WO2005095979A1 - Use of protein proc as a marker for colorectal cancer - Google Patents

Use of protein proc as a marker for colorectal cancer Download PDF

Info

Publication number
WO2005095979A1
WO2005095979A1 PCT/EP2005/003264 EP2005003264W WO2005095979A1 WO 2005095979 A1 WO2005095979 A1 WO 2005095979A1 EP 2005003264 W EP2005003264 W EP 2005003264W WO 2005095979 A1 WO2005095979 A1 WO 2005095979A1
Authority
WO
WIPO (PCT)
Prior art keywords
proc
colorectal cancer
diagnosis
protein
sample
Prior art date
Application number
PCT/EP2005/003264
Other languages
French (fr)
Inventor
Michael Tacke
Johannes Auer
Ursula Brehm
Johann Karl
Stefan Palme
Wolfgang Rollinger
Werner Zolg
Original Assignee
Roche Diagnostics Gmbh
F. Hoffmann-La Roche Ag
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 Roche Diagnostics Gmbh, F. Hoffmann-La Roche Ag filed Critical Roche Diagnostics Gmbh
Publication of WO2005095979A1 publication Critical patent/WO2005095979A1/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
    • 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
    • G01N33/573Immunoassay; Biospecific binding assay; Materials therefor for enzymes or isoenzymes

