acid molecules, polypeptides,and diagnostic and therapeutic methods
EP0785216B1 (en) Chomosome 13-linked breast cancer susceptibility gene BRCA2
US5837492A (en) Chromosome 13-linked breast cancer susceptibility gene
IE903024A1 (en) Cystic fibrosis gene
JP2002538789A (en) Novel human kallikrein-like gene
WO1993010228A1 (en) Dna encoding a glycine transporter and uses thereof
JP2003502041A (en) Lipocalin family proteins
US7388086B2 (en) NPHS2 gene involved in the steroid-resistant nephrotic syndrome, protein encoded by said gene and diagnostic and therapeutic uses
US20030017468A1 (en) Compositions and methods relating to lung specific genes
US7611892B2 (en) Prostate-specific or testis-specific nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods
US7189565B2 (en) Prostate-specific or testis-specific nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods
JP2003525599A (en) Methods for diagnosing, monitoring, staging, imaging and treating lung cancer with lung cancer-specific genes
WO2002000939A2 (en) Method of diagnosing, monitoring, staging, imaging and treating colon cancer
AU2001265239B2 (en) Method of diagnosing, monitoring, staging, imaging and treating colon cancer
US7867731B2 (en) HX2004-6 polypeptide expressed in cancerous cells
US6844189B1 (en) Chromosome 17P-linked prostate cancer susceptibility gene
AU2001265239A1 (en) Method of diagnosing, monitoring, staging, imaging and treating colon cancer
AU6082794A (en) Dna encoding a human betaine/gaba transporter and uses thereof
AU2006200418B2 (en) Gridlock nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods
KR100508845B1 (en) A Long QT Syndrome Gene Which Encodes KVLQT1 and Its Association with minK
WO2000012046A2 (en) Pkdl nucleic acids, polypeptides, and diagnostic and therapeutic methods
EP1661995A1 (en) Method of diagnosing, monitoring, staging, imaging and treating colon cancer
AU1464301A (en) Gridlock nucleic acid molecules, polypeptides, and diagnostic and therapeutic methods

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
SREP Specification republished
MK14 Patent ceased section 143(a) (annual fees not paid) or expired