WO2005094263A2 - Methods of determining the prognosis and treatment of subjects with colon cancer - Google Patents

Methods of determining the prognosis and treatment of subjects with colon cancer Download PDF

Info

Publication number
WO2005094263A2
WO2005094263A2 PCT/US2005/010020 US2005010020W WO2005094263A2 WO 2005094263 A2 WO2005094263 A2 WO 2005094263A2 US 2005010020 W US2005010020 W US 2005010020W WO 2005094263 A2 WO2005094263 A2 WO 2005094263A2
Authority
WO
WIPO (PCT)
Prior art keywords
pinl
ppinl
subject
determining
antibody
Prior art date
Application number
PCT/US2005/010020
Other languages
French (fr)
Other versions
WO2005094263A3 (en
Inventor
Janusz M. Sowadski
Original Assignee
Pintex Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pintex Pharmaceuticals, Inc. filed Critical Pintex Pharmaceuticals, Inc.
Publication of WO2005094263A2 publication Critical patent/WO2005094263A2/en
Publication of WO2005094263A3 publication Critical patent/WO2005094263A3/en

Links

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57419Specifically defined cancers of colon
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/99Isomerases (5.)

Definitions

  • PPIases The peptidyl-prolyl cis-trans isomerases (PPIases), or rotamases, are a family of ubiquitous enzymes that catalyze the cis/trans isomerization of the peptide bond on the n-terminal side of proline residues in proteins (Hunter, Cell 92: 141-142, 1998). PPIases are divided into three classes, cyclophilins (Cyps), FK-506 binding proteins (FKBPs) and the Pinl/parvulin class.
  • Pinl is a highly conserved protein that catalyzes the isomerization of only phosphorylated Ser/Thr-Pro bonds (Rananathan, R. et al. (1997) Cell 89:875-86; Yaffe, et al. 1997, Science 278: 1957-1960; Shen, et al. 1998,Genes Dev. 12:706-720; Lu, et al. 1999, Science 283: 1325-1328; Crenshaw, et al. 1998, ⁇ mbo J. 17:1315-1327; Lu, et al. 1999, Nature 399:784-788; Zhou, et al. 1999, Cell Mol. Life Sci. 56:788-806).
  • Pinl contains an N-terminal WW domain, which functions as a phosphorylated Ser/Thr-Pro binding module (Sudol, M. (1996) Prog. Biophys. Mol. Biol. 65:113-32).
  • This phosphorylation-dependent interaction targets Pinl to a subset of phosphorylated substrates, including Cdc25, Wee 1, Mytl, Tau-Rad4, and the C-terminal domain of RNA polymerase II large domain (Crenshaw, D.G., et al. (1998) Embo. J. 17: 1315-27; Shen, M. (1998) Genes Dev. 12:706-20; Wells, NJ. (1999) J. Cell. Sci. 1 12: 3861-71).
  • Pinl is involved in the onset and progression of many types of cell proliferative and neurodegenerative disorders (see, for example, Lu, K.P. (2003) Cancer Cell 4:175-80; Wulf, G. et al. (2003) Breast Cancer Res. 5:76-82; Ryo, A. et al. (2003) J. Cell Sci. 116: 773-83; Wulf, G.M. et al. (2001) EMBO J. 20:3459-72; Liou et al. (2003) Nature 424:556-61 ; Lu, P.J. et al. (1999)
  • ⁇ -catenin may also play a direct role in colorectal tumorigenesis because it binds the product of the tumor supressor gene APC (Rubinfeld et al. 1993 Science 262: 1731-34; Rubinfeld et al. 1996 Science 272:1023-1026; I inzler and Vogelstein 1996 Cell 87: 159-170).
  • Glycogen synthase kinase-3 ⁇ and wild-type APC regulate the level of free cytosolic ⁇ -catenin by promoting its degradation through the ubiquitin proteasome pathway (Rubinfeld et al. 1996 Science 272: 1023-1026; Aberle et al. 1997 EMBO J 16:3797-3804).
  • ⁇ -catenin When APC is mutated, as occurs in 85% of colorectal cancers, ⁇ -catenin accumulates in the cytosol and can translocate to the nucleus, where it binds transcription factors of the TCF/LEF gene family and activates the expression of target genes (Behrens et al. 1996 Nature 382:638-642; Huber et al. 1996 Mech Dev 59:3-10; Korinek et al. 1997 Science 275: 1784-1787; Morin et al. 1997 Science 275: 1787-90; Rubinfeld et al. 1997 Science 275: 1790-1792). Moreover, it was recently reported that several colon carcinoma cell lines (Korinek et al.
  • ⁇ -catenin plays a dual role, not only in the formation and maintenance of cell-cell interactions but also in the regulation of gene activity with a dominant oncogenic effect on tumorigenesis Accordingly, a need exists for modulators of Pinl, and methods of selecting and treating subjects with disorders characterized by the misexpression of Pinl and beta catenin, e.g., colon cancer.
  • the instant invention provides methods to determine which subjects in a population would benefit from treatment with a Pinl modulator.
  • the instant invention also provides, in at least one embodiment, methods for determining the prognosis of a subject with a cell proliferative disorder.
  • the invention also provides methods to monitor the efficacy of treatment of individuals with a Pinl associated disorder. Further, kits are provided to perform the methods of the invention.
  • the invention provides a method for treating a subject that has been preselected based on unphosphorylated Pinl levels by administering a Pinl modulator to the Pinl subject such that the treatment of said subject occurs.
  • the subject has high levels of cytoplasmic Pinl.
  • the subject has a Pinl associated state.
  • the Pinl associated state is a cell proliferative disorder.
  • the cell proliferative disorder is colon cancer.
  • the invention provides a method of treating a subject preselected based on their levels of Pinl, wherein said subject is selected based on the levels of phosphorylated Pinl and unphosphorylated Pinl, comprising administering a Pinl modulator to the subject such that the treatment of the subject occurs.
  • the levels of Pinl are determined by measuring the level of phosphorylated and unphosphorylated Pinl using immunohistochemistry.
  • the immunohistochemistry is performed using antibodies specific for phosphorylated and unphosphorylated Pinl.
  • the Pinl associated state is a cell proliferative disorder, e.g., cancer.
  • the invention provides a method of determining the Pinl status of a subject comprising determining the level and phosphorylation state of Pinl within a sample obtained from said subject.
  • the level of phosphorylated and unphosphorylated Pinl are determined, e.g., by immunohistochemistry.
  • the subject has a Pinl associated state.
  • the Pinl associated state is a cell proliferative disorder, e.g., cancer.
  • the cancer is colon cancer.
  • the subject is classified into a one of the following groups based on the level, location and phosphorylation state of Pinl : nPinlStr- cPinlneg, nPinlStr-cPinlpos, nPinlWeak-cPinlneg, nPinlWeak-cPinlpos, nPinlNeg- cPinlneg, or nPinlNeg-cPinlpos.
  • the invention provides a method of treating a subject having a Pinl associated state wherein the treatment is determined based on the subject's classification as nPinlStr-cPinlneg, nPinlStr-cPinlpos, nPinlWeak-cPinlneg, nPinl Weak-cPinlpos, nPinlNeg-cPinlneg, or nPinlNeg-cPinlpos.
  • the treatment comprises the administration of a Pinl modulator, and optionally, and one or more other anti-cancer treatments.
  • the invention provides a method of monitoring the efficacy of treatment of a subject by determining the levels of phosphorylated and unphosphorylated Pinl after the administration of a Pinl modulator to the subject; wherein a change in the ratio of phosphorylated to unphosphorylated Pinl is indicative of the efficacy of treatment.
  • the invention provides a kit for selecting a subject that would benefit from treatment with a Pinl modulator including a phosphorylated Pinl specific antibody and/or an unphosphorylated Pinl specific antibody, and directions for use.
  • the invention provides a method for determining the prognosis of a subject having a cell proliferative disorder by determining the levels of pPinl in a biological sample, wherein an elevated level of pPinl in the sample compared to the statistical mean of a population having a cell proliferative disorder is indicative of a good prognosis.
  • the invention provides a method for determining the prognosis of a subject having a cell proliferative disorder by determining the levels of pPinl in a biological sample, wherein a decreased level of pPinl in the sample compared to the statistical mean of a population having a cell proliferative disorder is indicative of a poor prognosis.
  • the biological sample is selected from the group consisting of colon and blood.
  • the sample is colon tissue.
  • the pPinl levels are determined using an antibody specific for pPinl .
  • the levels of pPinl are determined using a pPinl specific antibody using fluorescence in situ hybridization (FISH).
  • FISH fluorescence in situ hybridization
  • the levels of pPinl are determined using a pPinl specific antibody using immunohistochemistry (IHC).
  • the invention provides a method of determining the prognosis of a subject having a cell proliferative disorder by obtaining a first biological sample from the subject and determining the level of pPinl in the sample, then obtaining a second biological sample from the subject at a time after collection of the first biological sample and determining the level of pPinl in the sample, wherein an increase in the level of pPinl is indicative of good prognosis.
  • the invention provides a method of determining the prognosis of a subject having a cell proliferative disorder by obtaining a first biological sample from the subject and determining the level of pPinl in the sample, then obtaining a second biological sample from the subject at a time after collection of the first biological sample and determining the level of pPinl in the sample, wherein a decrease in the level of pPinl is indicative of poor prognosis.
  • the invention provides a kit for determining the prognosis of a subject with a cell proliferative disorder comprising an antibody specific for pPinl and instructions for use.
  • the antibody enclosed in the kit is a monoclonal or a polyclonal antibody.
  • the kit can contain a second antibody specific for a second cancer marker.
  • the invention provides a method for treating an individual having a cell proliferative disorder comprising determining the level of pPinl present in a biological sample, wherein the lower the level of pPinl the more aggressive the treatment of said subject with a Pinl inhibitor.
  • Figure 1 depicts the polypeptide sequence of Pinl (SEQ ID NO:l). The serine residue identified in Lu et al. ((2002) J Biol Chem. 277: 2381-4) is underlined and in bold.
  • Figure 2 depicts a survival curve based on subjects expression of pPinl. The graph depicts the fraction of subjects surviving as a function of time (in months). Subjects are divided based on nuclear Pinl levels being strong or negative.
  • Figures 3 and 4 depict the fraction of samples with nuclear and cytoplasmic Pinl staining as a function of pPinl staining, respectively.
  • Figure 5 and 6 depict the fraction of nuclear and membraneous beta catenin positive samples as a function of pPinl staining, respectively.
  • Figure 7 and 8 depict survival curves of subjects based on the expression of pPinl and membraneous and nuclear beta catenin, respectively.
  • the graph depicts the fraction of subjects surviving as a function of time (in months). Subjects are divided based on nuclear Pinl levels being strong or negative, and beta catenin levels being negative or positive.
  • Figure 9 depicts survival curves of subjects based on their expression of pPinl in the nucleus, cytoplasm only and nucleus and cytoplasm, and no expression of pPinl .
  • the graph depicts the fraction of subjects surviving as a function of time (in months).
  • biological sample includes solid and body fluid samples.
  • the biological samples of the present invention may include cells, protein or membrane extracts of cells, blood or biological fluids such as ascites fluid or brain fluid (e.g., cerebrospinal fluid).
  • biological fluids such as ascites fluid or brain fluid (e.g., cerebrospinal fluid).
  • solid biological samples include samples taken from the colon.
  • body fluid samples include samples taken from the blood, serum, cerebrospinal fluid, semen, prostate fluid, seminal fluid, urine, saliva, sputum, mucus, bone marrow, lymph, and tears.
  • Samples for use in the methods of the invention can be obtained by standard methods including venous puncture and surgical biopsy.
  • the biological sample is a colon sample obtained by needle biopsy.
  • Pinl status indicates a classification of a subject into one of a defined series of groups based on the presence and level of phosphorylated and unphosphorylated Pinl.
  • Exemplary classifications of Pinl status defined herein are: Pin lneg-pPinl Strong, Pinl neg-pPinl Weak Pinlneg-pPinlNeg, Pin lpos-pPinl Strong, Pinlpos-pPinWeak, and Pinlpos-pPinlNeg.
  • Pinl status can be represented as ratio of phosphorylated Pinl to unphosphorylated Pinl .
  • the presence and level of Pinl can be determined using art recognized techniques, e.g., immunohistochemistry using Pinl specific antibodies. Subjects can be treated for Pinl- associated states based on their Pinl status.
  • the term "nuclear Pinl” is intended to include Pinl polypeptide that is localized to the nucleus of a cell. In certain embodiments, nuclear Pinl is predominantly phosphorylated.
  • the term "cytoplasmic Pinl” is intended to include Pinl polypeptide that is localized to the cytoplasm of a cell. In certain embodiments, cytoplasmic Pinl is predominantly unphosphorylated.
  • phosphorylation state is intended to denote that the Pinl polypeptide can exist in either a phosphorylated or unphosphorylated state.
  • the phosphorylation state denotes whether the Pinl in a biological sample is phosphorylated or unphosphorylated, or the relative ratios of phosphorylated to unphosphorylated Pinl in a sample.
  • Lu et al. (2002) J Biol Chem. 277: 2381-4
  • the term "Pinl -associated state” or "Pinl associated disorder” includes disorders and states (e.g., a disease state) that are associated with the abnormal activity of Pinl .
  • This abnormal activity can be as a result of the misexpression or misregulation of the production, degradation, or regulation of Pinl, e.g., the phosphorylation/ dephosphorylation of Pinl .
  • Pinl associated disorders that are related to higher than necessary levels of Pinl can be caused by (1) an increase in the level of transcription or translation, or a decrease in the level of degradation, of Pinl such that an abnormally high amount of Pinl polypeptide is present in a cell, or (2) the amount Pinl that is present in the unphosphorylated, i.e., active form, is abnormally high due to either an increase in the dephosphorylation of Pinl or a decrease in the phosphorylation of Pinl.
  • Pinl disorders are often associated with abnormal cell growth, abnormal cell proliferation, or misexpression of Pinl (e.g., Pinl protein or nucleic acid).
  • Pinl -associated states include states resulting from an elevation in the expression of cyclin Dl and/or Pinl .
  • Pinl -associated states also include states resulting from an elevation in the phosphorylation level of c-Jun, particularly phosphorylation of c-Jun on Ser 63/73 -Pro and/or from an elevation in the level of c-Jun amino terminal kinases (J Ks) present in a cell.
  • Pinl -associated states include neoplasia, cancer, undesirable cell growth, and/or tumor growth.
  • Pinl -associated states include states caused by DNA damage, an oncogenic protein (i.e. Ha-Ras), loss of or reduced expression of a tumor suppressor (i.e. Brcal), and/or growth factors.
  • Pinl -associated state is also intended to include diseases or disorders caused by, or associated with, deregulation of genes and/or gene products involved in a biological pathway that includes Pinl and/or cyclin Dl (e.g. beta-catenin, APC or WNT). Beta-catenin, APC and WNT have been linked to cancer development as demonstrated in Biochim Biophys Acta. (2003) 1653: 1-24 and Eur J Surg Oncol. (2003) 29: 107-1 17.
  • Pinl associated states further include disorders and states associated with regulation or activity of Pinl in the brain, e.g., Alzheimer's disease, wherein the phosphorylation state of tau is influenced by the activity of Pinl.
  • the term "misexpression” includes a non-wild type pattern of gene expression. Expression as used herein includes transcriptional, post transcriptional, e.g., mRNA stability, translational, and post translational stages.
  • Misexpression includes: expression at non-wild type levels, i.e., over or under expression; a pattern of expression that differs from wild type in terms of the time or stage at which the gene is expressed, e.g., increased or decreased expression (as compared with wild type) at a predetermined developmental period or stage; a pattern of expression that differs from wild type in terms of decreased expression (as compared with wild type) in a predetermined cell type or tissue type; a pattern of expression that differs from wild type in terms of the splicing size, amino acid sequence, post-transitional modification, or biological activity of the expressed polypeptide; a pattern of expression that differs from wild type in terms of the effect of an environmental stimulus or extracellular stimulus on expression of the gene, e.g., a pattern of increased or decreased expression (as compared with wild type) in the presence of an increase or decrease in the strength of the stimulus.
  • Misexpression includes any expression from a transgenic nucleic acid. Misexpression includes the lack or non-expression of a gene or transgene, e.g., that can be caused by a deletion of all or part of the gene or its control sequences.
  • the term "carcinoma” includes malignancies of epithelial or endocrine tissues, including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostate carcinomas, endocrine system carcinomas, melanomas, choriocarcinoma, and carcinomas of the cervix, lung, head and neck, colon, and ovary.
  • carcinoma also includes carcinosarcomas, which include malignant tumors composed of carcinomatous and sarcomatous tissues.
  • adenocarcinoma includes carcinomas derived from glandular tissue or a tumor in which the tumor cells form recognizable glandular structures.
  • the therapeutic methods of the present invention can be applied to cancerous cells of mesenchymal origin, such as those producing sarcomas (e.g., fibrosarcoma, myxosarcoma, liosarcoma, chondrosarcoma, osteogenic sarcoma or chordosarcoma, angiosarcoma, endotheliosardcoma, lympangiosarcoma, synoviosarcoma or mesothelisosarcoma); leukemias and lymphomas such as granulocytic leukemia, monocytic leukemia, lymphocytic leukemia, malignant lymphoma, plasmocytoma, reticulum cell sarcoma, or Hodgkin's disease; sarcomas such as leiomysarcoma or rhabdomysarcoma, tumors of epithelial origin such as squamous cell carcinoma, basal cell carcinoma,
  • Additional cell types amenable to treatment according to the methods described herein include those giving rise to mammary carcinomas, gastrointestinal carcinoma, such as colonic carcinomas, bladder carcinoma, prostate carcinoma, and squamous cell carcinoma of the neck and head region.
  • Examples of cancers amenable to treatment according to the methods described herein include colorectal cancers, e.g., colon cancer.