Definitions

  • CRC colorectal cancer
  • the earlier cancer can be detected/diagnosed, the better is the overall survival rate.
  • More than one third of the patients will die from progressive disease within five years after diagnosis, corresponding to a survival rate of about 40% for five years.
  • CRC colorectal cancer
  • WO 01/96390 shall be mentioned and discussed.
  • This application describes and claims more than two hundred isolated polynucleotides and the corresponding polypeptides as such, as well as their use in the detection of CRC.
  • differences on the level of mRNA are not mirrored by the level of the corresponding proteins.
  • a protein encoded by a rare mRNA may be found in very high amounts and a protein encoded by an abundant mRNA may nonetheless be hard to detect and find at all.
  • This lack of correlation between mRNA-level and protein level is due to reasons like mRNA stability, efficiency of translation, stability of the protein, etc.
  • WO 02/078636 reports about nine colorectal cancer- associated spots as found by surface-enhanced laser desorption and ionization (SELDI). These spots are seen more frequently in sera obtained from patients with CRC as compared to sera obtained from healthy controls. However, the identity of the molecule(s) comprised in such spot, e.g., its (their sequence), is not known.
  • a new diagnostic marker as a single marker should be at least as good as the best single marker known in the art. Or, a new marker should lead to a progress in diagnostic sensitivity and/or specificity either if used alone or in combination with one or more other markers, respectively.
  • the diagnostic sensitivity and/or specificity of a test is best assessed by its receiver-operating characteristics, which will be described in detail below.
  • CEA carcinoembryonic antigen
  • a tumor-associated glycoprotein a tumor-associated glycoprotein
  • serum CEA determination possesses neither the sensitivity nor the specificity to enable its use as a screening test for colorectal cancer in the asymptomatic population (Reynoso, G., et al., JAMA 220 (1972) 361- 365; Sturgeon, C, Clinical Chemistry 48 (2002) 1151-1159).
  • CRC from blood. It is especially important to improve the early diagnosis of CRC, since for patients diagnosed early on chances of survival are much higher as compared to those diagnosed at a progressed stage of disease.
  • the present invention therefore relates to a method for the diagnosis of colorectal cancer comprising the steps of a) providing a- liquid sample obtained from an individual, b) contacting said sample with a specific binding agent for PROC under conditions appropriate for formation of a complex between said binding agent and PROC, and c) correlating the amount of complex formed in (b) to the diagnosis of colorectal cancer.
  • Another preferred embodiment of the invention is a method for the diagnosis of colorectal cancer comprising the steps of a) c ⁇ -ntacting a liquid sample obtained from an individual with a specific binding agent for PROC under conditions appropriate for formation of a complex between said binding agent and PROC, and b) correlating the amount of complex formed in (a) to the diagnosis of colorectal cancer.
  • any sucti diagnosis is made in vitro.
  • the patient sample is discarded afterwards.
  • the patient sample is solely used for the in vitro diagnostic method of the invention and tib.e material of the patient sample is not transferred back into the patient's body.
  • the sample is a liquid sample.
  • the protein PROC (Pyrroline-5-carboxylate reductase, also known as PYCR1;
  • PROC catalyzes the NAD(P)H-dependent conversion of pyrroline-5 -carboxylate to proline.
  • Merrill, M. J., et al., J. Biol. Chem. 264 (1989) 9352-9358 studied the properties of human erythrocyte pyrroline-5-carboxylate reductase. They concluded that in addition to the traditional role of catalyzing the obligatory and final unidirectional step in pyrroline biosynthesis, the enzyme may play a physiologic role in the generation of NADP(+) in some cell types including human erythrocytes.
  • Physiological or artificial fragments of PROC, secondary modifications of PROC-, as well as allelic variants of PROC are also encompassed by the present invention.
  • Artificial fragments preferably encompass a peptide produced synthetically or by recombinant techniques, which at least comprises one epitope of diagnostic interest consisting of at least 6 contiguous amino acids as derived from the sequence disclosed in SEQ ID NO:l. Such fragment may advantageously be used for generation of antibodies or as a standard in an immunoassay. More preferred the artificial fragment comprises at least two epitopes of interest appropriate for setting ULp a sandwich immunoassay.
  • novel marker PROC may be used for monitoring as well as for screening purposes.
  • the diagnostic method according to the present invention may help to assess tumor load, efficacy of treatment and tumor recurrence in the follow-up of patients.
  • Increased levels of PROC are directly correlated to tumor burden.
  • a skiort term (few hours to 14 days) increase in PROC may serve as an indicator of tumor cell death.
  • an in rease of PROC can be used as an indicator for tumor recurrence.
  • the diagnostic method according to the present invention is used for screening purposes. I.e., it is used to assess subjects without a prior diagnosis of CRC by measuring the level of PI OC and correlating the level measured to the presence or absence of CRC.
  • the staging of cancer is the classification of the disease in terms of extent, progression, and severity. It groups cancer patients so that generalizations can be made about prognosis and the choice of therapy.
  • TNM the most widely used classification of the anatomical extent of cancer. It represents an internationally accepted, uniform staging system. There are three basic variables: T (the extent of the primary tumor), N (the status of regional lymph nodes) and M (the presence or absence of distant metastases).
  • TNM criteria are published by the UICC (International Union against Cancer),
  • early diagnosis of CRC refers to a diagnosis at a pre-malignant state (adenoma) or at a tumor stage where no metastases at all (neither proximal nor distal), i.e., adenoma, T ⁇ s , NI0, M0 or Tl-4; NO; M0 are present.
  • T ls denotes carcinoma in situ.
  • the diagnostic method according to the present invention is biased on a liquid sample which is derived from an individual. Unlike to methods known from the art PROC is specifically measured from this liquid sample by use of -a specific binding agent.
  • a specific binding agent is, e.g., a receptor for PROC, a lectin binding to PROC or an antibody to PROC.
  • a specific binding agent has at least an affinity of 10 7 1/mol for its corresponding target molecule.
  • the specific binding agent preferably has an affinity of 10 8 1/mol or even more preferred of 10 9 1/mol for its ta-rget molecule.
  • specific is used to indicate that other biomolecules present in the sample do not significantly bind to with the binding agent specific for PROC.
  • the level of binding to a b>iomolecule other than the target molecule results in a binding affinity which is only 10%, more preferably only 5% of the affinity of the target molecule or less.
  • a most preferred specific binding agent will fulfill both the above minimum criteria for affinity as well as for specificity.
  • a specific binding agent preferably is an antibody reactive with PROC.
  • the term antibody refers to a polyclonal antibody, a monoclonal antibody, fragments of such antibodies, as well as to genetic constructs comprising the binding domain of an antibody.
  • Antibodies are generated by state of the art procedures, e. g., as described in
  • monoclonal antibodies can be produced in any amo ⁇ -nt required with constant properties, they represent ideal tools in development of an assay for clinical routine.
  • the generation and use of monoclonal antibodies to PROC in a method according to the present invention is yet another preferred embodiment.
  • PROC has been identified as a marker which is useful in the diagnosis of CRC
  • alternative ways may be used to reach a result comparable to the achievements of the present invention.
  • alternative strategies to generate antibodies may be used.
  • Such strategies comprise amongst others the use of synthetic peptides, representing an epitope of PROC for immunization.
  • DNA Immunization also known as DNA vaccination may be used.
  • the liquid sample obtained from an individual is incubated with the specific binding agent for PROC under conditions appropriate for formation of a binding agent PROC-complex.
  • Such conditions need not be specifie d, since the skilled artisan without any inventive effort can easily identify such appropriate incubation conditions.
  • the amount of complex is measured and correlated to the diagnosis of CRC.
  • the skilled artisan will appreciate there are numerous methods to measure the amount of the specific binding agent PROC-complex all described in detail in relevant textbooks (cf., e.g., Tijssen P., supra, or Diamandis, et al., eds. (1996) Immunoassay, Academic Press, Boston).
  • PROC is detected in a sandwich type assay format.
  • a first specific binding agent is used to capture PROC on the one side an-d a second specific binding agent, which is labeled to be directly or indirectiy detectable is used on the other side.
  • a second specific binding agent which is labeled to be directly or indirectiy detectable is used on the other side.
  • PROC can be measured from a liquid sample obtained from an individual sample. No tissue and no biopsy sample is required to apply the marker PROC in the diagnosis of CRC.
  • the method according to the present invention is practiced with serum as liquid sample material.
  • the method according to the present invention is practiced with plasma as liquid sample material.
  • the method according to the present invention is practiced with whole blood as liquid sample material.
  • stool can be prepared in various ways known to the skilled artisan to result in a liquid sample as well.
  • sample liquid derived from stool also represents a preferred embodiment according to the present invention.
  • PROC in a bodily fluid sample Even more surprising they have been able to demonstrate that the presence of PROC in such liquid sample obtained from an individual can be correlated to the diagnosis of colorectal cancer.
  • Antibodies to PROC with great advantage can be used in established procedures, e.g., to detect colorectal cancer cells in situ, in biopsies, or in immu-r-ohistological procedures.
  • an antibody to PROC is used in a qualitative (PROC pres ent or absent) or quantitative (PROC amount is determined) immunoassay.
  • the present in ention relates to use of protein PROC as a marker molecule in the diagnosis of colorectal cancer from a liquid sample obtained from an individual.
  • the term marker molecule is used to indicate that an increased level of the analyte PROC as measured from a bodily fluid of an individual marks the presence of CRC.
  • novel marker PROC in the early diagnosis of colorectal cancer.
  • the use of protein PROC itself represents a significant progress to the challenging field of CRC diagnosis. Combining measurements of PROC with other known markers, like CEA, or with other markers of CRC yet to be discovered, leads to further improvements. Therefore in a further preferred embodiment the present invention relates to the use of PROC as a marker molecule for colorectal cancer in combination with one or more marker molecules for colorectal cancer in the diagnosis of colorectal cancer from a liquid sample obtained from an individual.
  • the expression "one or more” denotes 1 to 10, preferably 1 to 5, more preferred 3.
  • Preferred selected other CRC markers with which the measurement of PROC may be combined are CEA, CA 19-9, CA 72-4, and/or CA 242.
  • a very much preferred embodiment of the present invention is the use of protein PROC as a marker molecule for colorectal cancer in combination with one or more marker molecules for colorectal cancer in the diagnosis of colorectal cancer from a liquid sample obtained from an individual, whereby the at least one other marker molecule is selected from the group consisting of CEA, CA 19-9, CA 72-4, and CA 242.
  • the marker PROC is used in combination with CEA.
  • Diagnostic reagents in the field of specific binding assays like immunoassays, usually are best provided in the form of a kit, which comprises the specific binding agent and the auxiliary reagents required to perform the assay.
  • the present invention therefore also relates to an immunological kit comprising at least one specific binding agent for PROC and auxiliary reagents for measurement of PROC.
  • ROC receiver-operating characteristics
  • the ROC graph is a plot of all of the sensitivity/specificity pairs resulting from continuously varying the decision thresh-hold over the entire range of data observed.
  • the clinical performance of a laboratory test depends on its diagnostic accuracy, or the ability to correctly classify subjects into clinically relevant subgroups. Diagnostic accuracy measures the test's ability to correctly distinguish two different conditions of the subjects investigated. Such conditions are for example health and disease or benign versus malignant disease.
  • the ROC plot depicts the overlap between the two distributions by plotting the sensitivity versus 1 - specificity for the complete range of decision thresholds.
  • sensitivity or the true-positive fraction [defined as (number of true-positive test results) (number of true-positive + number of false- negative test results)].
  • This has also been referred to as positivity in the presence of a disease or condition. It is calculated solely from the affected subgroup.
  • the false-positive fraction or 1 - specificity [defined as (number of false- positive results) / (number of true-negative + number of false-positive results)]. It is an index of specificity and is calculated entirely from the unaffected subgroup.
