WO2005040808A1 - Use of protein fkbp52 as a marker for breast cancer - Google Patents

Use of protein fkbp52 as a marker for breast cancer Download PDF

Info

Publication number
WO2005040808A1
WO2005040808A1 PCT/EP2004/011602 EP2004011602W WO2005040808A1 WO 2005040808 A1 WO2005040808 A1 WO 2005040808A1 EP 2004011602 W EP2004011602 W EP 2004011602W WO 2005040808 A1 WO2005040808 A1 WO 2005040808A1
Authority
WO
WIPO (PCT)
Prior art keywords
fkbp52
breast cancer
diagnosis
protein
sample
Prior art date
Application number
PCT/EP2004/011602
Other languages
French (fr)
Inventor
Gabriele Pestlin
Peter Berndt
Marie-Luise Hagmann
Johann Karl
Hanno Langen
Norbert Wild
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 WO2005040808A1 publication Critical patent/WO2005040808A1/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
    • G01N33/57415Specifically defined cancers of breast

Definitions

  • FKBP52 FK506-binding protein 4
  • BC breast cancer
  • 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.
  • BC as a public health problem, it is essential that more effective screening and preventative measures for breast cancer will be developed.
  • the earliest detection procedures available at present for breast cancer involve using clinical breast examination and mammography.
  • significant tumor size must typically exist before a tumor is palpable or can be detected by a mammogram.
  • the density of the breast tissue and the age are important predictors of the accuracy of screening mammography.
  • the sensitivity ranges from 63% in women with extremely dense breasts to 87% in women with almost entirely fatty- breasts.
  • the sensitivity increases with age from 69% in women of about 40 years of age to 83% in women 80 years and older (Carney, P.A., et al., Ann. Intern. Med. 138 (3) (2003) 168-175). Only 20 - 25 % of mammographically detected abnormalities that are biopsied prove to be malignant.
  • WO 00/60076 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 BC.
  • 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 (Chen, G., et al., Molecular and Cellular Proteomics, 1.4 (2002) 304- 313).
  • 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 02/23200 reports about twelve breast 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 BC as compared to sera obtained from healthy controls. However, the identity of the molecule(s) comprised in such spot, e.g their sequence, is not known.
  • Nipple aspirate fluid has been used for many years as a potential non- invasive method to identify breast cancer-specific markers.
  • Kuerer et al. compared bilateral matched pair nipple aspirate fluids from women with unilateral invasive breast carcinoma by 2D gel electrophoresis (Kuerer, H.M., et al, Cancer 95 (2002) 2276-2282).
  • 30 to 202 different protein spots were detected in the NAF of breasts suffering from breast carcinoma and not in the matched NAF of the healthy breasts. These spots were detected by a gel image analysis. But the identity of the protein spots 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.
  • CA 15-3 a tumor-associated mucin, and carcinoembryonic antigen (CEA), a tumor associated glycoprotein, are available to assist diagnosis in the field of BC.
  • CA 15-3 is usually increased in patients with advanced breast cancer.
  • CA 15-3 levels are rarely elevated in women with early stage breast cancer (Duffy, M.J., Critical Reviews in Clinical Laboratory Sciences 38 (2001) 225-262). Cancers of the ovary, lung and prostate may also raise CA 15-3 levels. Elevated levels of CA 15-3 may be associated with non-cancerous conditions, such as benign breast or ovary disease, endometriosis, pelvic inflammatory disease, and hepatitis.
  • Pregnancy and lactation can also cause CA 15-3 levels to raise (National Cancer Institute, Cancer Facts, Fact Sheet 5.18 (1998) 1-5).
  • the primary use of CEA is in monitoring colon cancer, especially when the disease has metastasized.
  • a variety of cancers can produce elevated levels of CEA, including breast cancer.
  • the present invention therefore relates to a method for the diagnosis of breast cancer comprising the steps of a) providing a liquid sample obtained from an individual, b) contacting said sample with a specific binding agent for FKBP52 under conditions appropriate for formation of a complex between said binding agent and FKBP52, and c) correlating the amount of complex formed in (b) to the diagnosis of breast cancer.
  • Another preferred embodiment of the invention is a method for the diagnosis of breast cancer comprising the steps of a) contacting a liquid sample obtained from an individual with a specific binding agent for FKBP52 under conditions appropriate for formation of a complex between said binding agent and FKBP52, and b) correlating the amount of complex formed in (a) to the diagnosis of breast cancer.