  • the language "inhibiting undesirable cell growth” is intended to include the inhibition of undesirable or inappropriate cell growth.
  • the inhibition is intended to include inhibition of cell proliferation, including rapid proliferation. For example, undesirable cell growth can result in benign masses or malignant tumors.
  • Examples of benign conditions which result from inappropriate cell growth or angiogenesis are diabetic retinopathy, retrolental fibrioplasia, neovascular glaucoma, psoriasis, angiofibromas, rheumatoid arthritis, hemangiomas, Karposi's sarcoma, and other conditions or dysfunctions characterized by dysregulated endothelial cell division.
  • the language "inhibiting tumor growth” or “inhibiting neoplasia” includes the prevention of the growth of a tumor in a subject or a reduction in the growth of a preexisting tumor in a subject, or can be the inhibition of the metastasis of a tumor from one site to another.
  • tumor is intended to encompass both in vitro, e.g., in cell culture, and in vivo tumors that form in any organ or body part of the subject.
  • the tumors preferably are tumors sensitive to the Pinl -modulating compounds of the present invention.
  • cancer includes malignancies characterized by deregulated or uncontrolled cell growth, for instance carcinomas, sarcomas, leukemias, and lymphomas.
  • carcinomas for instance carcinomas, sarcomas, leukemias, and lymphomas.
  • lymphomas for instance carcinomas, sarcomas, leukemias, and lymphomas.
  • cancer and “tumor” may be used interchangeably herein.
  • the term "cancer” includes primary malignant tumors, e.g., those whose cells have not migrated to sites in the subject's body other than the site of the original tumor, and secondary malignant tumors, e.g., those arising from metastasis, the migration of tumor cells to secondary sites that are different from the site of the original tumor.
  • Pinl modulating agents may be used to treat, inhibit, and/or prevent undesirable cell growth, neoplasia, neurodegenerative diseases, and/or cancer in a subject.
  • Pinl modulating compounds may be used to inhibit Pinl activity in a subject.
  • Pinl modulating compounds may be used to inhibit cyclin Dl expression in a subject.
  • Pinl modulating compounds of the invention can be used to treat subjects having Alzheimer's disease.
  • Pinl modulators have a Kj for Pinl of less than 0.2mM, less than 0.1 m-M, less than 750 ⁇ M, less than 500 ⁇ M, less than 250 ⁇ M, less than 100 ⁇ M, less than 50 ⁇ M, less than 500 nM, less than 250nM, less than 50 n-M, less than 10 nM, less than 5 nM, or less than 2 nM.
  • the language "Pinl modulating compound” refers to compounds that modulate, e.g., inhibit, promote, or otherwise alter, the activity of Pinl .
  • Pinl modulating compounds include both Pinl agonists and antagonists.
  • the Pinl modulating compounds include compounds that interact with the peptidyl prolyl isomrease domain (PPI) and/or the WW domain of Pinl .
  • the Pinl modulating compound is substantially specific to Pinl .
  • Pinl substantially specific for Pinl
  • inhibitors of the invention that have a Kj or ICj that is at least 2, 3, 4, 5, 10, 15, or 20 times less than the K, or K d for other peptidyl prolyl isomerases, e.g., hCyP-A, hCyP-B, hCyP-C, NKCA, hFKBP-12, hFKBP-13, and hFKBP-25.
  • Examples of Pinl modulating compounds include compounds described in U.S. Provisional Application No. 60/537,171, filed January 16, 2004, entitled "Pinl- Modulating Compounds and Methods of Use Thereof; U.S. Application No.
  • the Pinl inhibiting compounds include compounds that interact with the PPI and or the WW domain of Pinl .
  • the term "preselected” is intended to mean that a subject has been identified based on their level and/or phosphorylation state of Pinl to be likely to benefit from treatment with a Pinl modulator.
  • a subject is preselected based on the levels of unphosphorylated Pinl, phosphorylated Pinl, or the relative amounts of phosphorylated and unphosphorylated Pinl.
  • prognosis is intended to mean the probable course and outcome of a disease, e.g., a Pinl associated disease. In certain embodiments, the term is intended to mean the likelihood that a subject will live longer than the average length of time that a population of subjects with a similar disease will live. In related embodiments, a subject's prognosis is indicative of the aggressiveness and course of treatment that a subject will receive.
  • Pinl is a highly conserved protein (SEQ ID NO: l) that catalyzes the isomerization of only phosphorylated Ser/Thr-Pro bonds (Rananathan, R. et al. (1997) Cell 89:875-86; Yaffe, et al. 1997, Science 278: 1957- 1960; Shen, et al. 1998,Genes Dev. 12:706-720; Lu, et al. 1999, Science 283: 1325- 1328; Crenshaw, et al. 1998, Embo J. 17: 1315-1327; Lu, et al.
  • SEQ ID NO: l highly conserved protein
  • Pinl contains an N-terminal WW domain, which functions as a phosphorylated Ser/Thr-Pro binding module (Sudol, M. (1996) Prog. Biophys. Mol. Biol. 65:113-32). This phosphorylation- dependent interaction targets Pinl to a subset of phosphorylated substrates, including
  • Pinl is dramatically misexpressed in human cancer samples and the total level or concentration of Pinl is correlated with the aggressiveness of tumors.
  • inhibition of Pinl by various approaches including Pinl antisense polynucleotides or genetic depletion, kills human and yeast dividing cells by inducing premature mitotic entry and apoptosis.
  • the invention further provides a method of determining the prognosis of a subject with colon cancer comprising determining the levels of pPinl and beta catenin in a biological sample.
  • Beta catenin is a polypeptide involved in, among other functions, the wnt pathway and cell to cell adhesion. The sequence of the beta catenin polypeptide can be found at, for example, SwissProt Accession Number P35222.
  • the invention provides a method of detecting the presence and amount of unphosphorylated Pinl and/or phosphorylated Pinl in a biological sample.
  • the invention also provides a method of determining the amount of phosphorylated Pinl relative to the amount of unphosphorylated Pinl in a sample.
  • the methods of the invention may use antibodies that recognize phosphorylated Pinl and unphosphorylated Pinl, antibodies that are specific for phosphorylated Pinl , and antibodies that are specific for unphosphorylated Pinl .
  • Antibodies that are specific for Pinl are described in US patent 6,596,848, the entire contents of which are incorporated herein by reference. Phosphorylation specific Pinl antibodies are commercially available from, for example, Cell Signaling Technology (Beverly, MA).
  • the invention also provides methods of detecting levels of Pinl in combination with other known cancer markers, e.g., beta catenin.
  • levels of known cancer markers can be determined using antibodies, or nucleic acid probes, specific for these markers and methods standard to one of skill in the art, e.g., a mouse monoclonal antibody for detecting human ⁇ -catenin is commercially available (clone 14, catalogue reference C 19220; Transduction Laboratories, Lexington, KY).
  • the phrase "antibodies specific for phosphorylated Pinl" is intended to include antibodies that preferentially bind to an antigen of Pinl that contains a phosphorylated residue.
  • Antibodies that are specific for phosphorylated Pinl bind to Pinl with at least twice the affinity that they bind to a nonspecific antigen (e.g., BSA or casein). Further, an antibody that is specific for phosphorylated Pinl has more affinity for phosphorylated Pinl than it does unphosphorylated Pinl . In certain embodiments, the antibody specific for phosphorylated Pinl binds with at least 2, 3, 4, 5, 10, 20, 50, 100, 500, or 1000 times the affinity to phosphorylated Pinl as it does unphosphorylated Pinl . In at least one embodiment, the antibody specific for phosphorylated Pinl recognizes a Pinl molecule that is phosphorylated on serine 16 of SEQ ID NO:l .
  • antibodies specific for unphosphorylated Pinl is intended to include antibodies that preferentially bind a Pinl polypeptide that is not phosphorylated. Antibodies that are specific for unphosphorylated Pinl bind to Pinl with at least twice the affinity that they bind to a nonspecific antigen (e.g., BSA or casein). Further, an antibody that is specific for unphosphorylated Pinl has more affinity for unphosphorylated Pinl than it does phosphorylated Pinl . In certain embodiments, the antibody specific for unphosphorylated Pinl binds with at least 2, 3, 4, 5, 10, 20, 50, 100, 500, or 1000 times the affinity to unphosphorylated Pinl as it does phosphorylated Pinl .
  • the antibody specific for unphosphorylated Pinl recognizes an epitope of Pinl that comprising a residue that is capable of being phosphorylated, e.g., serine 16 of SEQ ID NO: l .
  • Polyclonal antibodies are produced by immunizing animals, usually a mammal, by multiple subcutaneous or intraperitoneal injections of an immunogen (antigen) and an adjuvant as appropriate.
  • animals are typically immunized against a protein, peptide or derivative by combining about 1 ⁇ g to 1 g of protein capable of eliciting an immune response, along with an enhancing carrier preparation, such as Freund's complete adjuvant, or an aggregating agent such as alum, and injecting the composition intradermally at multiple sites.
  • Animals are later boosted with at least one subsequent administration of a lower amount, as 1/5 to 1/10 the original amount of immunogen in Freund's complete adjuvant (or other suitable adjuvant) by subcutaneous injection at multiple sites.
  • Such populations of antibody molecules are referred to as "polyclonal" because the population comprises a large set of antibodies each of which is specific for one of the many differing epitopes found in the immunogen, and each of which is characterized by a specific affinity for that epitope.
  • An epitope is the smallest determinant of antigenicity, which for a protein, comprises a peptide of six to eight residues in length (Berzofsky, J. and I. Berkower, (1993) in Paul, W., Ed., Fundamental Immunology,
  • the polyclonal antibody fraction collected from mammalian serum is isolated by well known techniques, e.g. by chromatography with an affinity matrix that selectively binds immunoglobulin molecules such as protein A, to obtain the IgG fraction.
  • the specific antibodies may be further purified by immunoaffinity chromatography using solid phase-affixed immunogen.
  • the antibody is contacted with the solid phase-affixed immunogen for a period of time sufficient for the immunogen to immunoreact with the antibody molecules to form a solid phase-affixed immunocomplex.
  • Bound antibodies are eluted from the solid phase by standard techniques, such as by use of buffers of decreasing pH or increasing ionic strength, the eluted fractions are assayed, and those containing the specific antibodies are combined.
  • "Monoclonal antibody” or “monoclonal antibody composition” as used herein refers to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.
  • Monoclonal antibodies can be prepared using a technique which provides for the production of antibody molecules by continuous growth of cells in culture. These include but are not limited to the hybridoma technique originally described by Kohler and Milstein (1975, Nature 256:495-497; see also Brown et al.
  • Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind the polypeptide of interest, e.g., using a standard ELISA assay.
  • a monoclonal antibody can be produced by the following steps. In all procedures, an animal is immunized with an antigen such as a protein (or peptide thereof) as described above for preparation of a polyclonal antibody.
  • the immunization is typically accomplished by administering the immunogen to an immunologically competent mammal in an immunologically effective amount, i.e., an amount sufficient to produce an immune response.
  • the mammal is a rodent such as a rabbit, rat or mouse.
  • the mammal is then maintained on a booster schedule for a time period sufficient for the mammal to generate high affinity antibody molecules as described.
  • a suspension of antibody-producing cells is removed from each immunized mammal secreting the desired antibody.
  • the animal e.g., mouse
  • antibody-producing lymphocytes are obtained from one or more of the lymph nodes, spleens and peripheral blood.
  • Spleen cells are preferred, and can be mechanically separated into individual cells in a physiological medium using methods well known to one of skill in the art.
  • the antibody-producing cells are immortalized by fusion to cells of a mouse myeloma line.
  • Mouse lymphocytes give a high percentage of stable fusions with mouse homologous myelomas, however rat, rabbit and frog somatic cells can also be used.
  • Spleen cells of the desired antibody-producing animals are immortalized by fusing with myeloma cells, generally in the presence of a fusing agent such as polyethylene glycol.
  • myeloma cell lines suitable as a fusion partner are used with to standard techniques, for example, the P3-NSl/l-Ag4-l , P3-x63-Ag8.653 or Sp2/O-Agl4 myeloma lines, available from the American Type Culture Collection (ATCC), Rockville, MD.
  • the fusion-product cells which include the desired hybridomas, are cultured in selective medium such as HAT medium, designed to eliminate unfused parental myeloma or lymphocyte or spleen cells.
  • Hybridoma cells are selected and are grown under limiting dilution conditions to obtain isolated clones.
  • the supernatants of each clonal hybridoma is screened for production of antibody of desired specificity and affinity, e.g., by immunoassay techniques to determine the desired antigen such as that used for immunization.
  • Monoclonal antibody is isolated from cultures of producing cells by conventional methods, such as ammonium sulfate precipitation, ion exchange chromatography, and affinity chromatography (Zola et al., Monoclonal Hybridoma Antibodies: Techniques And Applications, Hurell (ed.), pp. 51-52, CRC Press, 1982).
  • Hybridomas produced according to these methods can be propagated in culture in vitro or in vivo (in ascites fluid) using techniques well known to those with skill in the art.
  • a monoclonal antibody directed against a polypeptide of the invention can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with the polypeptide of interest.
  • a recombinant combinatorial immunoglobulin library e.g., an antibody phage display library
  • a recombinant combinatorial immunoglobulin library e.g., an antibody phage display library
  • -Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01 ; and the Stratagene SurfZAP Phage Display Kit, Catalog No. 240612).
  • examples of methods and reagents particularly amenable for use in generating and screening an antibody display library can be found in, for example, U.S. Patent No.
  • recombinant antibodies such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention.
  • Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in PCT Publication No. WO 87/02671; European Patent Application 184,187; European Patent Application 171,496; European Patent Application 173,494; PCT Publication No. WO 86/01533; U.S. Patent No.
  • Labeled antibody includes antibodies that are labeled by a detectable means and includes enzymatically, radioactively, fluorescently, chemiluminescently, and/or bioluminescently labeled antibodies.
  • an antibody can be detectably labeled is by linking the same to an enzyme.
  • This enzyme when later exposed to its substrate, will react with the substrate in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorometric or by visual means.
  • Enzymes which can be used to detectably label the Pinl -specific or a cancer associated polypeptide-specific antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-V-steroid isomerase, yeast alcohol dehydrogenase, alpha- glycerophosphate dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-VI-phosphate dehydrogenase, glucoamylase and acetylcholinesterase.
  • Detection may be accomplished using any of a variety of immunoassays. For example, by radioactively labeling an antibody, it is possible to detect the antibody through the use of radioimmune assays.
  • a description of a radioimmune assay (RIA) may be found in Laboratory Techniques and Biochemistry in Molecular Biology, by Work, T. S., et al., North Holland Publishing Company, NY (1978), with particular reference to the chapter entitled "An Introduction to Radioimmune Assay and Related Techniques" by Chard, T.
  • the radioactive isotope can be detected by such means as the use of a gamma counter or a scintillation counter or by audioradiography.
  • Isotopes which are particularly useful for the purpose of the present invention are: 3 H, 13 I I, 35 S, l C, and preferably l25 I. It is also possible to label an antibody with a fluorescent compound. When the fluorescently labeled antibody is exposed to light of the proper wave length, its presence can then be detected due to fluorescence. Among the most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerytherin, phycocyanin, allophycocyanin, o-phthaldehyde and fiuorescamine. An antibody can also be detectably labeled using fluorescence emitting metals such as 152 Eu, or others of the lanthanide series.
  • metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentaacetic acid (DTP A) or ethylenediaminetetraacetic acid (EDTA).
  • DTP A diethylenetriaminepentaacetic acid
  • EDTA ethylenediaminetetraacetic acid
  • An antibody also can be detectably labeled by coupling it to a chemiluminescent compound. The presence of the chemiluminescent-tagged antibody is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction.
  • chemiluminescent labeling compounds are luminol, luciferin, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.
  • a bioluminescent compound may be used to label an antibody of the present invention.
  • Bioluminescence is a type of chemiluminescence found in biological systems in which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence.
  • Important bioluminescent compounds for purposes of labeling are luciferin, luciferase and aequorin.
  • the invention provides methods for evaluating subjects for the level and phosphorylation state of Pinl .
  • the invention provides methods for evaluating subjects for the level on Pinl and other markers, e.g., beta catenin. These results can be used to preselect a subject for treatment with a Pinl modulator.
  • the amount of phosphorylated and unphosphorylated Pinl in a biological sample may be determined by an assay, e.g., by a radioimmunoassay, an immunoradiometric assay, enzyme immunoassay, and /or by immunohistochemistry using antibodies specific for phosphorylated Pinl and/or unphosphorylated Pinl, respectively.
  • Radioimmunoassay is a technique for detecting and measuring the concentration of an antigen using a labeled (i.e. radioactively labeled) form of the antigen.
  • radioactive labels for antigens include 3 H, 14 C, and 125 I.
  • the concentration of phosphorylated and unphosphorylated Pinl in a sample is measured by having the antigen in the sample compete with a labeled (i.e. radioactively) antigen for binding to an antibody to the antigen.