  • the ROC plot is independent of the prevalence of disease in the sample.
  • Each point on the ROC plot represents a sensitivity/ 1 -specificity pair corresponding to a particular decision threshold.
  • a test with perfect discrimination has an ROC plot that passes through the upper left corner, where the true-positive fraction is 1.0, or 100% (perfect sensitivity), and the false-positive fraction is 0 (perfect specificity).
  • the theoretical plot for a test with no discrimination is a 45° diagonal line from the lower left corner to the upper right corner. Most plots fall in between these two extremes.
  • One convenient goal to quantify the diagnostic accuracy of a laboratory test is to express its performance by a single number.
  • Clinical utility of the novel marker PROC has been assessed in comparison to and in combination with the established marker CEA using a receiver operator curve analysis (ROC; Zweig, M. H., and Campbell, G., Clin. Chem. 39 (1993) 561-577).
  • This analysis has been based on well-defined patient cohorts consisting of 50 samples each from patients in Tl-3; NO; M0, more progressed tumor, i.e., T4 and/or various severity of metastasis (N+ and/or M+), and healthy controls, respectively.
  • Figure 1 shows a typical example of a 2D-gel, loaded with a tumor sample (left side), and a gel, loaded with a matched control sample (right side) obtained from adjacent healthy mucosa.
  • the apparent molecular weight and the isoelectric point of PROC correspond to the theoretical values of 33.4 kDa and 7.2, respectively.
  • the circle in the enlarged section of these gels indicates the position for the protein PROC. This protein is not detectable by the same method in healthy mucosa.
  • Figure 2 shows a typical example of a Western-Blot.
  • the gel is loaded with tissue lysates from colorectal tumor tissue and adjacent healthy control tissue from 3 patients (subject 5: colon ca (carcinoma), Dukes B; subject 7: colon ca, Dukes C; and subject 13: colon ca, Dukes B). Presence of PROC in the samples is tested using a polyclonal rabbit anti-PROC serum. Lanes containing tumor lysates are indicated with "T", lanes containing normal control tissue with "N”. The marker lane containing a molecular weight protein standard is indicated by "M”. The arrow indicates the position in the gel of the PROC band. All tumor samples give a strong signal at the position of PROC, whereas almost no signal can be detected in the lysates from adjacent normal control tissue. This strong overexpression of PROC in tumor tissue from colorectal cancer patients is shown in 10 out of 10 subjects tested.
  • tissue specimen from 10 patients suffering from colorectal cancer are analyzed. From each patient three different tissue types are collected from therapeutic resections: tumor tissue (>80% tumor) (T), adjacent healthy tissue (N) and stripped mucosa from adjacent healthy mucosa (M). The latter two tissue types serves as matched healthy control samples. Tissues are immediately snap frozen after resection and stored at -80°C before processing. Tumors are diagnosed by histopathological criteria.
  • 0.8-1.2 g of frozen tissue are put into a mortar and completely frozen by liquid nitrogen.
  • the tissue is pulverized in the mortar, dissolved in the 10-fold volume (w/v) of lysis buffer (40 mM Na-citrate, 5 mM MgCl 2 , 1% Genapol X-080, 0.02% Na-azide, Complete ® EDTA-free [Roche Diagnostics GmbH, Mannheim, Germany, Cat. No. 1 873 580] ) and subsequently homogenized in a Wheaton® glass homogenizer (20 x loose fitting, 20 x tight fitting).
  • lysis buffer 40 mM Na-citrate, 5 mM MgCl 2 , 1% Genapol X-080, 0.02% Na-azide, Complete ® EDTA-free
  • IPG strips pH 6-9 (Amersham Biosciences, Freiburg, Germany) overnight.
  • the IEF is performed using the following gradient protocol: 1.) 1 minute to 500 N; 2.) 2 h to 3,500 N; 3.) 22 h at constant 3,500 N giving rise to 82 kNh. After IEF, strips are stored at -80°C or directly used for SDS-PAGE.
  • the strips Prior to SDS-PAGE the strips are incubated in equilibration buffer (6 M urea, 50 mM Tris/HCl, pH 8.8, 30% glycerol, 2% SDS), for reduction DTT (15 min, + 50 mg DTT/10 ml), and for alkylation IAA (15 min, + 235 mg iodacetamide/10 ml) is added.
  • equilibration buffer 6 M urea, 50 mM Tris/HCl, pH 8.8, 30% glycerol, 2% SDS
  • DTT 15 min, + 50 mg DTT/10 ml
  • alkylation IAA 15 min, + 235 mg iodacetamide/10 ml
  • the strips are put on 12.5% polyacrylamide gels and subjected to electrophoresis at 1 W/gel for 1 h and thereafter at 17 W/gel. Subsequently, the gels are fixed (50% methanol, 10% acetate) and stained overnight with No vex Colloidal Blue Sta
  • protein PROC is found to be specifically expressed or strongly overexpressed in tumor tissue and not detectable or less strongly expressed in healthy control tissue.
  • Polyclonal antibody to the colorectal cancer marker protein PROC is generated for further use of the antibody in the measurement of serum and plasma and blood levels of PROC by immunodetection assays, e.g. Western Blotting and ELISA.
  • recombinant expression of the protein is performed for obtaining immunogens.
  • the expression is done applying a combination of the RTS 100 expression system and E.coli.
  • the DNA sequence is analyzed and recommendations for high yield cDNA silent mutational variants and respective PCR-primer sequences are obtained using the "ProteoExpert RTS E.coli HY” system. This is a commercial web based service (www.proteoexpert.com).
  • the "RTS 100 E. coli Linear Template Generation Set, His-tag” (Roche Diagnostics GmbH, Mannheim, Germany, Cat.No.
  • His-PROC fusion protein Purification of His-PROC fusion protein is done following standard procedures on a Ni-chelate column. Briefly, 1 1 of bacteria culture containing the expression vector for the His-PROC fusion protein is pelleted by centrifugation. The cell pellet is resuspended in lysis buffer, containing phosphate, pH 8.0, 7 M guanidium chloride, imidazole and thioglycerole, followed by homogenization using a Ultra-Turra ⁇ R . Insoluble material is pelleted by high speed centrifugation and the supernatant is applied to a Ni-chelate chromatographic column. The column is washed with several bed volumes of lysis buffer followed by washes with buffer, containing phosphate, pH 8,0 and Urea. Finally, bound antigen is eluted using a phosphate buffer containing SDS under acid conditions.
  • mice 12 week old A/J mice are initially immunized intraperitoneally with 100 ⁇ g PROC. This is followed after 6 weeks by two further intraperitoneal immunizations at monthly intervals. In this process each mouse is administered 100 ⁇ g PROC adsorbed to aluminum hydroxide and 10 germs of Bordetella pertussis.
  • the last two immunizations are carried out intravenously on the 3rd and 2nd day before fusion using 100 ⁇ g PROC in PBS buffer for each.
  • Spleen cells of the mice immunized according to a) are fused with myeloma cells according to Galfre, G., and Milstein, C, Methods in Enzymology 73 (1981) 3-46.
  • spleen cells of the immunized mouse are mixed with 2xl0 7 myeloma cells (P3X63-Ag8-653, ATCC CRL1580) and centrifuged (10 min at 300 g and 4° C). The cells are then washed once with RPMI 1640 medium without fetal calf serum (FCS) and centrifuged again at 400 g in a 50 ml conical tube. The supernatant is discarded, the cell sediment is gently loosened by tapping, 1 ml PEG
  • the hybridoma cells obtained are sown at a density of lxlO 5 cells per ml in RPMI 1640 medium containing 10% FCS and proliferated for 7 days in a fermenter (Thermodux Co., Wertheim/Main, Model MCS-104XL, Order No. 144-050). On average concentrations of 100 ⁇ g monoclonal antibody per ml are obtained in the culture supernatant. Purification of this antibody from the culture supernatant is carried out by conventional methods in protein chemistry (e.g. according to Bruck, C, et al., Methods in Enzymology 121 (1986) 587-695).
  • a fresh emulsion of the protein solution (100 ⁇ g/ml PROC protein) and complete Freund's adjuvant at the ratio of 1:1 is prepared.
  • Each rabbit is immunized with 1 ml of the emulsion at days 1, 7, 14 and 30, 60 and 90. Blood is drawn and resulting anti-PROC serum used for further experiments as described in examples 3 and 4.
  • IgG immunoglobulin G
  • One volume of rabbit serum is diluted with 4 volumes of acetate buffer (60 mM, pH 4.0). The pH is adjusted to 4.5 with 2 M Tris-base. Caprylic acid (25 ⁇ l/ml of diluted sample) is added drop-wise under vigorous stirring. After 30 min the sample is centrifuged (13,000 x g, 30 min, 4°C), the pellet discarded and the supernatant collected. The pH of the supernatant is adjusted to 7.5 by the addition of 2 M Tris-base and filtered (0.2 ⁇ m). The immunoglobulin in the supernatant is precipitated under vigorous stirring by the drop-wise addition of a 4 M ammonium sulfate solution to a final concentration of 2 M. The precipitated immunoglobulins are collected by centrifugation (8,000 x g, 15 min, 4°C).
  • the supernatant is discarded.
  • the pellet is dissolved in 10 mM NaH 2 PO_JNaOH, pH 7.5, 30 mM NaCl and exhaustively dialyzed.
  • the dialysate is centrifuged (13,000 x g, 15 min, 4°C) and filtered (0.2 ⁇ m).
  • Polyclonal rabbit IgG is brought to 10 mg/ml in 10 mM NaH 2 PO 4 /NaOH, pH 7.5, 30 mM NaCl. Per ml IgG solution 50 ⁇ l Biotin -N-hydroxysuccinimide (3.6 mg/ml in DMSO) are added. After 30 min at room temperature, the sample is chromatographed on Superdex 200 (10 mM NaH 2 PO 4 /NaOH, pH 7.5, 30 mM NaCl). The fraction containing biotinylated IgG are collected. Monoclonal antibodies are biotinylated according to the same procedure.
  • Polyclonal rabbit IgG is brought to 10 mg/ml in 10 mM NaH 2 PO 4 /NaOH, 30 mM NaCl, pH 7.5. Per ml IgG solution 50 ⁇ l digoxigenin-3-O-methylcarbonyl- ⁇ - aminocaproic acid-N-hydroxysuccinimide ester (Roche Diagnostics, Mannheim, Germany, Cat. No. 1 333 054) (3.8 mg/ml in DMSO) are added. After 30 min at room temperature, the sample is chromatographed on Superdex ® 200 (10 mM
  • Example 3 Western Blotting for the detection of PROC in human colorectal cancer tissue using polyclonal antibody as generated in Example 2.
  • Tissue lysates from tumor samples and healthy control samples are prepared as described in Example 1, "Tissue preparation”. SDS-PAGE and Western-Blotting are carried out using reagents and equipment of Invitrogen, Düsseldorf, Germany. For each tissue sample tested, 10 ⁇ g of tissue lysate are diluted in reducing NuPAGE ® (Invitrogen) SDS sample buffer and heated for 10 min at 95°C. Samples are run on 4-12% NuPAGE ® gels (Tris-Glycine) in the MES running buffer system. The gel-separated protein mixture is blotted onto nitrocellulose membranes using the Invitrogen XCell II Blot Module (Invitrogen) and the NuPAGE" transfer buffer system.
  • NuPAGE ® Invitrogen
  • the membranes are washed three times in PBS/0.05% Tween-20 and blocked with Roti®-Block blocking buffer (A151.1; Carl Roth GmbH, Düsseldorf, Germany) for 2 h.
  • the primary antibody, polyclonal rabbit anti-PROC serum (generation described in Example 2), is diluted 1:10,000 in
  • Roti ® -Block blocking buffer and incubated with the membrane for 1 h.
  • the membranes are washed 6 times in PBS/0.05% Tween-20.
  • the specifically bound primary rabbit antibody is labeled with an POD-conjugated polyclonal sheep anti- rabbit IgG antibody, diluted to 10 mU/ml in 0.5x Roti®-Block blocking buffer. After incubation for 1 h, the membranes are washed 6 times in PBS/0.05% Tween-
  • the membrane is incubated with the Lumi-Light PLUS Western Blotting Substrate (Order-No. 2015196, Roche Diagnostics GmbH, Mannheim, Germany) and exposed to an autoradiographic film.
  • Lumi-Light PLUS Western Blotting Substrate Order-No. 2015196, Roche Diagnostics GmbH, Mannheim, Germany
  • a sandwich ELISA For detection of PROC in human serum or plasma, a sandwich ELISA is developed. For capture and detection of the antigen, aliquots of the anti-PROC polyclonal antibody (see Example 2) are conjugated with biotin and digoxygenin, respectively.
  • Streptavidin-coated 96-well microwell plates are incubated with 100 ⁇ l biotinylated anti-PROC polyclonal antibody for 60 min at 10 ⁇ g/ml in 10 mM phosphate, pH 7.4, 1% BSA, 0.9% NaCl and 0.1% Tween-20. After incubation, plates are washed three times with 0.9% NaCl , 0.1% Tween-20. Wells are then incubated for 2 h with either a serial dilution of the recombinant protein (see Example 2) as standard antigen or with diluted plasma samples from patients. After binding of
  • PROC plates are washed three times with 0.9% NaCl , 0.1% Tween-20.
  • wells are incubated with 100 ⁇ l of digoxygenylated anti- PROC polyclonal antibody for 60 min at 10 ⁇ g/ml in 10 mM phosphate, pH 7.4, 1% BSA, 0.9% NaCl and 0.1% Tween-20. Thereafter, plates are washed three times to remove unbound antibody.
  • wells are incubated with 20 mU/ml anti-digoxigenin-POD conjugates (Roche Diagnostics GmbH, Mannheim, Germany, Catalog No. 1633716) for 60 min in 10 mM phosphate, pH 7.4, 1% BSA,
  • Accuracy is assessed by analyzing individual liquid samples obtained from well- characterized patient cohorts, i.e., 50 patients having undergone colonoscopy and found to be free of adenoma or CRC, 50 patients diagnosed and staged as Ti s -3, NO,
  • CEA as measured by a commercially available assay (Roche Diagnostics, CEA-assay (Cat. No. 1 173 1629 for Elecsys ® Systems immunoassay analyzer) and PROC measured as described above are quantified in a serum obtained from each of these individuals.
  • ROC-analysis is performed according to Zweig, M. H., and Campbell, supra. Discriminatory power for differentiating patients in the group Tj s -3, NO, M0 from healthy individuals as measured by the area under the curve is found to be at least as good for PROC as compared to the established marker CEA.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Chemical & Material Sciences (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Biotechnology (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