  • the labeled antigen is present in a concentration sufficient to saturate the binding sites of the antibody.
  • the concentration of antigen in the sample the lower the concentration of labeled antigen that will bind to the antibody.
  • the antigen-antibody complex must be separated from the free antigen.
  • One method for separating the antigen-antibody complex from the free antigen is by precipitating the antigen-antibody complex with an anti-isotype antiserum.
  • Another method for separating the antigen-antibody complex from the free antigen is by precipitating the antigen-antibody complex with formalin-killed S. aureus.
  • Yet another method for separating the antigen-antibody complex from the free antigen is by performing a "solid-phase radioimmunoassay" where the antibody is linked (i.e. covalently) to Sepharose beads, polystyrene wells, polyvinylchloride wells, or microtiter wells. By comparing the concentration of labeled antigen bound to antibody to a standard curve based on samples having a known concentration of antigen, the concentration of antigen in the biological sample can be determined.
  • a "Immunoradiometric assay” is an immunoassay in which the antibody reagent is radioactively labeled.
  • An IRMA requires the production of a multivalent antigen conjugate, by techniques such as conjugation to a protein e.g., rabbit serum albumin (RSA).
  • the multivalent antigen conjugate must have at least 2 antigen residues per molecule and the antigen residues must be of sufficient distance apart to allow binding by at least two antibodies to the antigen.
  • the multivalent antigen conjugate can be attached to a solid surface such as a plastic sphere.
  • Unlabeled "sample” antigen and antibody to antigen which is radioactively labeled are added to a test tube containing the multivalent antigen conjugate coated sphere. The antigen in the sample competes with the multivalent antigen conjugate for antigen antibody binding sites.
  • the unbound reactants are removed by washing and the amount of radioactivity on the solid phase is determined.
  • the amount of bound radioactive antibody is inversely proportional to the concentration of antigen in the sample.
  • the most common enzyme immunoassay is the "Enzyme-Linked Immunosorbent Assay (ELISA).”
  • ELISA Enzyme-Linked Immunosorbent Assay
  • the "Enzyme-Linked I munosorbent Assay (ELISA)” is a technique for detecting and measuring the concentration of an antigen using a labeled (i.e. enzyme linked) form of the antibody.
  • an antibody i.e. to phosphorylated and unphosphorylated Pinl
  • a solid phase i.e.
  • a labeled i.e. enzyme linked
  • a labeled is then bound to the bound-antigen (if present) forming an antibody-antigen-antibody sandwich.
  • enzymes that can be linked to the antibody are alkaline phosphatase, horseradish peroxidase, luciferase, urease, and -galactosidase.
  • the enzyme linked antibody reacts with a substrate to generate a colored reaction product that can be assayed for.
  • antibody is incubated with a sample containing phosphorylated and unphosphorylated Pinl .
  • the antigen-antibody mixture is then contacted with an antigen-coated solid phase (i.e. a microtiter plate).
  • an antigen-coated solid phase i.e. a microtiter plate.
  • a labeled (i.e. enzyme linked) secondary antibody is then added to the solid phase to determine the amount of primary antibody bound to the solid phase.
  • an "immunohistochemistry assay” a section of tissue for is tested for specific proteins by exposing the tissue to antibodies that are specific for the type of Pinl protein that is being assayed (e.g., phosphorylated and unphosphorylated Pinl.
  • the antibodies are then visualized by any of a number of methods to determine the presence and amount of the protein present. Examples of methods used to visualize antibodies are, for example, through enzymes linked to the antibodies (e.g., luciferase, alkaline phosphatase, horseradish peroxidase, or ⁇ -galactosidase), or chemical methods (e.g., DAB/Substrate chromagen). Examples of immunohistochemsitry assays are provided in the Examples.
  • nPinlStr-cPinlneg nPinl Str-cPinlpos
  • nPinlWeak-cPinlneg nPinl Weak-cPinlpos
  • nPinlNeg-cPinlneg nPinlNeg-cPinlpos
  • nPinlNeg-cPinlpos nPinlNeg-cPinlpos.
  • the data presented in the Examples indicates that the majority of phosphorylated Pinl localizes to the nucleus while the majority of unphosphorylated Pinl localizes to the cytoplasm.
  • subjects with high levels of unphosphorylated Pinl localized in the cytoplasm are preselected for treatment with a Pinl modulator.
  • the instant invention provides method of determining the prognosis of a subject with a Pinl associated disorder, e.g., a cell proliferative disorder such as .
  • the Pinl associated disorder is a type of cancer, e.g., colon cancer.
  • the instant invention provides for the determination of the prognosis of a subject by evaluating the levels of pPinl in the subject at one or more points in time.
  • the level of pPinl in a subject can be compared to the statistical mean level in a population of subjects with similar diseases and the prognosis of the subject can be determined based on the level of pPinl relative to the statistical mean.
  • the prognosis of the individual is considered poor, e.g., the subject will likely not survive for as long as the mean length of survival of the population. If the level of pPinl in a subject is higher than the mean , the prognosis of the individual is considered good, e.g., the subject will survive for as long as the mean length of survival of the population, or longer.
  • the term "statistical mean” is used herein in a manner consistent with the well- understood definitions in the art of statistics. The statistical mean can be determined by quantitating the level of pPinl in a statistically significant number of subjects and determining the mean value of that population.
  • the prognosis of an individual can be determined by evaluating the level of pPinl in biological samples isolated at different time points. If the levels of pPinl decrease from a first sample to a second sample, the prognosis of said subject is poor. If the levels of pPinl in a sample stay the same, or increase, the prognosis is good.
  • the levels of pPinl can be determined and compared with a survival curve generated with data from a statistically significant number of subjects having a similar disease. The comparison of the pPinl levels in a subject to the survival curve will determine the prognosis of a subject, i.e., the chance the subject has to survive for a given amount of time.
  • the levels of pPinl in a biological sample can be determined and used in combination with the levels of other known prognostic markers to determine the prognosis of a subject.
  • the levels of pPinl and one or more known cancer markers e.g., beta catenin
  • Exemplary cancer markers used to predict the prognosis of a subject in combination with the levels of pPinl are provided in the Examples. Survival curves are provided for subjects based on the determination of pPinl levels and membraneous or nuclear beta catenin levels.
  • these markers are exemplary and this analysis could be performed with any known cancer marker.
  • the use of pPinl and one or more additional makers allows for a more accurate determination of a subject's prognosis.
  • the prognosis of a subject as determined by the methods disclosed herein can aide in the determination of what course of treatment to provide a subject. Further, the prognosis can indicate the aggressiveness of treatment that is required.
  • subject is intended to include living organisms, e.g., prokaryotes and eukaryotes. Examples of subjects include mammals, e.g., humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals. Most preferably the subject is a human.
  • subject that would benefit from treatment with a Pinl modulator is intended to include subjects having a Pinl associated disorder wherein treatment of said subject with a Pinl modulator would alleviate, reduce or eliminate one or more symptoms of the Pinl disorder.
  • effective amount of the compound is that amount necessary or sufficient to treat or prevent a Pinl associated state, e.g.
  • an effective amount of a Pinl modulator of the invention is the amount sufficient to inhibit undesirable cell growth in a subject, i another example, an effective amount of the Pinl modulator compound is the amount sufficient to reduce the size of a pre-existing benign cell mass or malignant tumor in a subject.
  • the effective amount can vary depending on such factors as the size and weight of the subject, the type of illness, or the particular Pinl binding compound. For example, the choice of the Pinl modulator compound can affect what constitutes an "effective amount".
  • One of ordinary skill in the art would be able to study the aforementioned factors and make the determination regarding the effective amount of the Pinl modulating compound without undue experimentation.
  • a Pinl modulator compound can be administered to the subject either prior to or after the onset of a Pinl associated state. Further, several divided dosages, as well as staggered dosages, can be administered daily or sequentially, or the dose can be continuously infused, or can be a bolus injection. Further, the dosages of the Pinl modulator can be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.
  • the term "treated,” “treating” or “treatment” includes the diminishment or alleviation of at least one symptom associated or caused by the state, disorder or disease being treated. For example, treatment can be diminishment of one or several symptoms of a disorder or complete eradication of a disorder.
  • composition includes preparations suitable for administration to mammals, e.g., humans.
  • modulators are administered as pharmaceuticals to mammals, e.g., humans, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier is art recognized and includes a pharmaceutically acceptable material, composition or vehicle, suitable for administering compounds of the present invention to mammals.
  • the carriers include liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be "acceptable” in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient.
  • materials which can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer'
  • wetting agents such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.
  • antioxidants examples include: water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, ⁇ -tocopherol, and the like; and metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
  • water soluble antioxidants such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like
  • oil-soluble antioxidants such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin
  • Formulations of the present invention include those suitable for oral, nasal, topical, transdermal, buccal, sublingual, rectal, vaginal and or parenteral administration.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy.
  • the amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 per cent to about 30 per cent.
  • Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.
  • the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; humectants, such as glycerol; disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; solution retarding agents, such as paraffin; absorption accelerators, such as quaternary ammonium compounds; wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; absorbers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic
  • the pharmaceutical compositions may also comprise buffering agents.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.
  • a tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent.
  • Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets, and other solid dosage forms of the pharmaceutical compositions such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres.
  • compositions may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use.
  • These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner.
  • embedding compositions which can be used include polymeric substances and waxes.
  • the active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.
  • Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluent commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1 ,3- butylene glycol, oils (in particular, cottonseed, groundnut, com, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluent commonly used in the art, such as, for example, water or other solvents, solubilizing agents
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.
  • Suspensions in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar- agar and tragacanth, and mixtures thereof.
  • Formulations of pharmaceutical compositions for use in the methods of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.
  • Dosage forms for the topical or transdermal administration of a compound include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants.
  • the active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required.
  • the ointments, pastes, creams and gels may contain, in addition to an active compound, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.
  • Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispersing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin.
  • compositions suitable for parenteral administration comprise one or more compounds in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.
  • aqueous and nonaqueous carriers examples include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate.
  • polyols such as glycerol, propylene glycol, polyethylene glycol, and the like
  • vegetable oils such as olive oil
  • injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • Injectable depot forms are made by forming microencapsule matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled.
  • biodegradable polymers examples include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.
  • formulations may be given orally, parenterally, topically, or rectally. They are of course given by forms suitable for each administration route. For example, they are administered in tablets or capsule fomi, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administration is preferred.
  • parenteral administration and “administered parenterally” as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrastemal injection and infusion.
  • systemic administration means the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
  • These compounds may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally and topically, as by powders, ointments or drops, including buccally and sublingually.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.
  • a physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the phamiaceutical composition required.
  • the physician or veterinarian could start doses of the compounds of the invention employed in the phamiaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.
  • intravenous and subcutaneous doses of the compounds of this invention for a patient when used for the indicated analgesic effects, will range from about 0.0001 to about 100 mg per kilogram of body weight per day, more preferably from about 0.01 to about 50 mg per kg per day, and still more preferably from about 1.0 to about 100 mg per kg per day.
  • An effective amount is that amount treats an Pinl associated state.
  • the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
  • Immunohistochemistry assay was used to detect expression of phosphorylated PIN1 in tissue microarrays (TMAs). Freshly cut tissue microarray sections were used to avoid the possibility of reduced assay sensitivity due to age-dependent oxidation of stored slides. Protocol for unphosphorylated Pinl detection The following steps were taken to analyze the level of unphosphorylated Pinl in colon tissue samples.
  • Protocol for phosphorylated Pinl detection The following steps were taken to analyze the level of phosphorylated Pinl in colon tissue samples.
  • PRIMARY ANTIBODY and Visualization -dilution 1 100 (Pintex,PIN-l phospho,m-177) -incubate 2h at 37°C in moist chamber -wash 2x5 min in PBS buffer (1 :10) -place slides in LINK, reagent (Bioninylated Anti-rabbit Vector BA-1000) -wash 2x5 min in TRIS-PBS buffer
  • Figure 2 depicts the survival curves indicating that subjects positive for pPinl have a better prognosis than those negative for Pin 1.
  • uP-TNl nuclear and cytoplasmic staining was evaluated separately. In both cases, staining intensity was estimated by visual inspection in a four step scale (0, 1, 2, 3). In addition to staining intensity, the percentage of positive cells was estimated for nuclear staining. This was not done for cytoplasmatic staining as all cells showed a similar cytoplasmatic staining intensity in most cases.
  • Figures 3 and 4 depict the fraction of subjects with phosphorylated and unphosporylated Pinl staining as a function of pPinl levels.
  • Nuclear PIN1 staining was found in 55% of adenocarcinomas, the largest subgroup of colon cancers tissues. A decrease in nuclear staining intensity was significantly associated with tumor stage (p ⁇ 0.0001), but not with grade or nodal stage.
  • the group of subjects that expressed cytoplasmic pPinl (or nuclear and cytoplasmic pPinl) (n 36) showed shortened survival when compared to subjects with samples that were completely pPinl negative ( Figure 8).
  • Example 2 Analysis of pPinl Co-expression of pPinl and Beta Catenin pPinl levels were evaluated as described above. Immunohistochemistry was used to detect beta-Catenin expression in tissue microarrays. Freshly cut tissue microarray sections were used to avoid the possibility of reduced assay sensitivity due to age-dependent oxidation of stored slides. The nuclear and membranous stainings were evaluated separately. In both cases, staining intensity was estimated by visual inspection in a four step scale (0, 1, 2, 3). In addition to staining intensity, the percentage of positive cells was estimated for nuclear staining. The following thresholds were applied for beta-Catenin:
  • Beta-Catenin NUCLEAR staining negative: no staining weak: 1+ intensity or 2+ intensity in ⁇ 20% of cells moderate: 2+ intensity in > 20% but ⁇ 70% of cells or 3+ intensity in ⁇ 20% of cells strong: 2+ intensity in > 70% of cells or 3+ intensity in > 20% of cells
  • Beta-Catenin MEMBRANOUS staining negative: no staining weak: 1+ intensity or 2+ intensity in ⁇ 20% of cells moderate: 2+ intensity in > 20% but ⁇ 70% of cells or 3+ intensity in ⁇ 20% of cells strong: 2+ intensity in > 70% of cells or 3+ intensity in > 20% of cells
  • the results indicate that pPinl was associated with nuclear beta catenin expression and the loss of membraneous beta catenin (Figure 3 and 4).
  • FIG. 6 depict the survival curves of subjects based on pPinl expression and membraneous or nuclear beta catenin, respectively.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Urology & Nephrology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Medicinal Chemistry (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Investigating Or Analysing Biological Materials (AREA)

Abstract

The instant invention provides methods of selecting subjects that will benefit from treatment with a Pin 1 modulator by determining the level an phosphorylation state of Pin 1 present in a biological sample from the subject. The invention also provides methods of determining the prognosis of a subject having a cell proliferative disorder, e.g., colon cancer, by determining the level of pPin 1 in a biological sample. The invention further provides methods of treating subjects with a Pin 1 associated disorder, and methods of monitoring the efficacy of treatment.

Description

Methods of Determining the Prognosis and Treatment of Subjects with Colon Cancer Related Applications This application claims the benefit of U.S. Patent Application Serial No. 60/556,008 filed March 23, 2004, the contents of which are hereby incorporated by reference.
Background of the invention With an estimated 130,000 new cases and 58,000 deaths per year, colorectal cancer is the second leading cause of cancer death in the United States (American
Cancer Society 2001 Cancer Facts & Figures, Am. Cancer Soc, Atlanta, GA). Most of these tumors are initiated by truncating mutations of the adenomatous polyposis coli (APC) tumor suppressor (reviewed in Kinzler, K. W. & Vogelstein, B. 2002 in The Genetic Basis of Human Cancer, eds. Vogelstein, B. & -Kinzler, K. W., McGraw-Hill, New York, pp. 565-587). Among the dozen or more proteins that associate with full length APC, β-catenin seems to play an especially important role (Rubinfeld, B. et al. 1993 Science 262:1731-1734; Su, L. K. et al. 1993 Science 262: 1734-1737), as indicated by the identification of oncogenic mutations of the β-catenin gene (CTNNB1) in colorectal cancers that lack ECmutations (Morin, P. J. et al. 1997 Science 275:1787-1790; Ilyas, M. et al. 1997 Proc. Natl. Acad. Sci. USA 94: 10330-10334;
Iwao, K. et al. 1998 Cancer Res. 58:1021-1026; Sparks, A. B. et al. 1998 Cancer Res. 58: 1 130-1134). The peptidyl-prolyl cis-trans isomerases (PPIases), or rotamases, are a family of ubiquitous enzymes that catalyze the cis/trans isomerization of the peptide bond on the n-terminal side of proline residues in proteins (Hunter, Cell 92: 141-142, 1998). PPIases are divided into three classes, cyclophilins (Cyps), FK-506 binding proteins (FKBPs) and the Pinl/parvulin class. Pinl is a highly conserved protein that catalyzes the isomerization of only phosphorylated Ser/Thr-Pro bonds (Rananathan, R. et al. (1997) Cell 89:875-86; Yaffe, et al. 1997, Science 278: 1957-1960; Shen, et al. 1998,Genes Dev. 12:706-720; Lu, et al. 1999, Science 283: 1325-1328; Crenshaw, et al. 1998, Εmbo J. 17:1315-1327; Lu, et al. 1999, Nature 399:784-788; Zhou, et al. 1999, Cell Mol. Life Sci. 56:788-806). In addition, Pinl contains an N-terminal WW domain, which functions as a phosphorylated Ser/Thr-Pro binding module (Sudol, M. (1996) Prog. Biophys. Mol. Biol. 65:113-32). This phosphorylation-dependent interaction targets Pinl to a subset of phosphorylated substrates, including Cdc25, Wee 1, Mytl, Tau-Rad4, and the C-terminal domain of RNA polymerase II large domain (Crenshaw, D.G., et al. (1998) Embo. J. 17: 1315-27; Shen, M. (1998) Genes Dev. 12:706-20; Wells, NJ. (1999) J. Cell. Sci. 1 12: 3861-71). It is becoming increasingly apparent that Pinl is involved in the onset and progression of many types of cell proliferative and neurodegenerative disorders (see, for example, Lu, K.P. (2003) Cancer Cell 4:175-80; Wulf, G. et al. (2003) Breast Cancer Res. 5:76-82; Ryo, A. et al. (2003) J. Cell Sci. 116: 773-83; Wulf, G.M. et al. (2001) EMBO J. 20:3459-72; Liou et al. (2003) Nature 424:556-61 ; Lu, P.J. et al. (1999)
Nature 399:784-8). Pinl activity has also been shown to restore the conformation and function of phosphorylated tau by promoting its dephosphorylation, indicating that Pinl is involved in neurodegeneration (Liou, Y.C. et al. (2003) Nature 424:556-61). β-Catenin has recently been the subject of increasing interest because of the discovery of additional functions of this protein apart from its well-known role in cell adhesion. The implication of β-catenin in the transduction of Wingless/Wnt-dependent cell-cell signaling has been demonstrated (Cadigan and Nusse 1997 Genes Dev 11 :3286-3305). Furthermore, β-catenin may also play a direct role in colorectal tumorigenesis because it binds the product of the tumor supressor gene APC (Rubinfeld et al. 1993 Science 262: 1731-34; Rubinfeld et al. 1996 Science 272:1023-1026; I inzler and Vogelstein 1996 Cell 87: 159-170). Glycogen synthase kinase-3β and wild-type APC regulate the level of free cytosolic β-catenin by promoting its degradation through the ubiquitin proteasome pathway (Rubinfeld et al. 1996 Science 272: 1023-1026; Aberle et al. 1997 EMBO J 16:3797-3804). When APC is mutated, as occurs in 85% of colorectal cancers, β-catenin accumulates in the cytosol and can translocate to the nucleus, where it binds transcription factors of the TCF/LEF gene family and activates the expression of target genes (Behrens et al. 1996 Nature 382:638-642; Huber et al. 1996 Mech Dev 59:3-10; Korinek et al. 1997 Science 275: 1784-1787; Morin et al. 1997 Science 275: 1787-90; Rubinfeld et al. 1997 Science 275: 1790-1792). Moreover, it was recently reported that several colon carcinoma cell lines (Korinek et al. 1997 Science 275: 1784- 1787; Morin et al. 1997 Science 275: 1787-1790) as well as colorectal tumors (Iwao et al. 1998 Cancer Res 58:1021-1026) with wild-type APC present mutations affecting the NH2-terminal domain of β-catenin. All these functional data are supported by the structural organization of the protein: an NH2-terminal domain that regulates protein stability through several serine-threonine phosphorylation sites, a central domain composed of arm repeats responsible for the interactions with APC, TCF, and E- cadherin, and a COOH-terminal domain with transcriptional regulation capacity (Cadigan and Nusse 1997 Genes Dev 1 1 :3286-3305). Therefore, it is proposed that β-catenin plays a dual role, not only in the formation and maintenance of cell-cell interactions but also in the regulation of gene activity with a dominant oncogenic effect on tumorigenesis Accordingly, a need exists for modulators of Pinl, and methods of selecting and treating subjects with disorders characterized by the misexpression of Pinl and beta catenin, e.g., colon cancer.
Summary of the Invention As the mechanism by which Pinl is involved in cell proliferative disorders is elucidated, it has been determined that the level and location of Pinl is of great importance to the progression of disease. Further, it has been shown that the phosphorylation state of Pinl is important in determining the ability of Pinl to bind substrate. The phosphorylation state and levels of Pinl are important factors in deteπnining which subjects will be likely to benefit from treatment with a Pinl modulator and for determining the prognosis of subjects with a cell proliferative disorder, e.g., colon cancer. To this point, there has not been a method to determine the amount and phosphorylation state of Pinl in a biological sample. The ability of a clinician to obtain this information would be advantageous in the determination of which subjects are likely to benefit from treatment with a Pinl modulator, and in determining the prognosis of a subject with a cell proliferative disorder, e.g., cancer. Accordingly, in one aspect, the instant invention provides methods to determine which subjects in a population would benefit from treatment with a Pinl modulator. The instant invention also provides, in at least one embodiment, methods for determining the prognosis of a subject with a cell proliferative disorder. The invention also provides methods to monitor the efficacy of treatment of individuals with a Pinl associated disorder. Further, kits are provided to perform the methods of the invention. In one aspect, the invention provides a method for treating a subject that has been preselected based on unphosphorylated Pinl levels by administering a Pinl modulator to the Pinl subject such that the treatment of said subject occurs. In one embodiment, the subject has high levels of cytoplasmic Pinl. In another embodiment, the subject has a Pinl associated state. In certain embodiments, the Pinl associated state is a cell proliferative disorder. In one specific embodiment, the cell proliferative disorder is colon cancer. In one aspect, the invention provides a method of treating a subject preselected based on their levels of Pinl, wherein said subject is selected based on the levels of phosphorylated Pinl and unphosphorylated Pinl, comprising administering a Pinl modulator to the subject such that the treatment of the subject occurs. In one embodiment, the levels of Pinl are determined by measuring the level of phosphorylated and unphosphorylated Pinl using immunohistochemistry. In specific embodiments, the immunohistochemistry is performed using antibodies specific for phosphorylated and unphosphorylated Pinl. -In a related embodiment, the Pinl associated state is a cell proliferative disorder, e.g., cancer. In one aspect, the invention provides a method of determining the Pinl status of a subject comprising determining the level and phosphorylation state of Pinl within a sample obtained from said subject. In one embodiment, the level of phosphorylated and unphosphorylated Pinl are determined, e.g., by immunohistochemistry. In a related embodiment, the subject has a Pinl associated state. In a specific embodiment the Pinl associated state is a cell proliferative disorder, e.g., cancer. In a specific embodiment the cancer is colon cancer. In a related embodiment, the subject is classified into a one of the following groups based on the level, location and phosphorylation state of Pinl : nPinlStr- cPinlneg, nPinlStr-cPinlpos, nPinlWeak-cPinlneg, nPinlWeak-cPinlpos, nPinlNeg- cPinlneg, or nPinlNeg-cPinlpos. In a related embodiment, the invention provides a method of treating a subject having a Pinl associated state wherein the treatment is determined based on the subject's classification as nPinlStr-cPinlneg, nPinlStr-cPinlpos, nPinlWeak-cPinlneg, nPinl Weak-cPinlpos, nPinlNeg-cPinlneg, or nPinlNeg-cPinlpos. In related embodiments, the treatment comprises the administration of a Pinl modulator, and optionally, and one or more other anti-cancer treatments. In one aspect, the invention provides a method of monitoring the efficacy of treatment of a subject by determining the levels of phosphorylated and unphosphorylated Pinl after the administration of a Pinl modulator to the subject; wherein a change in the ratio of phosphorylated to unphosphorylated Pinl is indicative of the efficacy of treatment. In a related aspect, the invention provides a kit for selecting a subject that would benefit from treatment with a Pinl modulator including a phosphorylated Pinl specific antibody and/or an unphosphorylated Pinl specific antibody, and directions for use. In one aspect, the invention provides a method for determining the prognosis of a subject having a cell proliferative disorder by determining the levels of pPinl in a biological sample, wherein an elevated level of pPinl in the sample compared to the statistical mean of a population having a cell proliferative disorder is indicative of a good prognosis. In another aspect, the invention provides a method for determining the prognosis of a subject having a cell proliferative disorder by determining the levels of pPinl in a biological sample, wherein a decreased level of pPinl in the sample compared to the statistical mean of a population having a cell proliferative disorder is indicative of a poor prognosis. In one embodiment, the biological sample is selected from the group consisting of colon and blood. In a specific embodiment, the sample is colon tissue. In a related method, the pPinl levels are determined using an antibody specific for pPinl . In certain embodiments, the levels of pPinl are determined using a pPinl specific antibody using fluorescence in situ hybridization (FISH). In certain embodiments, the levels of pPinl are determined using a pPinl specific antibody using immunohistochemistry (IHC). In one aspect, the invention provides a method of determining the prognosis of a subject having a cell proliferative disorder by obtaining a first biological sample from the subject and determining the level of pPinl in the sample, then obtaining a second biological sample from the subject at a time after collection of the first biological sample and determining the level of pPinl in the sample, wherein an increase in the level of pPinl is indicative of good prognosis. In one aspect, the invention provides a method of determining the prognosis of a subject having a cell proliferative disorder by obtaining a first biological sample from the subject and determining the level of pPinl in the sample, then obtaining a second biological sample from the subject at a time after collection of the first biological sample and determining the level of pPinl in the sample, wherein a decrease in the level of pPinl is indicative of poor prognosis. In one aspect, the invention provides a kit for determining the prognosis of a subject with a cell proliferative disorder comprising an antibody specific for pPinl and instructions for use. In specific embodiments the antibody enclosed in the kit is a monoclonal or a polyclonal antibody. In a related embodiment, the kit can contain a second antibody specific for a second cancer marker. In one aspect, the invention provides a method for treating an individual having a cell proliferative disorder comprising determining the level of pPinl present in a biological sample, wherein the lower the level of pPinl the more aggressive the treatment of said subject with a Pinl inhibitor.
Brief Description of the Drawings
Figure 1 depicts the polypeptide sequence of Pinl (SEQ ID NO:l). The serine residue identified in Lu et al. ((2002) J Biol Chem. 277: 2381-4) is underlined and in bold. Figure 2 depicts a survival curve based on subjects expression of pPinl. The graph depicts the fraction of subjects surviving as a function of time (in months). Subjects are divided based on nuclear Pinl levels being strong or negative. Figures 3 and 4 depict the fraction of samples with nuclear and cytoplasmic Pinl staining as a function of pPinl staining, respectively. Figure 5 and 6 depict the fraction of nuclear and membraneous beta catenin positive samples as a function of pPinl staining, respectively. Figure 7 and 8 depict survival curves of subjects based on the expression of pPinl and membraneous and nuclear beta catenin, respectively. The graph depicts the fraction of subjects surviving as a function of time (in months). Subjects are divided based on nuclear Pinl levels being strong or negative, and beta catenin levels being negative or positive. Figure 9 depicts survival curves of subjects based on their expression of pPinl in the nucleus, cytoplasm only and nucleus and cytoplasm, and no expression of pPinl . The graph depicts the fraction of subjects surviving as a function of time (in months).
Detailed Description of the Invention The term "biological sample" includes solid and body fluid samples. The biological samples of the present invention may include cells, protein or membrane extracts of cells, blood or biological fluids such as ascites fluid or brain fluid (e.g., cerebrospinal fluid). Examples of solid biological samples include samples taken from the colon, Examples of "body fluid samples" include samples taken from the blood, serum, cerebrospinal fluid, semen, prostate fluid, seminal fluid, urine, saliva, sputum, mucus, bone marrow, lymph, and tears. Samples for use in the methods of the invention can be obtained by standard methods including venous puncture and surgical biopsy. In certain embodiments, the biological sample is a colon sample obtained by needle biopsy. The term "Pinl status" indicates a classification of a subject into one of a defined series of groups based on the presence and level of phosphorylated and unphosphorylated Pinl. Exemplary classifications of Pinl status defined herein are: Pin lneg-pPinl Strong, Pinl neg-pPinl Weak Pinlneg-pPinlNeg, Pin lpos-pPinl Strong, Pinlpos-pPinWeak, and Pinlpos-pPinlNeg. However, a skilled artisan could further define the categories presented herein to better define Pinl status. For example, Pinl status can be represented as ratio of phosphorylated Pinl to unphosphorylated Pinl . The presence and level of Pinl can be determined using art recognized techniques, e.g., immunohistochemistry using Pinl specific antibodies. Subjects can be treated for Pinl- associated states based on their Pinl status. The term "nuclear Pinl" is intended to include Pinl polypeptide that is localized to the nucleus of a cell. In certain embodiments, nuclear Pinl is predominantly phosphorylated. The term "cytoplasmic Pinl" is intended to include Pinl polypeptide that is localized to the cytoplasm of a cell. In certain embodiments, cytoplasmic Pinl is predominantly unphosphorylated. The term "phosphorylation state" is intended to denote that the Pinl polypeptide can exist in either a phosphorylated or unphosphorylated state. The phosphorylation state denotes whether the Pinl in a biological sample is phosphorylated or unphosphorylated, or the relative ratios of phosphorylated to unphosphorylated Pinl in a sample. For example, Lu et al. ((2002) J Biol Chem. 277: 2381-4) demonstrated the importance of the phosphorylation of serine 16 on the ability of Pinl to bind phosphorylated substrate. The term "Pinl -associated state" or "Pinl associated disorder" includes disorders and states (e.g., a disease state) that are associated with the abnormal activity of Pinl . This abnormal activity can be as a result of the misexpression or misregulation of the production, degradation, or regulation of Pinl, e.g., the phosphorylation/ dephosphorylation of Pinl . Without being bound by theory, Pinl associated disorders that are related to higher than necessary levels of Pinl can be caused by (1) an increase in the level of transcription or translation, or a decrease in the level of degradation, of Pinl such that an abnormally high amount of Pinl polypeptide is present in a cell, or (2) the amount Pinl that is present in the unphosphorylated, i.e., active form, is abnormally high due to either an increase in the dephosphorylation of Pinl or a decrease in the phosphorylation of Pinl. Pinl disorders are often associated with abnormal cell growth, abnormal cell proliferation, or misexpression of Pinl (e.g., Pinl protein or nucleic acid). Pinl -associated states include states resulting from an elevation in the expression of cyclin Dl and/or Pinl . Pinl -associated states also include states resulting from an elevation in the phosphorylation level of c-Jun, particularly phosphorylation of c-Jun on Ser63/73-Pro and/or from an elevation in the level of c-Jun amino terminal kinases (J Ks) present in a cell. Pinl -associated states include neoplasia, cancer, undesirable cell growth, and/or tumor growth. Pinl -associated states include states caused by DNA damage, an oncogenic protein (i.e. Ha-Ras), loss of or reduced expression of a tumor suppressor (i.e. Brcal), and/or growth factors. Pinl -associated state is also intended to include diseases or disorders caused by, or associated with, deregulation of genes and/or gene products involved in a biological pathway that includes Pinl and/or cyclin Dl (e.g. beta-catenin, APC or WNT). Beta-catenin, APC and WNT have been linked to cancer development as demonstrated in Biochim Biophys Acta. (2003) 1653: 1-24 and Eur J Surg Oncol. (2003) 29: 107-1 17. Pinl associated states further include disorders and states associated with regulation or activity of Pinl in the brain, e.g., Alzheimer's disease, wherein the phosphorylation state of tau is influenced by the activity of Pinl. The term "misexpression" includes a non-wild type pattern of gene expression. Expression as used herein includes transcriptional, post transcriptional, e.g., mRNA stability, translational, and post translational stages. Misexpression includes: expression at non-wild type levels, i.e., over or under expression; a pattern of expression that differs from wild type in terms of the time or stage at which the gene is expressed, e.g., increased or decreased expression (as compared with wild type) at a predetermined developmental period or stage; a pattern of expression that differs from wild type in terms of decreased expression (as compared with wild type) in a predetermined cell type or tissue type; a pattern of expression that differs from wild type in terms of the splicing size, amino acid sequence, post-transitional modification, or biological activity of the expressed polypeptide; a pattern of expression that differs from wild type in terms of the effect of an environmental stimulus or extracellular stimulus on expression of the gene, e.g., a pattern of increased or decreased expression (as compared with wild type) in the presence of an increase or decrease in the strength of the stimulus. Misexpression includes any expression from a transgenic nucleic acid. Misexpression includes the lack or non-expression of a gene or transgene, e.g., that can be caused by a deletion of all or part of the gene or its control sequences. The term "carcinoma" includes malignancies of epithelial or endocrine tissues, including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostate carcinomas, endocrine system carcinomas, melanomas, choriocarcinoma, and carcinomas of the cervix, lung, head and neck, colon, and ovary. The term "carcinoma" also includes carcinosarcomas, which include malignant tumors composed of carcinomatous and sarcomatous tissues. The term "adenocarcinoma" includes carcinomas derived from glandular tissue or a tumor in which the tumor cells form recognizable glandular structures. For example, the therapeutic methods of the present invention can be applied to cancerous cells of mesenchymal origin, such as those producing sarcomas (e.g., fibrosarcoma, myxosarcoma, liosarcoma, chondrosarcoma, osteogenic sarcoma or chordosarcoma, angiosarcoma, endotheliosardcoma, lympangiosarcoma, synoviosarcoma or mesothelisosarcoma); leukemias and lymphomas such as granulocytic leukemia, monocytic leukemia, lymphocytic leukemia, malignant lymphoma, plasmocytoma, reticulum cell sarcoma, or Hodgkin's disease; sarcomas such as leiomysarcoma or rhabdomysarcoma, tumors of epithelial origin such as squamous cell carcinoma, basal cell carcinoma, sweat gland carcinoma, sebaceous gland carcinoma, adenocarcinoma, papillary carcinoma, papillary adenocarcinoma, cystadenocarcinoma, medullary carcinoma, undifferentiated carcinoma, bronchogenic carcinoma, melanoma, renal cell carcinoma, hepatoma-liver cell carcinoma, bile duct carcinoma, cholangiocarcinoma, papillary carcinoma, transitional cell carcinoma, chorioaencinoma, semonoma, or embryonal carcinoma; and tumors of the nervous system including gioma, menigoma, medulloblastoma, schwannoma or epidymoma. Additional cell types amenable to treatment according to the methods described herein include those giving rise to mammary carcinomas, gastrointestinal carcinoma, such as colonic carcinomas, bladder carcinoma, prostate carcinoma, and squamous cell carcinoma of the neck and head region. Examples of cancers amenable to treatment according to the methods described herein include colorectal cancers, e.g., colon cancer. The language "inhibiting undesirable cell growth" is intended to include the inhibition of undesirable or inappropriate cell growth. The inhibition is intended to include inhibition of cell proliferation, including rapid proliferation. For example, undesirable cell growth can result in benign masses or malignant tumors. Examples of benign conditions which result from inappropriate cell growth or angiogenesis are diabetic retinopathy, retrolental fibrioplasia, neovascular glaucoma, psoriasis, angiofibromas, rheumatoid arthritis, hemangiomas, Karposi's sarcoma, and other conditions or dysfunctions characterized by dysregulated endothelial cell division. The language "inhibiting tumor growth" or "inhibiting neoplasia" includes the prevention of the growth of a tumor in a subject or a reduction in the growth of a preexisting tumor in a subject, or can be the inhibition of the metastasis of a tumor from one site to another. In particular, the language "tumor" is intended to encompass both in vitro, e.g., in cell culture, and in vivo tumors that form in any organ or body part of the subject. The tumors preferably are tumors sensitive to the Pinl -modulating compounds of the present invention. . The term "cancer" includes malignancies characterized by deregulated or uncontrolled cell growth, for instance carcinomas, sarcomas, leukemias, and lymphomas. The terms "cancer" and "tumor" may be used interchangeably herein. The term "cancer" includes primary malignant tumors, e.g., those whose cells have not migrated to sites in the subject's body other than the site of the original tumor, and secondary malignant tumors, e.g., those arising from metastasis, the migration of tumor cells to secondary sites that are different from the site of the original tumor. Pinl modulating agents may be used to treat, inhibit, and/or prevent undesirable cell growth, neoplasia, neurodegenerative diseases, and/or cancer in a subject. Pinl modulating compounds may be used to inhibit Pinl activity in a subject. In one embodiment, Pinl modulating compounds may be used to inhibit cyclin Dl expression in a subject. In a further embodiment, Pinl modulating compounds of the invention can be used to treat subjects having Alzheimer's disease. In certain embodiments Pinl modulators have a Kj for Pinl of less than 0.2mM, less than 0.1 m-M, less than 750 μM, less than 500 μM, less than 250 μM, less than 100 μM, less than 50 μM, less than 500 nM, less than 250nM, less than 50 n-M, less than 10 nM, less than 5 nM, or less than 2 nM. The language "Pinl modulating compound" refers to compounds that modulate, e.g., inhibit, promote, or otherwise alter, the activity of Pinl . Pinl modulating compounds include both Pinl agonists and antagonists. In certain embodiments, the Pinl modulating compounds include compounds that interact with the peptidyl prolyl isomrease domain (PPI) and/or the WW domain of Pinl . In certain embodiments, the Pinl modulating compound is substantially specific to Pinl . The phrase "substantially specific for Pinl" is intended to include inhibitors of the invention that have a Kj or ICj that is at least 2, 3, 4, 5, 10, 15, or 20 times less than the K, or Kd for other peptidyl prolyl isomerases, e.g., hCyP-A, hCyP-B, hCyP-C, NKCA, hFKBP-12, hFKBP-13, and hFKBP-25. Examples of Pinl modulating compounds include compounds described in U.S. Provisional Application No. 60/537,171, filed January 16, 2004, entitled "Pinl- Modulating Compounds and Methods of Use Thereof; U.S. Application No.