The present invention relates to the diagnosis of colorectal cancer. It discloses the use of the protein pyrroline-5-carboxylate reductase (PROC) in the diagnosis of colorectal cancer. It relates to a method for diagnosis of colorectal cancer from a liquid sample, derived from an individual by measuring PROC in said sample. Measurement of PROC can, e.g., be used in the early detection or diagnosis of colorectal cancer.

Description

Use of protein PROC as a marker for colorectal cancer
The present invention relates to the diagnosis of colorectal cancer. It discloses the use of the protein PROC (= Pyrroline-5-carboxylate reductase) in the diagnosis of colorectal cancer. Furthermore, it especially relates to a method for diagnosis of colorectal cancer from a liquid sample, derived from an individual by measuring PROC in said sample. Measurement of PROC can, e.g., be used in the early detection or diagnosis of colorectal cancer.
Cancer remains a major public health challenge despite progress in detection and therapy. Amongst the various types of cancer, colorectal cancer (= CRC) is one of the most frequent cancers in the Western world.
The earlier cancer can be detected/diagnosed, the better is the overall survival rate.
This is especially true for CRC. The prognosis in advanced stages of tumor is poor.
More than one third of the patients will die from progressive disease within five years after diagnosis, corresponding to a survival rate of about 40% for five years.
Current treatment is only curing a fraction of the patients and clearly has the best effect on those patients diagnosed in an early stage of disease.
With regard to CRC as a public health problem, it is essential that more effective screening and preventative measures for colorectal cancer be developed.
The earliest detection procedures available at present for colorectal cancer involve using tests for fecal blood or endoscopic procedures. However, significant tumor size must typically exist before fecal blood is detected. The sensitivity of the guaiac- based fecal occult blood tests is ~26%, which means 74% of patients with malignant lesions will remain undetected (Ahlquist, D.A., Gastroenterol. Clin. North Am. 26 (1997) 41-55). The visualization of precancerous and cancerous lesions represents the best approach to early detection, but colonoscopy is invasive with significant costs, risks, and complications (Silvis, S.E., et al, JAMA 235 (1976) 928-930;
Geenen, J.E., et al., Am. J. Dig. Dis. 20 (1975) 231-235; Anderson, W.F., et al, J. Natl. Cancer Institute 94 (2002) 1126-1133).
In the recent years a tremendous amount of so-called colon specific or even so- called colorectal cancer specific genes has been reported. The vast majority of the corresponding research papers or patent applications are based on data obtained by analysis of RNA expression patterns in colon (cancer) tissue versus a different tissue or an adjacent normal tissue, respectively. Such approaches may be summarized as differential mRNA display techniques.
As an example for data available from mRNA-display techniques, WO 01/96390 shall be mentioned and discussed. This application describes and claims more than two hundred isolated polynucleotides and the corresponding polypeptides as such, as well as their use in the detection of CRC. However, it is general knowledge that differences on the level of mRNA are not mirrored by the level of the corresponding proteins. A protein encoded by a rare mRNA may be found in very high amounts and a protein encoded by an abundant mRNA may nonetheless be hard to detect and find at all. This lack of correlation between mRNA-level and protein level is due to reasons like mRNA stability, efficiency of translation, stability of the protein, etc.
There also are recent approaches investigating the differences in protein patterns between different tissues or between healthy and diseased tissue in order to identify candidate marker molecules which might be used in the diagnosis of CRC. Brϋnagel, G., et al., Cancer Research 62 (2002) 2437-2442 have identified seven nuclear matrix proteins which appear to be more abundant in CRC tissixe as compared to adjacent normal tissue. No data from liquid samples obtained from an individual are reported.
WO 02/078636 reports about nine colorectal cancer- associated spots as found by surface-enhanced laser desorption and ionization (SELDI). These spots are seen more frequently in sera obtained from patients with CRC as compared to sera obtained from healthy controls. However, the identity of the molecule(s) comprised in such spot, e.g., its (their sequence), is not known.
Despite the large and ever growing list of candidate protein markers in the field of CRC, to date clinical/diagnostic utility of these molecules is not known. In order to be of clinical utility a new diagnostic marker as a single marker should be at least as good as the best single marker known in the art. Or, a new marker should lead to a progress in diagnostic sensitivity and/or specificity either if used alone or in combination with one or more other markers, respectively. The diagnostic sensitivity and/or specificity of a test is best assessed by its receiver-operating characteristics, which will be described in detail below.
At present, only diagnostic blood tests based on the detection of carcinoembryonic antigen (CEA), a tumor-associated glycoprotein, are available to assist diagnosis in the field of CRC. CEA is increased in 95% of tissue samples obtained from patients with colorectal, gastric, and pancreatic cancers and in the majority of breast, lung, and head and neck carcinomas (Goldenberg, D.M., et al., J. Natl. Cancer Inst. (Bethesda) 57 (1976) 11-22). Elevated CEA levels have also been reported in patients with nonmalignant disease, and many patients with colorectal cancer have normal CEA levels in the serum, especially during the early stage of the disease
(Carriquiry, L.A., and Pineyro, A., Dis. Colon Rectum 42 (1999) 921-929; Herrera, M.A., et al., Ann. Surg. 183 (1976) 5-9; Wanebo, H.J., et al., N. Engl. J. Med. 299 (1978) 448-451). The utility of CEA as measured from serum or plasma in detecting recurrences is reportedly controversial and has yet to be widely applied (Martell, R.E., et al, Int. J. Biol. Markers 13 (1998) 145-149; Moertel, C.G., et al., JAMA 270
(1993) 943-947).
In light of the available data, serum CEA determination possesses neither the sensitivity nor the specificity to enable its use as a screening test for colorectal cancer in the asymptomatic population (Reynoso, G., et al., JAMA 220 (1972) 361- 365; Sturgeon, C, Clinical Chemistry 48 (2002) 1151-1159).
Whole blood, serum or plasma are the most widely used sources of sample in clinical routine. The identification of an early CRC tumor marker that would allow reliable cancer detection or provide early prognostic information could lead to a diagnostic assay that would greatly aid in the diagnosis and in the management of this disease. Therefore, an urgent clinical need exists to improve the diagnosis of
CRC from blood. It is especially important to improve the early diagnosis of CRC, since for patients diagnosed early on chances of survival are much higher as compared to those diagnosed at a progressed stage of disease.
It was the task of the present invention to investigate whether a new marker can be identified which may aid in CRC diagnosis. Surprisingly, it has been found that use of protein PROC can at least partially overcome the problems known from the state of the art.
The present invention therefore relates to a method for the diagnosis of colorectal cancer comprising the steps of a) providing a- liquid sample obtained from an individual, b) contacting said sample with a specific binding agent for PROC under conditions appropriate for formation of a complex between said binding agent and PROC, and c) correlating the amount of complex formed in (b) to the diagnosis of colorectal cancer.
Another preferred embodiment of the invention is a method for the diagnosis of colorectal cancer comprising the steps of a) cα-ntacting a liquid sample obtained from an individual with a specific binding agent for PROC under conditions appropriate for formation of a complex between said binding agent and PROC, and b) correlating the amount of complex formed in (a) to the diagnosis of colorectal cancer.
As the skilled artisan will appreciate, any sucti diagnosis is made in vitro. The patient sample is discarded afterwards. The patient sample is solely used for the in vitro diagnostic method of the invention and tib.e material of the patient sample is not transferred back into the patient's body. Typically, the sample is a liquid sample.
The protein PROC (Pyrroline-5-carboxylate reductase, also known as PYCR1;
Swiss-PROT: P32322) is characterized by the seq uence given SEQ ID NO:
PROC catalyzes the NAD(P)H-dependent conversion of pyrroline-5 -carboxylate to proline. Merrill, M. J., et al., J. Biol. Chem. 264 (1989) 9352-9358 studied the properties of human erythrocyte pyrroline-5-carboxylate reductase. They concluded that in addition to the traditional role of catalyzing the obligatory and final unidirectional step in pyrroline biosynthesis, the enzyme may play a physiologic role in the generation of NADP(+) in some cell types including human erythrocytes.
Dougherty, K. M., et al, J. Biol. Chem. 267 ( 1992) 871-875 cloned a cDNA by complementation of proline auxotrophy in a Saccharomyces cerevisiae mutant strain ('pro3'). The 1,810-bp cDNA hybridized to a 1.85-kb mRNA in samples from human cell lines and predicated a 319-amino acid, 33.4-kDa protein. Studies of somatic cell hybrids indicated that PYCR1 is a single-copy gene that is located on chromosome 17.
Greengard, O., and Herzfeld, A., Cancer Res. 37 (1977) 884-891 found elevated enzymatic activity of PROC in lung neoplasm compared to normal adult lung. Herzfeld, A., et al., Cancer 42 (1978) 1280-1283 found elevated enzymatic activity of PROC in colon adenocarcinoma compared to norm-al colon tissues. However, it was not clear if more than one isoform of PROC contributed to the measured enzymatic activity. Direct measurement of the protein, e.g. by immunological technologies was not done in any of the studies listed.
As obvious to the skilled artisan, the present invention shall not be construed to be limited to the full-length protein PROC of SEQ ID NO= 1. Physiological or artificial fragments of PROC, secondary modifications of PROC-, as well as allelic variants of PROC are also encompassed by the present invention. Artificial fragments preferably encompass a peptide produced synthetically or by recombinant techniques, which at least comprises one epitope of diagnostic interest consisting of at least 6 contiguous amino acids as derived from the sequence disclosed in SEQ ID NO:l. Such fragment may advantageously be used for generation of antibodies or as a standard in an immunoassay. More preferred the artificial fragment comprises at least two epitopes of interest appropriate for setting ULp a sandwich immunoassay.
In preferred embodiments, the novel marker PROC may be used for monitoring as well as for screening purposes.
When used in patient monitoring the diagnostic method according to the present invention may help to assess tumor load, efficacy of treatment and tumor recurrence in the follow-up of patients. Increased levels of PROC are directly correlated to tumor burden. After chemotherapy a skiort term (few hours to 14 days) increase in PROC may serve as an indicator of tumor cell death. In the follow-up of patients (from 3 months to 10 years) an in rease of PROC can be used as an indicator for tumor recurrence. In a preferred embodiment the diagnostic method according to the present invention is used for screening purposes. I.e., it is used to assess subjects without a prior diagnosis of CRC by measuring the level of PI OC and correlating the level measured to the presence or absence of CRC.
Colorectal cancer most frequently progresses from adenomas (polyps) to malignant carcinomas. The different stages of CRC used to be classified according to Dukes' stages A to D.
The staging of cancer is the classification of the disease in terms of extent, progression, and severity. It groups cancer patients so that generalizations can be made about prognosis and the choice of therapy.
Today, the TNM system is the most widely used classification of the anatomical extent of cancer. It represents an internationally accepted, uniform staging system. There are three basic variables: T (the extent of the primary tumor), N (the status of regional lymph nodes) and M (the presence or absence of distant metastases). The TNM criteria are published by the UICC (International Union Against Cancer),
Sobin, L.H., Wittekind, Ch. (eds): TNM Classification of Malignant Tumours, fifth edition, 1997.
What is especially important is, that early diagnosis of CRC translates to a much better prognosis. Malignant tumors of the colorectiαm arise from benign tumors, i.e. from adenoma. Therefore, best prognosis have those patients diagnosed at the adenoma stage. Patients diagnosed as early as in stage TiS, NO, M0 or Tl-3; NO; M0, if treated properly have a more than 90% chance of survival 5 years after diagnosis as compared to a 5-years survival rate of only 10%> for patients diagnosed when distant metastases are already present.
In the sense of the present invention early diagnosis of CRC refers to a diagnosis at a pre-malignant state (adenoma) or at a tumor stage where no metastases at all (neither proximal nor distal), i.e., adenoma, Tιs, NI0, M0 or Tl-4; NO; M0 are present. Tls denotes carcinoma in situ.
In a preferred embodiment PROC is used to diagnose CRC as early as in the adenoma stage. It is further preferred, that CRC is diagnosed when it has not yet fully grown through the bowel wall and thus neither the visceral peritoneum is perforated nor other organs or structures are invaded, i.e., that diagnosis is made at stage Tιs, NO, MO or Tl-3; NO; MO (=^-3; NO; MO).
The diagnostic method according to the present invention is biased on a liquid sample which is derived from an individual. Unlike to methods known from the art PROC is specifically measured from this liquid sample by use of -a specific binding agent.
A specific binding agent is, e.g., a receptor for PROC, a lectin binding to PROC or an antibody to PROC. A specific binding agent has at least an affinity of 107 1/mol for its corresponding target molecule. The specific binding agent preferably has an affinity of 108 1/mol or even more preferred of 109 1/mol for its ta-rget molecule. As the skilled artisan will appreciate the term specific is used to indicate that other biomolecules present in the sample do not significantly bind to with the binding agent specific for PROC. Preferably, the level of binding to a b>iomolecule other than the target molecule results in a binding affinity which is only 10%, more preferably only 5% of the affinity of the target molecule or less. A most preferred specific binding agent will fulfill both the above minimum criteria for affinity as well as for specificity.
A specific binding agent preferably is an antibody reactive with PROC. The term antibody refers to a polyclonal antibody, a monoclonal antibody, fragments of such antibodies, as well as to genetic constructs comprising the binding domain of an antibody.
Any antibody fragment retaining the above criteria of a specific binding agent can be used. Antibodies are generated by state of the art procedures, e. g., as described in
Tijssen (Tijssen, P., Practice and theory of enzyme immunoassays 11 (1990) the whole book, especially pages 43-78; Elsevier, Amsterdam). In addition, the skilled artisan is well aware of methods based on immunosorbents that can be used for the specific isolation of antibodies. By these means the quality of polyclonal antibodies and hence their performance in immunoassays can be enhanced. CTijssen, P., supra, pages 108-115). For the achievements as disclosed in the present invention polyclonal antibodies raised in rabbits have been used. However, clearly also polyclonal antibodies from different species , e.g. rats or guinea pigs, as well as monoclonal antibodies can also be used. Since monoclonal antibodies can be produced in any amoα-nt required with constant properties, they represent ideal tools in development of an assay for clinical routine. The generation and use of monoclonal antibodies to PROC in a method according to the present invention is yet another preferred embodiment.
As the skilled artisan will appreciate now, that PROC has been identified as a marker which is useful in the diagnosis of CRC, alternative ways may be used to reach a result comparable to the achievements of the present invention. For example, alternative strategies to generate antibodies may be used. Such strategies comprise amongst others the use of synthetic peptides, representing an epitope of PROC for immunization. Alternatively, DNA Immunization also known as DNA vaccination may be used.
For measurement the liquid sample obtained from an individual is incubated with the specific binding agent for PROC under conditions appropriate for formation of a binding agent PROC-complex. Such conditions need not be specifie d, since the skilled artisan without any inventive effort can easily identify such appropriate incubation conditions.
As a final step according to the method disclosed in the present indention the amount of complex is measured and correlated to the diagnosis of CRC. As the skilled artisan will appreciate there are numerous methods to measure the amount of the specific binding agent PROC-complex all described in detail in relevant textbooks (cf., e.g., Tijssen P., supra, or Diamandis, et al., eds. (1996) Immunoassay, Academic Press, Boston).
Preferably PROC is detected in a sandwich type assay format. In such assay a first specific binding agent is used to capture PROC on the one side an-d a second specific binding agent, which is labeled to be directly or indirectiy detectable is used on the other side. As mentioned above, it has surprisingly been found that PROC can be measured from a liquid sample obtained from an individual sample. No tissue and no biopsy sample is required to apply the marker PROC in the diagnosis of CRC.
In a preferred embodiment the method according to the present invention is practiced with serum as liquid sample material.
In a further preferred embodiment the method according to the present invention is practiced with plasma as liquid sample material.
In a further preferred embodiment the method according to the present invention is practiced with whole blood as liquid sample material.
Furthermore stool can be prepared in various ways known to the skilled artisan to result in a liquid sample as well. Such sample liquid derived from stool also represents a preferred embodiment according to the present invention.
Whereas application of routine proteomics methods to tissue samples, leads to the identification of many potential marker candidates for the tissue selected, the inventors of the present invention have surprisingly been able to -detect protein
PROC in a bodily fluid sample. Even more surprising they have been able to demonstrate that the presence of PROC in such liquid sample obtained from an individual can be correlated to the diagnosis of colorectal cancer.
Antibodies to PROC with great advantage can be used in established procedures, e.g., to detect colorectal cancer cells in situ, in biopsies, or in immu-r-ohistological procedures.
Preferably, an antibody to PROC is used in a qualitative (PROC pres ent or absent) or quantitative (PROC amount is determined) immunoassay.
Measuring the level of protein PROC has proven very advantageous in the field of CRC. Therefore, in a further preferred embodiment, the present in ention relates to use of protein PROC as a marker molecule in the diagnosis of colorectal cancer from a liquid sample obtained from an individual. The term marker molecule is used to indicate that an increased level of the analyte PROC as measured from a bodily fluid of an individual marks the presence of CRC.
It is especially preferred to use the novel marker PROC in the early diagnosis of colorectal cancer.
The use of protein PROC itself, represents a significant progress to the challenging field of CRC diagnosis. Combining measurements of PROC with other known markers, like CEA, or with other markers of CRC yet to be discovered, leads to further improvements. Therefore in a further preferred embodiment the present invention relates to the use of PROC as a marker molecule for colorectal cancer in combination with one or more marker molecules for colorectal cancer in the diagnosis of colorectal cancer from a liquid sample obtained from an individual. In this regard, the expression "one or more" denotes 1 to 10, preferably 1 to 5, more preferred 3. Preferred selected other CRC markers with which the measurement of PROC may be combined are CEA, CA 19-9, CA 72-4, and/or CA 242. Thus, a very much preferred embodiment of the present invention is the use of protein PROC as a marker molecule for colorectal cancer in combination with one or more marker molecules for colorectal cancer in the diagnosis of colorectal cancer from a liquid sample obtained from an individual, whereby the at least one other marker molecule is selected from the group consisting of CEA, CA 19-9, CA 72-4, and CA 242. Very preferred the marker PROC is used in combination with CEA.
Diagnostic reagents in the field of specific binding assays, like immunoassays, usually are best provided in the form of a kit, which comprises the specific binding agent and the auxiliary reagents required to perform the assay. The present invention therefore also relates to an immunological kit comprising at least one specific binding agent for PROC and auxiliary reagents for measurement of PROC.
Accuracy of a test is best described by its receiver-operating characteristics (ROC) (see especially Zweig, M. H., and Campbell, G., Clin. Chem. 39 (1993) 561-577). The ROC graph is a plot of all of the sensitivity/specificity pairs resulting from continuously varying the decision thresh-hold over the entire range of data observed. The clinical performance of a laboratory test depends on its diagnostic accuracy, or the ability to correctly classify subjects into clinically relevant subgroups. Diagnostic accuracy measures the test's ability to correctly distinguish two different conditions of the subjects investigated. Such conditions are for example health and disease or benign versus malignant disease.