10/361 ,206 filed March 3, 2003, entitled "Pinl -Modulating Compounds and Methods of Use Thereof; U.S. Application Serial No. 10/379,410, filed March 3, 2003, entitled "Pinl -Modulating Compounds and Methods of Use Thereof; U.S. Application Serial No: 10/379,404, filed March 3, 2003, entitled "Pinl -Modulating Compounds and Methods of Use Thereof; U.S. Application Serial No. 10/379,1 15, filed on March 3, 2003; entitled "Methods for Designing Specific Inhibitors for Pinl Proline Isomerase and Pinl-Related Molecules"; U.S. Application No. 10/379,161 , filed March 3, 2003, entitled "Methods of Treating Pinl Associated Disorders"; U.S. Provisional Application No. 60/463271, entitled "PHOTOCHEMOTHERAPEUTIC COMPOUNDS FOR USE
IN TREATMENT OF PIN1 -ASSOCIATED STATES", filed April 16, 2003; and U.S.
Application No. 10/379,408, entitled "Pinl -Modulating Compounds and Methods of
Use Thereof, filed March 3, 2003. The entire contents of each of the aforementioned applications are hereby expressly incorporated herein by reference in their entireties. In certain embodiments, the Pinl inhibiting compounds include compounds that interact with the PPI and or the WW domain of Pinl . The term "preselected" is intended to mean that a subject has been identified based on their level and/or phosphorylation state of Pinl to be likely to benefit from treatment with a Pinl modulator. In certain embodiments, a subject is preselected based on the levels of unphosphorylated Pinl, phosphorylated Pinl, or the relative amounts of phosphorylated and unphosphorylated Pinl. The term "prognosis" is intended to mean the probable course and outcome of a disease, e.g., a Pinl associated disease. In certain embodiments, the term is intended to mean the likelihood that a subject will live longer than the average length of time that a population of subjects with a similar disease will live. In related embodiments, a subject's prognosis is indicative of the aggressiveness and course of treatment that a subject will receive.
Polypeptides This invention relates to methods of diagnosis, treatment and prognosis of subjects with a Pinl associated disorder. Pinl is a highly conserved protein (SEQ ID NO: l) that catalyzes the isomerization of only phosphorylated Ser/Thr-Pro bonds (Rananathan, R. et al. (1997) Cell 89:875-86; Yaffe, et al. 1997, Science 278: 1957- 1960; Shen, et al. 1998,Genes Dev. 12:706-720; Lu, et al. 1999, Science 283: 1325- 1328; Crenshaw, et al. 1998, Embo J. 17: 1315-1327; Lu, et al. 1999, Nature 399:784- 788; Zhou, et al. 1999, Cell Mol. Life Sci. 56:788-806). In addition, Pinl contains an N-terminal WW domain, which functions as a phosphorylated Ser/Thr-Pro binding module (Sudol, M. (1996) Prog. Biophys. Mol. Biol. 65:113-32). This phosphorylation- dependent interaction targets Pinl to a subset of phosphorylated substrates, including
Cdc25, Wee 1 , Mytl, Tau-Rad4, and the C-terminal domain of RNA polymerase II large domain (Crenshaw, D.G., et al. (1998) Embo. J. 17:1315-27; Shen, M. (1998) Genes Dev. 12:706-20; Wells, NJ. (1999) J. Cell. Sci. 1 12: 3861-71). The specificity of Pinl activity is essential for cell growth; depletion or mutations of Pinl cause growth arrest, affect cell cycle checkpoints and induce premature mitotic entry, mitotic arrest and apoptosis in human tumor cells, yeast or Xenopus extracts (Lu, et al. 1996, Nature 380:544-547; Winkler, et al. 200, Science 287: 1644-1647; Hani, et al. 1999. J. Biol. Chem. 274: 108-1 16). In addition, Pinl is dramatically misexpressed in human cancer samples and the total level or concentration of Pinl is correlated with the aggressiveness of tumors. Moreover, inhibition of Pinl by various approaches, including Pinl antisense polynucleotides or genetic depletion, kills human and yeast dividing cells by inducing premature mitotic entry and apoptosis. Thus, Pinl -catalyzed prolyl isomerization regulates the conformation and function of these phosphoprotein substrates and facilitates dephosphorylation because of the conformational specificity of some phosphatases. Thus, Pinl -dependent peptide bond isomerization is an important post-phosphorylation regulatory mechanism, allowing cells to turn phosphoprotein function on or off with high efficiency and specificity during temporally regulated events, including the cell cycle (Lu et al., supra). The invention further provides a method of determining the prognosis of a subject with colon cancer comprising determining the levels of pPinl and beta catenin in a biological sample. Beta catenin is a polypeptide involved in, among other functions, the wnt pathway and cell to cell adhesion. The sequence of the beta catenin polypeptide can be found at, for example, SwissProt Accession Number P35222.
Antibodies The invention provides a method of detecting the presence and amount of unphosphorylated Pinl and/or phosphorylated Pinl in a biological sample. The invention also provides a method of determining the amount of phosphorylated Pinl relative to the amount of unphosphorylated Pinl in a sample. Accordingly, the methods of the invention may use antibodies that recognize phosphorylated Pinl and unphosphorylated Pinl, antibodies that are specific for phosphorylated Pinl , and antibodies that are specific for unphosphorylated Pinl . Antibodies that are specific for Pinl are described in US patent 6,596,848, the entire contents of which are incorporated herein by reference. Phosphorylation specific Pinl antibodies are commercially available from, for example, Cell Signaling Technology (Beverly, MA). The invention also provides methods of detecting levels of Pinl in combination with other known cancer markers, e.g., beta catenin. Levels of known cancer markers can be determined using antibodies, or nucleic acid probes, specific for these markers and methods standard to one of skill in the art, e.g., a mouse monoclonal antibody for detecting human β-catenin is commercially available (clone 14, catalogue reference C 19220; Transduction Laboratories, Lexington, KY). The phrase "antibodies specific for phosphorylated Pinl" is intended to include antibodies that preferentially bind to an antigen of Pinl that contains a phosphorylated residue. Antibodies that are specific for phosphorylated Pinl bind to Pinl with at least twice the affinity that they bind to a nonspecific antigen (e.g., BSA or casein). Further, an antibody that is specific for phosphorylated Pinl has more affinity for phosphorylated Pinl than it does unphosphorylated Pinl . In certain embodiments, the antibody specific for phosphorylated Pinl binds with at least 2, 3, 4, 5, 10, 20, 50, 100, 500, or 1000 times the affinity to phosphorylated Pinl as it does unphosphorylated Pinl . In at least one embodiment, the antibody specific for phosphorylated Pinl recognizes a Pinl molecule that is phosphorylated on serine 16 of SEQ ID NO:l . The phrase "antibodies specific for unphosphorylated Pinl" is intended to include antibodies that preferentially bind a Pinl polypeptide that is not phosphorylated. Antibodies that are specific for unphosphorylated Pinl bind to Pinl with at least twice the affinity that they bind to a nonspecific antigen (e.g., BSA or casein). Further, an antibody that is specific for unphosphorylated Pinl has more affinity for unphosphorylated Pinl than it does phosphorylated Pinl . In certain embodiments, the antibody specific for unphosphorylated Pinl binds with at least 2, 3, 4, 5, 10, 20, 50, 100, 500, or 1000 times the affinity to unphosphorylated Pinl as it does phosphorylated Pinl . In at least one embodiment, the antibody specific for unphosphorylated Pinl recognizes an epitope of Pinl that comprising a residue that is capable of being phosphorylated, e.g., serine 16 of SEQ ID NO: l . Polyclonal antibodies are produced by immunizing animals, usually a mammal, by multiple subcutaneous or intraperitoneal injections of an immunogen (antigen) and an adjuvant as appropriate. As an illustrative embodiment, animals are typically immunized against a protein, peptide or derivative by combining about 1 μg to 1 g of protein capable of eliciting an immune response, along with an enhancing carrier preparation, such as Freund's complete adjuvant, or an aggregating agent such as alum, and injecting the composition intradermally at multiple sites. Animals are later boosted with at least one subsequent administration of a lower amount, as 1/5 to 1/10 the original amount of immunogen in Freund's complete adjuvant (or other suitable adjuvant) by subcutaneous injection at multiple sites. Animals are subsequently bled, serum assayed to determine the specific antibody titer, and the animals are again boosted and assayed until the titer of antibody no longer increases (i.e., plateaus). Such populations of antibody molecules are referred to as "polyclonal" because the population comprises a large set of antibodies each of which is specific for one of the many differing epitopes found in the immunogen, and each of which is characterized by a specific affinity for that epitope. An epitope is the smallest determinant of antigenicity, which for a protein, comprises a peptide of six to eight residues in length (Berzofsky, J. and I. Berkower, (1993) in Paul, W., Ed., Fundamental Immunology,
Raven Press, N.Y., p.246). Affinities range from low, e.g. 10"°" M, to high, e.g., 10'^ M. The polyclonal antibody fraction collected from mammalian serum is isolated by well known techniques, e.g. by chromatography with an affinity matrix that selectively binds immunoglobulin molecules such as protein A, to obtain the IgG fraction. To enhance the purity and specificity of the antibody, the specific antibodies may be further purified by immunoaffinity chromatography using solid phase-affixed immunogen. The antibody is contacted with the solid phase-affixed immunogen for a period of time sufficient for the immunogen to immunoreact with the antibody molecules to form a solid phase-affixed immunocomplex. Bound antibodies are eluted from the solid phase by standard techniques, such as by use of buffers of decreasing pH or increasing ionic strength, the eluted fractions are assayed, and those containing the specific antibodies are combined. "Monoclonal antibody" or "monoclonal antibody composition" as used herein refers to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope. Monoclonal antibodies can be prepared using a technique which provides for the production of antibody molecules by continuous growth of cells in culture. These include but are not limited to the hybridoma technique originally described by Kohler and Milstein (1975, Nature 256:495-497; see also Brown et al. 1981 J Immunol 127:539-46; Brown et al., 1980, J Biol Chem 255:4980-83; Yeh et al., 1976, ENΛS 76:2927-31 ; and Yeh et al., 1982, Int. J. Cancer 29:269-75) and the more recent human B cell hybridoma technique (Kozbor et al., 1983, Immunol Today 4:72), EBV-hybridoma technique (Cole et al., 1985, Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96), and trioma techniques. The technology for producing hybridomas is well known (see generally Current Protocols in Immunology, Coligan et al. ed., John Wiley & Sons, New York, 1994). Hybridoma cells producing a monoclonal antibody of the invention are detected by screening the hybridoma culture supernatants for antibodies that bind the polypeptide of interest, e.g., using a standard ELISA assay. A monoclonal antibody can be produced by the following steps. In all procedures, an animal is immunized with an antigen such as a protein (or peptide thereof) as described above for preparation of a polyclonal antibody. The immunization is typically accomplished by administering the immunogen to an immunologically competent mammal in an immunologically effective amount, i.e., an amount sufficient to produce an immune response. Preferably, the mammal is a rodent such as a rabbit, rat or mouse. The mammal is then maintained on a booster schedule for a time period sufficient for the mammal to generate high affinity antibody molecules as described. A suspension of antibody-producing cells is removed from each immunized mammal secreting the desired antibody. After a sufficient time to generate high affinity antibodies, the animal (e.g., mouse) is sacrificed and antibody-producing lymphocytes are obtained from one or more of the lymph nodes, spleens and peripheral blood. Spleen cells are preferred, and can be mechanically separated into individual cells in a physiological medium using methods well known to one of skill in the art. The antibody-producing cells are immortalized by fusion to cells of a mouse myeloma line. Mouse lymphocytes give a high percentage of stable fusions with mouse homologous myelomas, however rat, rabbit and frog somatic cells can also be used. Spleen cells of the desired antibody-producing animals are immortalized by fusing with myeloma cells, generally in the presence of a fusing agent such as polyethylene glycol. Any of a number of myeloma cell lines suitable as a fusion partner are used with to standard techniques, for example, the P3-NSl/l-Ag4-l , P3-x63-Ag8.653 or Sp2/O-Agl4 myeloma lines, available from the American Type Culture Collection (ATCC), Rockville, MD. The fusion-product cells, which include the desired hybridomas, are cultured in selective medium such as HAT medium, designed to eliminate unfused parental myeloma or lymphocyte or spleen cells. Hybridoma cells are selected and are grown under limiting dilution conditions to obtain isolated clones. The supernatants of each clonal hybridoma is screened for production of antibody of desired specificity and affinity, e.g., by immunoassay techniques to determine the desired antigen such as that used for immunization. Monoclonal antibody is isolated from cultures of producing cells by conventional methods, such as ammonium sulfate precipitation, ion exchange chromatography, and affinity chromatography (Zola et al., Monoclonal Hybridoma Antibodies: Techniques And Applications, Hurell (ed.), pp. 51-52, CRC Press, 1982). Hybridomas produced according to these methods can be propagated in culture in vitro or in vivo (in ascites fluid) using techniques well known to those with skill in the art. Alternative to preparing monoclonal antibody-secreting hybridomas, a monoclonal antibody directed against a polypeptide of the invention can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with the polypeptide of interest. -Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01 ; and the Stratagene SurfZAP Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and reagents particularly amenable for use in generating and screening an antibody display library can be found in, for example, U.S. Patent No. 5,223,409; PCT Publication No. WO 92/18619; PCT Publication No. WO 91/17271 ; PCT Publication No. WO 92/20791 ; PCT Publication No. WO 92/15679; PCT Publication No. WO 93/01288; PCT Publication No. WO 92/01047; PCT Publication No. WO 92/09690; PCT Publication No. WO 90/02809; Fuchs et al. (1991) Bio/Technology 9: 1370-1372; Hay et al. (1992) Hum. Antibod. Hybridomas 3:81-85; Huse et al. (1989) Science 246: 1275- 1281 ; Griffiths et al. (1993) EMBO J. 12:725-734. Additionally, recombinant antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention. Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art, for example using methods described in PCT Publication No. WO 87/02671; European Patent Application 184,187; European Patent Application 171,496; European Patent Application 173,494; PCT Publication No. WO 86/01533; U.S. Patent No. 4,816,567; European Patent Application 125,023; Better et al. (1988) Science 240: 1041-1043; Liu et al. (1987) Proc. Natl. Acad. Sci. USA 84:3439-3443; Liu et α/. (1987) J. Immunol. 139:3521- 3526; Sun et /. (1987) Proc. Natl. Acad. Sci. USA 84:214-218; Nishimura et al. (1987) Cancer Res. 47:999-1005; Wood et al. (1985) Nature 314:446-449; and Shaw et al. (1988) J Natl. Cancer Inst. 80: 1553-1559); Morrison (1985) Science 229: 1202-1207; Oi et al. (1986) Bio/Techniques 4:214; U.S. Patent 5,225,539; Jones et al. (1986) Nature 321. -552-525; Verhoeyan et al. (1988) Science 239:1534; and Beidler et al. (1988) J. Immunol. 141 :4053-4060. "Labeled antibody" as used herein includes antibodies that are labeled by a detectable means and includes enzymatically, radioactively, fluorescently, chemiluminescently, and/or bioluminescently labeled antibodies. One of the ways in which an antibody can be detectably labeled is by linking the same to an enzyme. This enzyme, in turn, when later exposed to its substrate, will react with the substrate in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorometric or by visual means. Enzymes which can be used to detectably label the Pinl -specific or a cancer associated polypeptide-specific antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-V-steroid isomerase, yeast alcohol dehydrogenase, alpha- glycerophosphate dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-VI-phosphate dehydrogenase, glucoamylase and acetylcholinesterase. Detection may be accomplished using any of a variety of immunoassays. For example, by radioactively labeling an antibody, it is possible to detect the antibody through the use of radioimmune assays. A description of a radioimmune assay (RIA) may be found in Laboratory Techniques and Biochemistry in Molecular Biology, by Work, T. S., et al., North Holland Publishing Company, NY (1978), with particular reference to the chapter entitled "An Introduction to Radioimmune Assay and Related Techniques" by Chard, T. The radioactive isotope can be detected by such means as the use of a gamma counter or a scintillation counter or by audioradiography. Isotopes which are particularly useful for the purpose of the present invention are: 3H, 13 II, 35S, l C, and preferably l25I. It is also possible to label an antibody with a fluorescent compound. When the fluorescently labeled antibody is exposed to light of the proper wave length, its presence can then be detected due to fluorescence. Among the most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerytherin, phycocyanin, allophycocyanin, o-phthaldehyde and fiuorescamine. An antibody can also be detectably labeled using fluorescence emitting metals such as 152Eu, or others of the lanthanide series. These metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentaacetic acid (DTP A) or ethylenediaminetetraacetic acid (EDTA). An antibody also can be detectably labeled by coupling it to a chemiluminescent compound. The presence of the chemiluminescent-tagged antibody is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction. Examples of particularly useful chemiluminescent labeling compounds are luminol, luciferin, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester. Likewise, a bioluminescent compound may be used to label an antibody of the present invention. Bioluminescence is a type of chemiluminescence found in biological systems in which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence. Important bioluminescent compounds for purposes of labeling are luciferin, luciferase and aequorin.