In each case, the ROC plot depicts the overlap between the two distributions by plotting the sensitivity versus 1 - specificity for the complete range of decision thresholds. On the y-axis is sensitivity, or the true-positive fraction [defined as (number of true-positive test results) (number of true-positive + number of false- negative test results)]. This has also been referred to as positivity in the presence of a disease or condition. It is calculated solely from the affected subgroup. On the x- axis is the false-positive fraction, or 1 - specificity [defined as (number of false- positive results) / (number of true-negative + number of false-positive results)]. It is an index of specificity and is calculated entirely from the unaffected subgroup. Because the true- and false-positive fractions are calculated entirely separately, by using the test results from two different subgroups, the ROC plot is independent of the prevalence of disease in the sample. Each point on the ROC plot represents a sensitivity/ 1 -specificity pair corresponding to a particular decision threshold. A test with perfect discrimination (no overlap in the two distributions of results) has an ROC plot that passes through the upper left corner, where the true-positive fraction is 1.0, or 100% (perfect sensitivity), and the false-positive fraction is 0 (perfect specificity). The theoretical plot for a test with no discrimination (identical distributions of results for the two groups) is a 45° diagonal line from the lower left corner to the upper right corner. Most plots fall in between these two extremes. (If the ROC plot falls completely below the 45° diagonal, this is easily remedied by reversing the criterion for "positivity" from "greater than" to "less than" or vice versa.) Qualitatively, the closer the plot is to the upper left corner, the higher the overall accuracy of the test.
One convenient goal to quantify the diagnostic accuracy of a laboratory test is to express its performance by a single number. The most common global measure is the area under the ROC plot. By convention, this area is always > 0.5 (if it is not, one can reverse the decision rule to make it so). Values range between 1.0 (perfect separation of the test values of the two groups) and 0.5 (no apparent distributional difference between the two groups of test values). The area does not depend only on a particular portion of the plot such as the point closest to the diagonal or the sensitivity at 90% specificity, but on the entire plot. This is a quantitative, descriptive expression of how close the ROC plot is to the perfect one (area = 1.0).
Clinical utility of the novel marker PROC has been assessed in comparison to and in combination with the established marker CEA using a receiver operator curve analysis (ROC; Zweig, M. H., and Campbell, G., Clin. Chem. 39 (1993) 561-577). This analysis has been based on well-defined patient cohorts consisting of 50 samples each from patients in Tl-3; NO; M0, more progressed tumor, i.e., T4 and/or various severity of metastasis (N+ and/or M+), and healthy controls, respectively.
The following examples, references, sequence listing and figures are provided to aid the understanding of the present invention, the true scope of which is set forth in the appended claims. It is understood that modifications can be made in the procedures set forth without departing from the spirit of the invention.
Description of the Figures
Figure 1 Figure 1 shows a typical example of a 2D-gel, loaded with a tumor sample (left side), and a gel, loaded with a matched control sample (right side) obtained from adjacent healthy mucosa. The apparent molecular weight and the isoelectric point of PROC correspond to the theoretical values of 33.4 kDa and 7.2, respectively. The circle in the enlarged section of these gels indicates the position for the protein PROC. This protein is not detectable by the same method in healthy mucosa.
Figure 2 Figure 2 shows a typical example of a Western-Blot. The gel is loaded with tissue lysates from colorectal tumor tissue and adjacent healthy control tissue from 3 patients (subject 5: colon ca (carcinoma), Dukes B; subject 7: colon ca, Dukes C; and subject 13: colon ca, Dukes B). Presence of PROC in the samples is tested using a polyclonal rabbit anti-PROC serum. Lanes containing tumor lysates are indicated with "T", lanes containing normal control tissue with "N". The marker lane containing a molecular weight protein standard is indicated by "M". The arrow indicates the position in the gel of the PROC band. All tumor samples give a strong signal at the position of PROC, whereas almost no signal can be detected in the lysates from adjacent normal control tissue. This strong overexpression of PROC in tumor tissue from colorectal cancer patients is shown in 10 out of 10 subjects tested.
Abbreviations
ABTS 2,2'-Azino-di- [3-ethylbenzthiazoline sulfonate (6)] diammonium salt
BSA bovine serum albumin cDNA complementary DNA
CHAPS (3-[(3-Cholamidopropyl)-dimethylammonio]- 1 -propane sulfonate)
DMSO dimethyl sulfoxide
DTT dithiothreitol
EDTA ethylene diamine tetraacetic acid
ELISA enzyme-linked immunosorbent assay
HRP horseradish peroxidase
IAA iodacetamid
IgG immunoglobulin G
IEF isoelectric focussing
IPG immobilized pH gradient
LDS lithium dodecyl sulfate
MALDI-TOF matrix-assisted laser desorption/ionisation-time of flight mass spectrometry
MES mesityl, 2,4,6-trimethylphenyl
OD optical density
PAGE polyacrylamide gel electrophoresis
PBS phosphate buffered saline
PI isoelectric point
RTS rapid translation system
SDS sodium dodecyl sulfate Example 1
Identification of PROC as a potential colorectal cancer marker
Sources of tissue
In order to identify tumor-specific proteins as potential diagnostic markers for colorectal cancer, analysis of three different kinds of tissue using proteomics methods is performed.
In total, tissue specimen from 10 patients suffering from colorectal cancer are analyzed. From each patient three different tissue types are collected from therapeutic resections: tumor tissue (>80% tumor) (T), adjacent healthy tissue (N) and stripped mucosa from adjacent healthy mucosa (M). The latter two tissue types serves as matched healthy control samples. Tissues are immediately snap frozen after resection and stored at -80°C before processing. Tumors are diagnosed by histopathological criteria.
Tissue preparation
0.8-1.2 g of frozen tissue are put into a mortar and completely frozen by liquid nitrogen. The tissue is pulverized in the mortar, dissolved in the 10-fold volume (w/v) of lysis buffer (40 mM Na-citrate, 5 mM MgCl2, 1% Genapol X-080, 0.02% Na-azide, Complete® EDTA-free [Roche Diagnostics GmbH, Mannheim, Germany, Cat. No. 1 873 580] ) and subsequently homogenized in a Wheaton® glass homogenizer (20 x loose fitting, 20 x tight fitting). 3 ml of the homogenate are subjected to a sucrose-density centrifugation (10-60% sucrose) for 1 h at 4,500 x g. After this centrifugation step three fractions are obtained. The fraction on top of the gradient contains the soluble proteins and is used for further analysis.
Isoelectric focussing (IEF) and SDS-PAGE
For IEF, 3 ml of the suspension are mixed with 12 ml sample buffer (7 M urea, 2 M thiourea, 2% CHAPS, 0.4% IPG buffer pH 6-9, 0.5% DTT) and incubated for 1 h. The samples are concentrated in an Amicon" Ultra- 15 device (Millipore GmbH, Schwalbach, Germany) and the protein concentration is determined using the Bio- Rad® protein assay (Cat.No. 500-0006; Bio-Rad Laboratories GmbH, Mϋnchen, Germany) following the instructions of the supplier's manual. To a volume corresponding to 1.5 mg of protein sample buffer is added to a final volume of 350 μl. This solution is used to rehydrate IPG strips pH 6-9 (Amersham Biosciences, Freiburg, Germany) overnight. The IEF is performed using the following gradient protocol: 1.) 1 minute to 500 N; 2.) 2 h to 3,500 N; 3.) 22 h at constant 3,500 N giving rise to 82 kNh. After IEF, strips are stored at -80°C or directly used for SDS-PAGE.
Prior to SDS-PAGE the strips are incubated in equilibration buffer (6 M urea, 50 mM Tris/HCl, pH 8.8, 30% glycerol, 2% SDS), for reduction DTT (15 min, + 50 mg DTT/10 ml), and for alkylation IAA (15 min, + 235 mg iodacetamide/10 ml) is added. The strips are put on 12.5% polyacrylamide gels and subjected to electrophoresis at 1 W/gel for 1 h and thereafter at 17 W/gel. Subsequently, the gels are fixed (50% methanol, 10% acetate) and stained overnight with No vex Colloidal Blue Staining Kit (Invitrogen, Karlsruhe, Germany, Cat No. LC6025, 45- 7101)
Detection of PROC as a potential marker for colorectal cancer
Each patient is analyzed separately by image analysis with the ProteomeWeaver® software (Definiens AG, Germany, Mϋnchen). In addition, all spots of the gel are excised by a picking robot and the proteins present in the spots are identified by MALDI-TOF mass spectrometry (Ultraflex Tof/Tof, Bruker Daltonik GmbH,
Bremen, Germany). For each patient, 4 gels from the tumor sample are compared with 4 gels each from adjacent normal and stripped mucosa tissue and analyzed for distinctive spots corresponding to differentially expressed proteins. By this means, protein PROC is found to be specifically expressed or strongly overexpressed in tumor tissue and not detectable or less strongly expressed in healthy control tissue.
It therefore - amongst many other proteins - qualifies as a candidate marker for use in the diagnosis of colorectal cancer. Example 2
Generation of antibodies to the colorectal cancer marker protein PROC
Polyclonal antibody to the colorectal cancer marker protein PROC is generated for further use of the antibody in the measurement of serum and plasma and blood levels of PROC by immunodetection assays, e.g. Western Blotting and ELISA.
Recombinant protein expression in E. coli
In order to generate antibodies to PROC, recombinant expression of the protein is performed for obtaining immunogens. The expression is done applying a combination of the RTS 100 expression system and E.coli. In a first step, the DNA sequence is analyzed and recommendations for high yield cDNA silent mutational variants and respective PCR-primer sequences are obtained using the "ProteoExpert RTS E.coli HY" system. This is a commercial web based service (www.proteoexpert.com). Using the recommended primer pairs, the "RTS 100 E. coli Linear Template Generation Set, His-tag" (Roche Diagnostics GmbH, Mannheim, Germany, Cat.No. 3186237) system to generate linear PCR templates from the cDNA and for in vitro transcription and expression of the nucleotide sequence coding for the PROC protein is used. For Western-blot detection and later purification, the expressed protein contains a His-tag. The best expressing variant is identified. All steps from PCR to expression and detection are carried out according to the instructions of the manufacturer. The respective PCR product, containing all necessary T7 regulatory regions (promoter, ribosomal binding site and T7 terminator) is cloned into the pBAD TOPO® vector (Invitrogen, Karlsruhe, Germany, Cat. No. K 4300/01) following the manufacturer's instructions. For expression using the T7 regulatory sequences, the construct is transformed into E. coli BL 21 (DE 3) (Studier, F.W., et al., Methods Enzymol. 185 (1990) 60-89) and the transformed bacteria are cultivated in a 1 1 batch for protein expression.
Purification of His-PROC fusion protein is done following standard procedures on a Ni-chelate column. Briefly, 1 1 of bacteria culture containing the expression vector for the His-PROC fusion protein is pelleted by centrifugation. The cell pellet is resuspended in lysis buffer, containing phosphate, pH 8.0, 7 M guanidium chloride, imidazole and thioglycerole, followed by homogenization using a Ultra-TurraχR . Insoluble material is pelleted by high speed centrifugation and the supernatant is applied to a Ni-chelate chromatographic column. The column is washed with several bed volumes of lysis buffer followed by washes with buffer, containing phosphate, pH 8,0 and Urea. Finally, bound antigen is eluted using a phosphate buffer containing SDS under acid conditions.
Production of monoclonal antibodies against the PROC
a) Immunization of mice
12 week old A/J mice are initially immunized intraperitoneally with 100 μg PROC. This is followed after 6 weeks by two further intraperitoneal immunizations at monthly intervals. In this process each mouse is administered 100 μg PROC adsorbed to aluminum hydroxide and 10 germs of Bordetella pertussis.
Subsequently the last two immunizations are carried out intravenously on the 3rd and 2nd day before fusion using 100 μg PROC in PBS buffer for each.
b) Fusion and cloning
Spleen cells of the mice immunized according to a) are fused with myeloma cells according to Galfre, G., and Milstein, C, Methods in Enzymology 73 (1981) 3-46.
In this process ca. 1*108 spleen cells of the immunized mouse are mixed with 2xl07 myeloma cells (P3X63-Ag8-653, ATCC CRL1580) and centrifuged (10 min at 300 g and 4° C). The cells are then washed once with RPMI 1640 medium without fetal calf serum (FCS) and centrifuged again at 400 g in a 50 ml conical tube. The supernatant is discarded, the cell sediment is gently loosened by tapping, 1 ml PEG
(molecular weight 4000, Merck, Darmstadt) is added and mixed by pipetting. After 1 min in a water-bath at 37°C, 5 ml RPMI 1640 without FCS is added drop-wise at roo temperature within a period of 4-5 min. Afterwards 5 ml RPMI 1640 containing 10% FCS is added drop-wise within ca. 1 min, mixed thoroughly, filled to 50 ml with medium (RPMI 1640+10% FCS) and subsequently centrifuged for
10 min at 400 x g and 4°C. The sedimented cells are taken up in RPMI 1640 medium containing 10% FCS and sown in hypoxanthine-azaserine selection medium (100 mmol/1 hypoxanthine, 1 μg/ml azaserine in RPMI 1640+10% FCS). Interleukin 6 at 100 U/ml is added to the medium as a growth factor. After ca. 10 days the primary cultures are tested for specific antibody. PROC- positive primary cultures are cloned in 96-well cell culture plates by means of a fluorescence activated cell sorter. In this process again interleukin 6 at 100 U/ml is added to the medium as a growth additive.
c) Immunoglobulin isolation from the cell culture supernatants
The hybridoma cells obtained are sown at a density of lxlO5 cells per ml in RPMI 1640 medium containing 10% FCS and proliferated for 7 days in a fermenter (Thermodux Co., Wertheim/Main, Model MCS-104XL, Order No. 144-050). On average concentrations of 100 μg monoclonal antibody per ml are obtained in the culture supernatant. Purification of this antibody from the culture supernatant is carried out by conventional methods in protein chemistry (e.g. according to Bruck, C, et al., Methods in Enzymology 121 (1986) 587-695).
Generation of polyclonal antibodies
a) Immunization
For immunization, a fresh emulsion of the protein solution (100 μg/ml PROC protein) and complete Freund's adjuvant at the ratio of 1:1 is prepared. Each rabbit is immunized with 1 ml of the emulsion at days 1, 7, 14 and 30, 60 and 90. Blood is drawn and resulting anti-PROC serum used for further experiments as described in examples 3 and 4.
b) Purification of IgG (immunoglobulin G) from rabbit serum by sequential precipitation with caprylic acid and ammonium sulfate
One volume of rabbit serum is diluted with 4 volumes of acetate buffer (60 mM, pH 4.0). The pH is adjusted to 4.5 with 2 M Tris-base. Caprylic acid (25 μl/ml of diluted sample) is added drop-wise under vigorous stirring. After 30 min the sample is centrifuged (13,000 x g, 30 min, 4°C), the pellet discarded and the supernatant collected. The pH of the supernatant is adjusted to 7.5 by the addition of 2 M Tris-base and filtered (0.2 μm). The immunoglobulin in the supernatant is precipitated under vigorous stirring by the drop-wise addition of a 4 M ammonium sulfate solution to a final concentration of 2 M. The precipitated immunoglobulins are collected by centrifugation (8,000 x g, 15 min, 4°C).
The supernatant is discarded. The pellet is dissolved in 10 mM NaH2PO_JNaOH, pH 7.5, 30 mM NaCl and exhaustively dialyzed. The dialysate is centrifuged (13,000 x g, 15 min, 4°C) and filtered (0.2 μm).
Biotinylation of polyclonal rabbit IgG
Polyclonal rabbit IgG is brought to 10 mg/ml in 10 mM NaH2PO4/NaOH, pH 7.5, 30 mM NaCl. Per ml IgG solution 50 μl Biotin -N-hydroxysuccinimide (3.6 mg/ml in DMSO) are added. After 30 min at room temperature, the sample is chromatographed on Superdex 200 (10 mM NaH2PO4/NaOH, pH 7.5, 30 mM NaCl). The fraction containing biotinylated IgG are collected. Monoclonal antibodies are biotinylated according to the same procedure.
Digoxygenylation of polyclonal rabbit IgG
Polyclonal rabbit IgG is brought to 10 mg/ml in 10 mM NaH2PO4/NaOH, 30 mM NaCl, pH 7.5. Per ml IgG solution 50 μl digoxigenin-3-O-methylcarbonyl-ε- aminocaproic acid-N-hydroxysuccinimide ester (Roche Diagnostics, Mannheim, Germany, Cat. No. 1 333 054) (3.8 mg/ml in DMSO) are added. After 30 min at room temperature, the sample is chromatographed on Superdex® 200 (10 mM
NaH2PO4/NaOH, pH 7.5, 30 mM NaCl). The fractions containing digoxigenylated IgG are collected. Monoclonal antibodies are labeled with digoxigenin according to the same procedure.
Example 3 Western Blotting for the detection of PROC in human colorectal cancer tissue using polyclonal antibody as generated in Example 2.
Tissue lysates from tumor samples and healthy control samples are prepared as described in Example 1, "Tissue preparation". SDS-PAGE and Western-Blotting are carried out using reagents and equipment of Invitrogen, Karlsruhe, Germany. For each tissue sample tested, 10 μg of tissue lysate are diluted in reducing NuPAGE® (Invitrogen) SDS sample buffer and heated for 10 min at 95°C. Samples are run on 4-12% NuPAGE® gels (Tris-Glycine) in the MES running buffer system. The gel-separated protein mixture is blotted onto nitrocellulose membranes using the Invitrogen XCell II Blot Module (Invitrogen) and the NuPAGE" transfer buffer system. The membranes are washed three times in PBS/0.05% Tween-20 and blocked with Roti®-Block blocking buffer (A151.1; Carl Roth GmbH, Karlsruhe, Germany) for 2 h. The primary antibody, polyclonal rabbit anti-PROC serum (generation described in Example 2), is diluted 1:10,000 in
Roti®-Block blocking buffer and incubated with the membrane for 1 h. The membranes are washed 6 times in PBS/0.05% Tween-20. The specifically bound primary rabbit antibody is labeled with an POD-conjugated polyclonal sheep anti- rabbit IgG antibody, diluted to 10 mU/ml in 0.5x Roti®-Block blocking buffer. After incubation for 1 h, the membranes are washed 6 times in PBS/0.05% Tween-
20. For detection of the bound POD-conjugated anti-rabbit antibody, the membrane is incubated with the Lumi-LightPLUS Western Blotting Substrate (Order-No. 2015196, Roche Diagnostics GmbH, Mannheim, Germany) and exposed to an autoradiographic film.
Example 4
ELISA for the measurement of PROC in human serum and plasma samples.
For detection of PROC in human serum or plasma, a sandwich ELISA is developed. For capture and detection of the antigen, aliquots of the anti-PROC polyclonal antibody (see Example 2) are conjugated with biotin and digoxygenin, respectively.
Streptavidin-coated 96-well microwell plates are incubated with 100 μl biotinylated anti-PROC polyclonal antibody for 60 min at 10 μg/ml in 10 mM phosphate, pH 7.4, 1% BSA, 0.9% NaCl and 0.1% Tween-20. After incubation, plates are washed three times with 0.9% NaCl , 0.1% Tween-20. Wells are then incubated for 2 h with either a serial dilution of the recombinant protein (see Example 2) as standard antigen or with diluted plasma samples from patients. After binding of
PROC, plates are washed three times with 0.9% NaCl , 0.1% Tween-20. For specific detection of bound PROC, wells are incubated with 100 μl of digoxygenylated anti- PROC polyclonal antibody for 60 min at 10 μg/ml in 10 mM phosphate, pH 7.4, 1% BSA, 0.9% NaCl and 0.1% Tween-20. Thereafter, plates are washed three times to remove unbound antibody. In a next step, wells are incubated with 20 mU/ml anti-digoxigenin-POD conjugates (Roche Diagnostics GmbH, Mannheim, Germany, Catalog No. 1633716) for 60 min in 10 mM phosphate, pH 7.4, 1% BSA,
0.9% NaCl and 0.1% Tween-20. Plates are subsequently washed three times with the same buffer. For detection of antigen-antibody complexes, wells are incubated with 100 μl ABTS solution (Roche Diagnostics GmbH, Mannheim, Germany, Catalog No. 11685767) and OD is measured after 30-60 min at 405 nm with an ELISA reader.
Example 5
ROC analysis to assess clinical utility in terms of diagnostic accuracy
Accuracy is assessed by analyzing individual liquid samples obtained from well- characterized patient cohorts, i.e., 50 patients having undergone colonoscopy and found to be free of adenoma or CRC, 50 patients diagnosed and staged as Tis-3, NO,
M0 of CRC, and 50 patients diagnosed with progressed CRC, having at least tumor infiltration in at least one proximal lymph node or more severe forms of metastasis, respectively. CEA as measured by a commercially available assay (Roche Diagnostics, CEA-assay (Cat. No. 1 173 1629 for Elecsys® Systems immunoassay analyzer) and PROC measured as described above are quantified in a serum obtained from each of these individuals. ROC-analysis is performed according to Zweig, M. H., and Campbell, supra. Discriminatory power for differentiating patients in the group Tjs-3, NO, M0 from healthy individuals as measured by the area under the curve is found to be at least as good for PROC as compared to the established marker CEA.
Preliminary data indicate that PROC may also be very helpful in the follow-up of patients after surgery. List of References
Ahlquist, D.A., Gastroenterol. Clin. North Am. 26 (1997) 41-55
Anderson, W.F., et al., J. Natl. Cancer Institute 94 (2002) 1126-1133
Bruck, C, et al., Methods Enzymol. 121 (1986) 587-695 Brunagel, G., et al., Cancer Research 62 (2002) 2437-2442
Carriquiry, L.A., and Pineyro, A., Dis. Colon Rectum 42 (1999) 921-929
Diamandis, et al., eds. (1996) Immunoassay, Academic Press, Boston.
Dougherty, K. M., et al., J. Biol. Chem. 267 (1992) 871-875
Galfre, G., and Milstein, C, Methods Enzymol. 73 (1981) 3-46 Geenen, J.E., et al., Am. J. Dig. Dis. 20 (1975) 231-235
Goldenberg, D.M., et al., J. Natl. Cancer Inst. (Bethesda) 57 (1976) 11-22
Greengard, O., and Herzfeld, A., Cancer Res. 37 (1977) 884-891
Herrera, M.A., et al., Ann. Surg. 183 (1976) 5-9
Herzfeld, A., et al., Cancer 42 (1978) 1280-1283 Martell, R.E., et al., Int. J. Biol. Markers 13 (1998) 145-149
Merrill, M. J., et al., J. Biol. Chem. 264 (1989) 9352-9358
Moertel, C.G., et al., JAMA 270 (1993) 943-947
Reynoso, G., et al., JAMA 220 (1972) 361-365
Silvis, S.E., et al., JAMA 235 (1976) 928-930 Sobin, L.H., Wittekind, Ch. (eds), TNM Classification of Malignant Tumours, fifth edition, 1997
Studier, F.W., et al., Methods Enzymol. 185 (1990) 60-89
Sturgeon, C, Clinical Chemistry 48 (2002) 1151-1159
Tijssen, P., Practice and theory of enzyme immunoassays 11 (1990) the whole book, especially pages 43-78; Elsevier, Amsterdam
UICC (International Union Against Cancer), Sobin, L.H., Wittekind, Ch. (eds), TNM Classification of Malignant Tumours, fifth edition, 1997
Wanebo, H.J., et al., N. Engl. J. Med. 299 (1978) 448-451
WO 01/96390 WO 02/078636
Zweig, M. H., and Campbell, G., Clin. Chem. 39 (1993) 561-577