Methods of the Invention Selection Methods In one embodiment, the invention provides methods for evaluating subjects for the level and phosphorylation state of Pinl . In related embodiments, the invention provides methods for evaluating subjects for the level on Pinl and other markers, e.g., beta catenin. These results can be used to preselect a subject for treatment with a Pinl modulator. The amount of phosphorylated and unphosphorylated Pinl in a biological sample may be determined by an assay, e.g., by a radioimmunoassay, an immunoradiometric assay, enzyme immunoassay, and /or by immunohistochemistry using antibodies specific for phosphorylated Pinl and/or unphosphorylated Pinl, respectively. "Radioimmunoassay" is a technique for detecting and measuring the concentration of an antigen using a labeled (i.e. radioactively labeled) form of the antigen. Examples of radioactive labels for antigens include 3H, 14C, and 125I. The concentration of phosphorylated and unphosphorylated Pinl in a sample (i.e. biological sample) is measured by having the antigen in the sample compete with a labeled (i.e. radioactively) antigen for binding to an antibody to the antigen. To ensure competitive binding between the labeled antigen and the unlabeled antigen, the labeled antigen is present in a concentration sufficient to saturate the binding sites of the antibody. The higher the concentration of antigen in the sample, the lower the concentration of labeled antigen that will bind to the antibody. In a radioimmunoassay, to determine the concentration of labeled antigen bound to antibody, the antigen-antibody complex must be separated from the free antigen. One method for separating the antigen-antibody complex from the free antigen is by precipitating the antigen-antibody complex with an anti-isotype antiserum. Another method for separating the antigen-antibody complex from the free antigen is by precipitating the antigen-antibody complex with formalin-killed S. aureus. Yet another method for separating the antigen-antibody complex from the free antigen is by performing a "solid-phase radioimmunoassay" where the antibody is linked (i.e. covalently) to Sepharose beads, polystyrene wells, polyvinylchloride wells, or microtiter wells. By comparing the concentration of labeled antigen bound to antibody to a standard curve based on samples having a known concentration of antigen, the concentration of antigen in the biological sample can be determined. A "Immunoradiometric assay" (IRMA) is an immunoassay in which the antibody reagent is radioactively labeled. An IRMA requires the production of a multivalent antigen conjugate, by techniques such as conjugation to a protein e.g., rabbit serum albumin (RSA). The multivalent antigen conjugate must have at least 2 antigen residues per molecule and the antigen residues must be of sufficient distance apart to allow binding by at least two antibodies to the antigen. For example, in an IRMA the multivalent antigen conjugate can be attached to a solid surface such as a plastic sphere. Unlabeled "sample" antigen and antibody to antigen which is radioactively labeled are added to a test tube containing the multivalent antigen conjugate coated sphere. The antigen in the sample competes with the multivalent antigen conjugate for antigen antibody binding sites. After an appropriate incubation period, the unbound reactants are removed by washing and the amount of radioactivity on the solid phase is determined. The amount of bound radioactive antibody is inversely proportional to the concentration of antigen in the sample. The most common enzyme immunoassay is the "Enzyme-Linked Immunosorbent Assay (ELISA)." The "Enzyme-Linked I munosorbent Assay (ELISA)" is a technique for detecting and measuring the concentration of an antigen using a labeled (i.e. enzyme linked) form of the antibody. In a "sandwich ELISA", an antibody (i.e. to phosphorylated and unphosphorylated Pinl) is linked to a solid phase (i.e. a microtiter plate) and exposed to a biological sample containing, phosphorylated and/or unphosphorylated Pinl . The solid phase is then washed to remove unbound antigen. A labeled (i.e. enzyme linked) is then bound to the bound-antigen (if present) forming an antibody-antigen-antibody sandwich. Examples of enzymes that can be linked to the antibody are alkaline phosphatase, horseradish peroxidase, luciferase, urease, and -galactosidase. The enzyme linked antibody reacts with a substrate to generate a colored reaction product that can be assayed for. In a "competitive ELISA", antibody is incubated with a sample containing phosphorylated and unphosphorylated Pinl . The antigen-antibody mixture is then contacted with an antigen-coated solid phase (i.e. a microtiter plate). The more antigen present in the sample, the less free antibody that will be available to bind to the solid phase. A labeled (i.e. enzyme linked) secondary antibody is then added to the solid phase to determine the amount of primary antibody bound to the solid phase. In an "immunohistochemistry assay" a section of tissue for is tested for specific proteins by exposing the tissue to antibodies that are specific for the type of Pinl protein that is being assayed (e.g., phosphorylated and unphosphorylated Pinl. The antibodies are then visualized by any of a number of methods to determine the presence and amount of the protein present. Examples of methods used to visualize antibodies are, for example, through enzymes linked to the antibodies (e.g., luciferase, alkaline phosphatase, horseradish peroxidase, or β-galactosidase), or chemical methods (e.g., DAB/Substrate chromagen). Examples of immunohistochemsitry assays are provided in the Examples. Once the levels of phosphorylated and unphosphorylated Pinl in a biological sample are determined the subject can be classified based on the level and/or ratio of phosphorylated and or unphosphorylated Pinl . Exemplary classifications provided in the Examples are nPinlStr-cPinlneg, nPinl Str-cPinlpos, nPinlWeak-cPinlneg, nPinl Weak-cPinlpos, nPinlNeg-cPinlneg, and nPinlNeg-cPinlpos. nPinl is used to indicate nuclear Pinl, and cPinl is used to indicate cytoplasmic Pinl . The data presented in the Examples indicates that the majority of phosphorylated Pinl localizes to the nucleus while the majority of unphosphorylated Pinl localizes to the cytoplasm. In one embodiment, subjects with high levels of unphosphorylated Pinl localized in the cytoplasm are preselected for treatment with a Pinl modulator.
Methods of Prognosis The instant invention provides method of determining the prognosis of a subject with a Pinl associated disorder, e.g., a cell proliferative disorder such as . In certain embodiments, the Pinl associated disorder is a type of cancer, e.g., colon cancer. The instant invention provides for the determination of the prognosis of a subject by evaluating the levels of pPinl in the subject at one or more points in time. In one embodiment, the level of pPinl in a subject can be compared to the statistical mean level in a population of subjects with similar diseases and the prognosis of the subject can be determined based on the level of pPinl relative to the statistical mean. If the level of pPinl in a subject is lower than the mean, the prognosis of the individual is considered poor, e.g., the subject will likely not survive for as long as the mean length of survival of the population. If the level of pPinl in a subject is higher than the mean , the prognosis of the individual is considered good, e.g., the subject will survive for as long as the mean length of survival of the population, or longer. The term "statistical mean" is used herein in a manner consistent with the well- understood definitions in the art of statistics. The statistical mean can be determined by quantitating the level of pPinl in a statistically significant number of subjects and determining the mean value of that population. In another embodiment, the prognosis of an individual can be determined by evaluating the level of pPinl in biological samples isolated at different time points. If the levels of pPinl decrease from a first sample to a second sample, the prognosis of said subject is poor. If the levels of pPinl in a sample stay the same, or increase, the prognosis is good. The levels of pPinl can be determined and compared with a survival curve generated with data from a statistically significant number of subjects having a similar disease. The comparison of the pPinl levels in a subject to the survival curve will determine the prognosis of a subject, i.e., the chance the subject has to survive for a given amount of time. In other embodiments, the levels of pPinl in a biological sample can be determined and used in combination with the levels of other known prognostic markers to determine the prognosis of a subject. For example, the levels of pPinl and one or more known cancer markers, e.g., beta catenin, can be evaluated and together used to determine the prognosis of a subject. Exemplary cancer markers used to predict the prognosis of a subject in combination with the levels of pPinl are provided in the Examples. Survival curves are provided for subjects based on the determination of pPinl levels and membraneous or nuclear beta catenin levels. One of ordinary skill in the art would understand that these markers are exemplary and this analysis could be performed with any known cancer marker. The use of pPinl and one or more additional makers allows for a more accurate determination of a subject's prognosis. The prognosis of a subject as determined by the methods disclosed herein, can aide in the determination of what course of treatment to provide a subject. Further, the prognosis can indicate the aggressiveness of treatment that is required.
Methods of Treatment The term "subject" is intended to include living organisms, e.g., prokaryotes and eukaryotes. Examples of subjects include mammals, e.g., humans, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals. Most preferably the subject is a human. The term "subject that would benefit from treatment with a Pinl modulator" is intended to include subjects having a Pinl associated disorder wherein treatment of said subject with a Pinl modulator would alleviate, reduce or eliminate one or more symptoms of the Pinl disorder. The language "effective amount" of the compound is that amount necessary or sufficient to treat or prevent a Pinl associated state, e.g. prevent the various morphological and somatic symptoms of a Pinl associated state. In an example, an effective amount of a Pinl modulator of the invention is the amount sufficient to inhibit undesirable cell growth in a subject, i another example, an effective amount of the Pinl modulator compound is the amount sufficient to reduce the size of a pre-existing benign cell mass or malignant tumor in a subject. The effective amount can vary depending on such factors as the size and weight of the subject, the type of illness, or the particular Pinl binding compound. For example, the choice of the Pinl modulator compound can affect what constitutes an "effective amount". One of ordinary skill in the art would be able to study the aforementioned factors and make the determination regarding the effective amount of the Pinl modulating compound without undue experimentation. The regimen of administration can affect what constitutes an effective amount. A Pinl modulator compound can be administered to the subject either prior to or after the onset of a Pinl associated state. Further, several divided dosages, as well as staggered dosages, can be administered daily or sequentially, or the dose can be continuously infused, or can be a bolus injection. Further, the dosages of the Pinl modulator can be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation. The term "treated," "treating" or "treatment" includes the diminishment or alleviation of at least one symptom associated or caused by the state, disorder or disease being treated. For example, treatment can be diminishment of one or several symptoms of a disorder or complete eradication of a disorder. While it is possible for a compound of the present invention to be administered alone, it is preferable to administer the compound as a pharmaceutical composition. The language "pharmaceutical composition" includes preparations suitable for administration to mammals, e.g., humans. When the modulators are administered as pharmaceuticals to mammals, e.g., humans, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier. The phrase "pharmaceutically acceptable carrier" is art recognized and includes a pharmaceutically acceptable material, composition or vehicle, suitable for administering compounds of the present invention to mammals. The carriers include liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject agent from one organ, or portion of the body, to another organ, or portion of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol; phosphate buffer solutions; and other non-toxic compatible substances employed in pharmaceutical formulations. Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions. Examples of pharmaceutically acceptable antioxidants include: water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, α-tocopherol, and the like; and metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like. Formulations of the present invention include those suitable for oral, nasal, topical, transdermal, buccal, sublingual, rectal, vaginal and or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred per cent, this amount will range from about 1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 per cent to about 30 per cent. Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product. In solid dosage forms for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; humectants, such as glycerol; disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; solution retarding agents, such as paraffin; absorption accelerators, such as quaternary ammonium compounds; wetting agents, such as, for example, cetyl alcohol and glycerol monostearate; absorbents, such as kaolin and bentonite clay; lubricants, such a talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like. A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets, and other solid dosage forms of the pharmaceutical compositions, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients. Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluent commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1 ,3- butylene glycol, oils (in particular, cottonseed, groundnut, com, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert dilutents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents. Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar- agar and tragacanth, and mixtures thereof. Formulations of pharmaceutical compositions for use in the methods of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound. Dosage forms for the topical or transdermal administration of a compound include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers, or propellants which may be required. The ointments, pastes, creams and gels may contain, in addition to an active compound, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof. Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispersing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the active compound in a polymer matrix or gel. Pharmaceutical compositions suitable for parenteral administration comprise one or more compounds in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents. Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions for use in the methods of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants. These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin. Injectable depot forms are made by forming microencapsule matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue. For the methods of treatment of the instant invention, formulations may be given orally, parenterally, topically, or rectally. They are of course given by forms suitable for each administration route. For example, they are administered in tablets or capsule fomi, by injection, inhalation, eye lotion, ointment, suppository, etc. administration by injection, infusion or inhalation; topical by lotion or ointment; and rectal by suppositories. Oral administration is preferred. The phrases "parenteral administration" and "administered parenterally" as used herein means modes of administration other than enteral and topical administration, usually by injection, and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrastemal injection and infusion. The phrases "systemic administration," "administered systemically," "peripheral administration" and "administered peripherally" as used herein mean the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration. These compounds may be administered to humans and other animals for therapy by any suitable route of administration, including orally, nasally, as by, for example, a spray, rectally, intravaginally, parenterally, intracisternally and topically, as by powders, ointments or drops, including buccally and sublingually. Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient. The selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion of the particular compound being employed, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts. A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the phamiaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the phamiaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. In general, a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. Generally, intravenous and subcutaneous doses of the compounds of this invention for a patient, when used for the indicated analgesic effects, will range from about 0.0001 to about 100 mg per kilogram of body weight per day, more preferably from about 0.01 to about 50 mg per kg per day, and still more preferably from about 1.0 to about 100 mg per kg per day. An effective amount is that amount treats an Pinl associated state. If desired, the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
Exemplification The invention is further illustrated by the following examples which should not be construed as limiting.
Examples
Example 1 : Analysis of Phosphorylated Pinl levels in Colon Tissue
Immunostainins protocol
Immunohistochemistry assay was used to detect expression of phosphorylated PIN1 in tissue microarrays (TMAs). Freshly cut tissue microarray sections were used to avoid the possibility of reduced assay sensitivity due to age-dependent oxidation of stored slides. Protocol for unphosphorylated Pinl detection The following steps were taken to analyze the level of unphosphorylated Pinl in colon tissue samples.
SLIDE PREPARATION -Deparaffinize TMAs overnight in Xylene -descending ethanol series -rinse 5 minutes in PBS buffer PRETREATMENT
-Microwave 30 minutes at 90°C in citrate buffer pH 6 (ProTaqstura Code No.:
400300692)
-wash 5 minutes in PBS buffer
PEROXIDASE-BLOCKING
-incubate 30 min in 0,3%H2O2/Methanol
-wash 2x5 min in PBS buffer
-incubate 30 min in normal serum(Normal horse Serum, Vector S-2000)
PRIMARY ANTIBODY AND VISUALIZATION
-dilution 1 :600 (Pintex,monoclonal,m-156)
-incubate 2h at 37°C in moist chamber
-wash 2x5 min in PBS buffer (1 :10) -place slides 30 min in LINK reagent (Biotinylated Anti-mouse Vector BA-2000)
-wash 2x5 min in TRIS-PBS buffer
-incubate 30 min with ABC reagent (Vectastain ABC-Kit, Elite standard, Vector PK
6100)
-wash 2x5 min in TRIS-PBS buffer
CHROMOGEN
-cover slides for 6 min with DAB-Chromogen(Liquid DAB DAKO Code No.: K 3467)
-wash slides in PBS-Puffer thoroughly
-counterstain for 20 sec with Hamatoxylin(Harris Hamatoxylin HTX 31000, Medite GmbH)
-rinse with water
-differentiate in HCL-Ethanol
-hemalaun for 5 min in water
-ascending ethanol series -xylene
-cover
Protocol for phosphorylated Pinl detection The following steps were taken to analyze the level of phosphorylated Pinl in colon tissue samples.
SLIDE PREPARATION
-Deparaffinize TMAs overnight in Xylene
-descending ethanol series
-rinse 5 minutes in PBS buffer
PRETREATMENT
-Water bath 30 minutes at 99°C in (DAKO Target Retrieval Solution Puffer Code No.: SI 699)
-Let slides cool -wash 5 minutes in PBS buffer PEROXIDASE-BLOCKING -incubate 30 min in 0,3%H2O2/Methanol -wash 2x5 min in PBS buffer
-incubate 30 min in normal serum (Normal goat Serum, Vector S-1000)
PRIMARY ANTIBODY and Visualization -dilution 1 : 100 (Pintex,PIN-l phospho,m-177) -incubate 2h at 37°C in moist chamber -wash 2x5 min in PBS buffer (1 :10) -place slides in LINK, reagent (Bioninylated Anti-rabbit Vector BA-1000) -wash 2x5 min in TRIS-PBS buffer
-incubate with ABC reagent (Vectastain ABC-Kit, Elite standard, Vector PK 6100) -wash 2x5 min in TRIS-PBS buffer CHROMOGEN
-cover slides for 6 min with DAB-Chromogen(Liquid DAB DAKO)
-wash slides in PBS-Puffer thoroughly
-counterstain for 20 sec with Hamatoxylin(Harris Hamatoxylin HTX 31000, Medite
GmbH) -rinse with water
-differentiate in HCL-Ethanol
-hemalaun for 5 min in water
-ascending ethanol series
-xylene -cover
Materials A colon cancer tissue microarray containing 1475 colon tissue samples was analyzed. The breakdown of histopathological features is presented in the results section. A Pinl antibody that is specific for phosphorylated Pinl was used for IHC analysis to determine the level of phosphorylated Pinl .
Methods PIN1 immunostaining was analyzable in a colon tissue microarray. In a fraction of the samples analysis failed. During the analysis nuclear or cytoplasmic pPINl positivity was noted. It was further observed that only 36 samples analyzed with the phosphorylated Pinl antibody showed cytoplasmic positivity. Re- t-s Analysis of the immunostaining For consistency, a single pathologist evaluated all immunostainings. For phosphorylated Pinl staining intensity was estimated by visual inspection in a four step scale (0, 1 , 2, 3). In addition to staining intensity, the percentage of positive cells was estimated. The criteria for pPINl classification was as follows: Negative: pPESf 1 int = 0 Weak: pPINl int = 1 or pPINl int = 2 and pPINl pet <30%
Moderate: pPINl int = 2 and pPINl pet >30% but <= 70% or pPINl int = 3 and pPINl pet <30%
Strong: pPINl int = 2 and pPINl pet >70% or pPINl int = 3 and pPINl pet >30%
For analysis puφoses, the negative and weak groups were called Negative, whereas the moderate and strong groups were called Positive. Figure 2 depicts the survival curves indicating that subjects positive for pPinl have a better prognosis than those negative for Pin 1. For uP-TNl, nuclear and cytoplasmic staining was evaluated separately. In both cases, staining intensity was estimated by visual inspection in a four step scale (0, 1, 2, 3). In addition to staining intensity, the percentage of positive cells was estimated for nuclear staining. This was not done for cytoplasmatic staining as all cells showed a similar cytoplasmatic staining intensity in most cases. Based on both score components (percentage and intensity), the nuclear PIN1 stainings were classified as "negative", "weak", "moderate" or "strong". The cut-off criteria for classification was as follows: Negative: no staining Weak: 1+ intensity 2+ intensity in 10% of cells
Moderate: 2+ intensity in >10% but <70% of cells or 3+ intensity in 30% of cells
Strong: 2+ intensity in >70% of cells or 3+ intensity in >30% of cells For analysis puφoses, negative subjects were considered uPinl negative, and subjects with weak, moderate and strong uPinl levels were considered uPinl positive.
Figures 3 and 4 depict the fraction of subjects with phosphorylated and unphosporylated Pinl staining as a function of pPinl levels.
pPIN expression in relation to tumor phenotype and patient prognosis The association between pPINl expression and histological parameters is summarized in Table 1. In cancer samples, loss of pPINl expression was significantly linked to advanced cancers (higher pT and pN stages). Table 1 : Nuclear pPINl expression and tumor phenotype in colon cancers. N on TMA analyzable negative weak moderate strong p-Valυe Histology Adenocarcinoma 1261 939 44.6% 35.4% 8.5% 11.5% Medullary ca. 5 4 100.0% 0.0% 0.0% 0.0% Mucinous ca. 119 68 38.2% 39.7% 14.7% 7.4% Signet cell ca. 5 3 33.3% 0.0% 33.3% 33.3% Others 12 2 70.0% 20.0% 10.0% 0.0%
Stage ppTTll 6622 4488 37.5% 25.0% 8.3% 29.2% <0.0001 pT2 203 162 34.6% 35.8% 12.4% 17.3% pT3 899 648 44.9% 36.6% 8.8% 9.7% pT4 223 155 55.5% 31.6% 7.1% 5.8% Grade 1 31 24.0% 45.8% 37.5% 8.3% 8.3% 0.2960 2 1177 870.0% 43.8% 35.4% 8.7% 12.1% 3 177 118.0% 50.0% 33.9% 11.0% 5.1% Nodal stage pNO 711 530 44.0% 34.7% 9.3% 12.1% 0.4326 pNl 357 259 44.8% 36.3% 8.5% 10.4% pN2 294 208 49.0% 36.5% 7.7% 6.7% In 41 samples, one or more parameters were unknown. pPINl immunostaining result Nuclear PIN1 staining was found in 55% of adenocarcinomas, the largest subgroup of colon cancers tissues. A decrease in nuclear staining intensity was significantly associated with tumor stage (p<0.0001), but not with grade or nodal stage. The group of subjects that expressed cytoplasmic pPinl (or nuclear and cytoplasmic pPinl) (n=36) showed shortened survival when compared to subjects with samples that were completely pPinl negative (Figure 8).
Example 2: Analysis of pPinl Co-expression of pPinl and Beta Catenin pPinl levels were evaluated as described above. Immunohistochemistry was used to detect beta-Catenin expression in tissue microarrays. Freshly cut tissue microarray sections were used to avoid the possibility of reduced assay sensitivity due to age-dependent oxidation of stored slides. The nuclear and membranous stainings were evaluated separately. In both cases, staining intensity was estimated by visual inspection in a four step scale (0, 1, 2, 3). In addition to staining intensity, the percentage of positive cells was estimated for nuclear staining. The following thresholds were applied for beta-Catenin:
Beta-Catenin NUCLEAR staining: negative: no staining weak: 1+ intensity or 2+ intensity in < 20% of cells moderate: 2+ intensity in > 20% but < 70% of cells or 3+ intensity in < 20% of cells strong: 2+ intensity in > 70% of cells or 3+ intensity in > 20% of cells Beta-Catenin MEMBRANOUS staining: negative: no staining weak: 1+ intensity or 2+ intensity in < 20% of cells moderate: 2+ intensity in > 20% but ≤ 70% of cells or 3+ intensity in < 20% of cells strong: 2+ intensity in > 70% of cells or 3+ intensity in > 20% of cells The results indicate that pPinl was associated with nuclear beta catenin expression and the loss of membraneous beta catenin (Figure 3 and 4). Survival analysis indicates that pPinl is a better prognostic marker than beta catenin in subjects with colon cancer, although the combination of both markers is beneficial in further defining the prognosis of a subject. Figures 6 and 7 depict the survival curves of subjects based on pPinl expression and membraneous or nuclear beta catenin, respectively.
Equivalents Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

What is claimed is:
1. A method for determining the prognosis of a subject having a cell proliferative disorder comprising the steps of: determining the levels of pPinl in a biological sample from the colon; wherein an elevated level of pPinl in the sample compared to the statistical mean of a population having a cell proliferative disorder is indicative of a good prognosis.
2. A method for determining the prognosis of a subject having a cell proliferative disorder comprising the steps of: determining the levels of pPinl in a biological sample from the colon; wherein an decreased level of pPinl in the sample compared to the statistical mean of a population having a cell proliferative disorder is indicative of a poor prognosis.
3. The method of claim 1 or 2, wherein levels are determined using an antibody specific for pPinl .
4. The method of claim 3, wherein said levels of pPinl are determined using FISH.
5. The method of claim 3, wherein said levels of pPinl are determined using IHC.
6. A method of determining the prognosis of a subject having a cell proliferative disorder comprising: obtaining a first biological sample from said subject and determining the level of pPinl in said sample; obtaining a second biological sample from said subject at a time after collection of said first biological sample and determining the level of pPinl in said sample; wherein an increase in the level of pPinl is indicative of good prognosis.
7. A method of determining the prognosis of a subject having a cell 5 proliferative disorder comprising: obtaining a first biological sample from said subject and determining the level of pPinl in said sample; obtaining a second biological sample from said subject at a time after collection of said first biological sample and determining the level of 10 pPinl in said sample; wherein a decrease in the level of pPinl is indicative of poor prognosis.
8. The method of claim 6 or 7 wherein said biological sample is from the colon of said subject.
15 9. A kit for determining the prognosis of a subject with colon cancer comprising an antibody specific for pPinl and instructions for use.
10. The kit of claim 9, wherein said antibody is a monoclonal antibody.
>0 1 1. The kit of claim 10, wherein said antibody is a polyclonal antibody.
12. The kit of claim 10, further comprising a second antibody specific for a second cancer marker.
>5 13. The kit of claim 12, wherein said antibody is specific for beta catenin.
14. A method for determining the course of treatment for a subject having a cell proliferative disorder comprising determining the level of pPinl in a 50 biological sample from said subject, wherein the lower the level of pPinl the more aggressive the treatment of said subject with a anticancer agent.
15. The method of claim 14, wherein said cell proliferative disorder is colon cancer.
16. The method of claim 15, wherein said anticancer agent is a Pinl inhibitor.
17. A method of determining if a subject has a cell proliferative disorder comprising the steps of: Obtaining a biological sample from said subject; Determining the level of pPinl in said sample; Thereby determining if said subject has cancer.
18. The method of claim 17, wherein said cell proliferative disorder is colon cancer.
19. The method of claim 17, wherein the level of beta catenin is determined.
PCT/US2005/010020 2004-03-23 2005-03-23 Methods of determining the prognosis and treatment of subjects with colon cancer WO2005094263A2 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US55600804P 2004-03-23 2004-03-23
US60/556,008 2004-03-23