Claims

Patent Claims
1. A method for the diagnosis of colorectal cancer comprising the steps of a) providing a liquid sample obtained from an individual, b) contacting said sample with a specific binding agent for pyrroline-5- carboxylate reductase (PROC) under conditions appropriate for formation of a complex between said binding agent and PROC, and c) correlating the amount of complex formed in (b) to the diagnosis of colorectal cancer.
2. The method according to claim 1, further characterized in that said sample is serum.
3. The method according to claim 1, further characterized in that said sample is plasma.
4. The method according to claim 1, further characterized in that said sample is whole blood.
5. Use of protein PROC as a marker molecule in the diagnosis of colorectal cancer from a liquid sample obtained from an individual.
6. Use of protein PROC as a marker molecule in the early diagnosis of colorectal cancer from a liquid sample obtained from an individual.
7. Use according to claim 6, wherein the early diagnosis is made with sample derived from CRC patients in the adenoma stage.
8. Use according to claim 6, wherein the early diagnosis is made with sample derived from CRC patients in stage TJS -3; NO; M0.
9. Use of protein PROC as a marker molecule for colorectal cancer in combination with another marker molecule for colorectal cancer in the diagnosis of colorectal cancer from a liquid sample obtained from an individual.
0. An immunological kit comprising at least one specific binding agent for PROC and auxiliary reagents for measurement of PROC.
PCT/EP2005/003264 2004-03-30 2005-03-29 Use of protein proc as a marker for colorectal cancer WO2005095979A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP04007620.0 2004-03-30
EP04007620 2004-03-30

Publications (1)

Publication Number Publication Date
WO2005095979A1 true WO2005095979A1 (en) 2005-10-13

Family

ID=34966839

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/003264 WO2005095979A1 (en) 2004-03-30 2005-03-29 Use of protein proc as a marker for colorectal cancer

Country Status (1)

Country Link
WO (1) WO2005095979A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002078636A2 (en) * 2001-03-28 2002-10-10 Matritech, Inc. Detection and treatment of colorectal cancer
WO2002099043A2 (en) * 2001-06-05 2002-12-12 Exelixis, Inc. P5CRS AS MODIFIERS OF THE p53 PATHWAY AND METHODS OF USE

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002078636A2 (en) * 2001-03-28 2002-10-10 Matritech, Inc. Detection and treatment of colorectal cancer
WO2002099043A2 (en) * 2001-06-05 2002-12-12 Exelixis, Inc. P5CRS AS MODIFIERS OF THE p53 PATHWAY AND METHODS OF USE

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BRUENAGEL G ET AL: "Identification of nuclear matrix protein alterations associated with human colon cancer", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, BALTIMORE, MD, US, vol. 62, no. 8, 15 April 2002 (2002-04-15), pages 2437 - 2442, XP002237185, ISSN: 0008-5472 *
HERZFELD A ET AL: "HUMAN COLON TUMORS ENZYMATIC AND HISTOLOGICAL CHARACTERISTICS", CANCER, AMERICAN CANCER SOCIETY, PHILADELPHIA, PA, US, vol. 42, no. 3, 1978, pages 1280 - 1283, XP008048941, ISSN: 0008-543X *

Similar Documents

Publication Publication Date Title
US7205118B2 (en) Nicotinamide N-methyltransferase as a marker for colorectal cancer
US20060177880A1 (en) Use of PRN3/ILEU as a marker for colorectal cancer
US20060188949A1 (en) Use of protein PLST as a marker for colorectal cancer
US20060121540A1 (en) Use of protein MASP as a marker for colorectal cancer
WO2005015218A1 (en) Use of proteins proteinase 3 (prn3) and leukocyte elastase inhibitor (ileu) as a marker for colorectal cancer
WO2005095978A1 (en) Pyrroline-5-carboxylate reductase as a marker for colorectal concer
US20060188950A1 (en) Use of protein spee as a marker for colorectal cancer
WO2004071267A2 (en) Diagnosis of colorectal cancer by detection of nicotinamide n-methyltransferase in a stool sample
US20060199232A1 (en) Use of protein PSE3 as a marker for colorectal cancer
US20060194266A1 (en) Use of protein RLA-0 as a marker for colorectal cancer
WO2005015221A1 (en) Use of protein sahh as a marker for colorectal cancer
WO2005095979A1 (en) Use of protein proc as a marker for colorectal cancer
WO2005015234A1 (en) Use of protein sahh as a marker for colorectal cancer
US20070218510A1 (en) Use of protein PSA3 as a marker for colorectal cancer
WO2005015233A1 (en) Use of protein spermidine synthase (spee) as a marker for colorectal cancer
WO2005015223A1 (en) Use of protein acidic ribosomal protein p0 (rla-0) as a marker for colorectal cancer
WO2005015222A1 (en) Use of the far upstream element (fuse) binding protein (fubp) as a marker for colorectal cancer
WO2005015225A1 (en) Use of the far upstream element (fuse) binding protein (fubp) as a marker for colorectal cancer
WO2005015227A1 (en) Use of protein t- plastin (plst) as a marker for colorectal cancer
WO2005015230A1 (en) Use of protein psa3 as a marker for colorectal cancer
WO2004104592A1 (en) Use of protein masp as a marker for colorectal cancer

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

122 Ep: pct application non-entry in european phase