Publications (2)

Publication Number Publication Date
WO2005094263A2 true WO2005094263A2 (en) 2005-10-13
WO2005094263A3 WO2005094263A3 (en) 2006-11-09

Family

ID=35064247

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/010020 WO2005094263A2 (en) 2004-03-23 2005-03-23 Methods of determining the prognosis and treatment of subjects with colon cancer

Country Status (1)

Country Link
WO (1) WO2005094263A2 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2369017A1 (en) * 2006-07-13 2011-09-28 The Ohio State University Research Foundation Micro-RNA-based methods and compositions for the diagnosis and treatment of colon related diseases
US8859202B2 (en) 2012-01-20 2014-10-14 The Ohio State University Breast cancer biomarker signatures for invasiveness and prognosis
US8865885B2 (en) 2006-03-20 2014-10-21 The Ohio State University Research Foundation MicroRNA fingerprints during human megakaryocytopoiesis
US8916533B2 (en) 2009-11-23 2014-12-23 The Ohio State University Materials and methods useful for affecting tumor cell growth, migration and invasion
US8946187B2 (en) 2010-11-12 2015-02-03 The Ohio State University Materials and methods related to microRNA-21, mismatch repair, and colorectal cancer
US9017940B2 (en) 2006-01-05 2015-04-28 The Ohio State University Methods for diagnosing colon cancer using MicroRNA signatures
US9085804B2 (en) 2007-08-03 2015-07-21 The Ohio State University Research Foundation Ultraconserved regions encoding ncRNAs
US9249468B2 (en) 2011-10-14 2016-02-02 The Ohio State University Methods and materials related to ovarian cancer
US9481885B2 (en) 2011-12-13 2016-11-01 Ohio State Innovation Foundation Methods and compositions related to miR-21 and miR-29a, exosome inhibition, and cancer metastasis
US10758619B2 (en) 2010-11-15 2020-09-01 The Ohio State University Controlled release mucoadhesive systems

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020025521A1 (en) * 1999-11-29 2002-02-28 Lu Kun Ping Pin1 as a marker for abnormal cell growth
US20030064384A1 (en) * 2001-04-02 2003-04-03 Mien-Chie Hung Beta-catenin is a strong and independent prognostic factor for cancer

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020025521A1 (en) * 1999-11-29 2002-02-28 Lu Kun Ping Pin1 as a marker for abnormal cell growth
US20030064384A1 (en) * 2001-04-02 2003-04-03 Mien-Chie Hung Beta-catenin is a strong and independent prognostic factor for cancer

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
RYO ET AL.: 'Prolyl isomerase Pin 1: a catalyst for oncogenesis and a potential therapeutic target in cancer' J. OF CELL SCIENCE vol. 116, 2003, pages 773 - 783 *
WULF ET AL.: 'Pin1 is overexpressed in breast cancer and cooperates with Ras signaling in increasing the transcritional activity of c-Jun towards cyclin D1' THE EMBO JOURNAL vol. 20, no. 13, 2001, pages 3459 - 3472 *

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9017940B2 (en) 2006-01-05 2015-04-28 The Ohio State University Methods for diagnosing colon cancer using MicroRNA signatures
US9017939B2 (en) 2006-01-05 2015-04-28 The Ohio State University Methods for diagnosing breast, colon, lung, pancreatic and prostate cancer using miR-21 and miR-17-5p
US8865885B2 (en) 2006-03-20 2014-10-21 The Ohio State University Research Foundation MicroRNA fingerprints during human megakaryocytopoiesis
EP2369017A1 (en) * 2006-07-13 2011-09-28 The Ohio State University Research Foundation Micro-RNA-based methods and compositions for the diagnosis and treatment of colon related diseases
US8084199B2 (en) 2006-07-13 2011-12-27 The Ohio State University Research Foundation Method of diagnosing poor survival prognosis colon cancer using microRNA-21
US9085804B2 (en) 2007-08-03 2015-07-21 The Ohio State University Research Foundation Ultraconserved regions encoding ncRNAs
US8916533B2 (en) 2009-11-23 2014-12-23 The Ohio State University Materials and methods useful for affecting tumor cell growth, migration and invasion
US8946187B2 (en) 2010-11-12 2015-02-03 The Ohio State University Materials and methods related to microRNA-21, mismatch repair, and colorectal cancer
US10758619B2 (en) 2010-11-15 2020-09-01 The Ohio State University Controlled release mucoadhesive systems
US11679157B2 (en) 2010-11-15 2023-06-20 The Ohio State University Controlled release mucoadhesive systems
US9249468B2 (en) 2011-10-14 2016-02-02 The Ohio State University Methods and materials related to ovarian cancer
US9481885B2 (en) 2011-12-13 2016-11-01 Ohio State Innovation Foundation Methods and compositions related to miR-21 and miR-29a, exosome inhibition, and cancer metastasis
US8859202B2 (en) 2012-01-20 2014-10-14 The Ohio State University Breast cancer biomarker signatures for invasiveness and prognosis
US9434995B2 (en) 2012-01-20 2016-09-06 The Ohio State University Breast cancer biomarker signatures for invasiveness and prognosis

Also Published As

Publication number Publication date
WO2005094263A3 (en) 2006-11-09

Similar Documents

Publication Publication Date Title
WO2005094263A2 (en) Methods of determining the prognosis and treatment of subjects with colon cancer
US10273308B2 (en) Methods of producing antibodies specific for p95
US7811774B2 (en) Reagents and methods for use in cancer diagnosis, classification and therapy
US20060074222A1 (en) Determination of a phosphorylation site in PPIase domain of Pin1 and uses therefor
WO2006015079A2 (en) Erm family binding agents and their use in diagnosis and treatment of proliferative conditions
US9297812B2 (en) Means and methods for diagnosing cancer using an antibody which specifically binds to BRAF V600E
JP2010145410A (en) Method and composition relevant to shc protein for predicting cancers of breast, prostate and ovary
US20210018504A1 (en) Shon as a prognostic biomarker for cancer and as a predictor of response to endocrine therapy
US20030068626A1 (en) Pin1 as a marker for abnormal cell growth
WO2005107803A2 (en) Methods of determining the prognosis and treatment of subjects with lung cancer
WO2003087760A2 (en) Methods for detecting bcr-abl signaling activity in tissues using phospho- specific antibodies
US20050214301A1 (en) Antibodies specific for BCR-ABL fusion protein and uses thereof
US20050239095A1 (en) Use of Pin1 inhibitors for treatment of cancer
EP1172654B1 (en) Diagnostic method based on the detection of the L1 adhesion molecule for ovarian and endometrial tumors
CA2408297A1 (en) Cancer diagnosis and assays for screening anti-cancer agents
WO2005076958A2 (en) Methods of determining the prognosis and treatment of subjects with pin 1 associated disorders
US7501255B2 (en) Levels of Pin1 in normal and cancerous tissue
US20120107837A1 (en) Method for screening anticancer substances, set or kit for implementing said method
US20080131916A1 (en) Reagents and Methods For Use In Cancer Diagnosis, Classification and Therapy
WO2002022862A2 (en) Methods for identifying modulators of protein interactions
JP2001302691A (en) Antibody against human kgf
EP1884522A1 (en) Antibody binding to a polo-like kinase 1 phosphorylation site of the human cohesin Wapl
JPWO2007037532A1 (en) Therapeutic or diagnostic use of SRMS gene
WO2012024643A2 (en) MST1/STK4 PHOSPHO-THREONINE 120 (pMST-T120) ANTIBODY

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SM SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

NENP Non-entry into the national phase in:

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

121 Ep: the epo has been informed by wipo that ep was designated in this application
32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: RULE 69 1 EPC

122 Ep: pct application non-entry in european phase

Ref document number: 05726138

Country of ref document: EP

Kind code of ref document: A2