  • any such diagnosis is made in vitro.
  • the patient sample is discarded afterwards.
  • the patient sample is merely used for the in vitro diagnostic method of the invention and the material of the patient sample is not transferred back into the patient's body.
  • the sample is a liquid sample.
  • the FK506-binding protein 4 (FKBP52; Swiss-PROT: Q02790) is characterized by the sequence given in SEQ ID NO:l. This sequence translates to a theoretical molecular weight of 51,673 Da and to an isoelectric point at pH 5.23.
  • Ward et al. examined breast carcinoma cell lines for mRNA expression of FKBP52.
  • the mRNA expression correlated strongly with protein expression and was significantly higher in estrogen receptor (ER) alpha-positive compared with ER alpha-negative cell lines.
  • FKBP52 was overexpressed in breast tumors compared to matched normal controls (Ward, B.K., et al., Breast Cancer Research and Treatment 58 (1999) 267-280).
  • the immunophilin FKBP52 is associated with the unactivated estrogen receptor in mutually exclusive heterocomplexes and may differentially modulate receptor activity.
  • Kumar et al. showed that FKBP52 mRNA is differentially elevated in breast carcinomas compared with normal breast tissue. Other studies suggest that such alterations in the ratio of immunophilines might potentially influence steroid receptor functions. Therefore, Kumar et al. investigated the influence of estradiol on FKBP52 expression in MCF-7 breast cancer cell lines and found out that estradiol regulates the FKBP52 mRNA expression through both transscriptional and post-transcriptional mechanisms (Kumar, P., et al., Biochemical and Biophysical Research Communications 284 (2001) 219-225).
  • the unactivated steroid receptors are chaperoned into a confirmation that is optimal for binding hormones by a number of heat shock proteins and the immunophilin FKBP52. Together with its partner co-chaperones, cyclophilin 40 and FKBP51, FKBP52 belongs to a distinct group of structurally related immunophilins that modulate steroid receptor function through their association with heat shock protein 90 (Mark, P.J., et al., Cell Stress & Chaperones 6 (2001) 59-
  • Cheung-Flynn et al. identified the structural features that differentiate binding of FKBP51 and FKBP52 to heat shock protein 90 (Hsp 90). They found out that the final C-terminal 30 amino acids of FKBP51 enhance binding to Hsp 90, whereas the corresponding region of FKBP52 moderates binding to Hsp 90 (Cheung-Flynn, J. et al., Journal of Biological Chemistry 278 (2003) 17388-17294). Li et al. determined the crystal structure of the N-terminal domain of FKBP52. This domain is responsible for the peptidyl-prolyl isomerase activity and binding of the immunosuppressive drug FK 506 (Li, P., et al., Acta Crystallographica 59 (2003) 16- 22).
  • the present invention shall not be construed to be limited to the full-length protein FKBP52 of SEQ ID NO:l.
  • Physiological or artificial fragments of FKBP52, secondary modifications of FKBP52, as well as allelic variants of FKBP52 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 up a sandwich immunoassay.
  • novel marker FKBP52 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 FKBP52 are directly correlated to tumor burden. After chemotherapy a short term (few hours to 14 days) increase in FKBP52 may serve as an indicator of tumor cell death. In the follow-up of patients (from 3 months to 10 years) an increase of FKBP52 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 BC by measuring the level of FKBP52 and correlating the level measured to the presence or absence of BC.
  • 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 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).
  • TNM criteria are published by the UICC (International Union against Cancer)
  • T is , 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 18% for patients diagnosed when distant metastases are already present.
  • early diagnosis of BC refers to a diagnosis at a pre-cancerous state (DCIS) or at a tumor stage where no metastases at all (neither proximal nor distal), i.e., T ⁇ s , NO, M0 or Tl-4; NO; M0 are present.
  • T; s denotes carcinoma in situ.
  • the diagnostic method according to the present invention is based on a liquid sample which is derived from an individual. Unlike to methods known from the art FKBP52 is specifically measured from this liquid sample by use of a specific binding agent.
  • a specific binding agent is, e.g., a receptor for FKBP52, a lectin binding to FKBP52 or an antibody to FKBP52.
  • 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 target molecule.
  • specific is used to indicate that other biomolecules present in the sample do not significantly bind to with the binding agent specific for FKBP52.
  • the level of binding to a biomolecule 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 FKBP52.
  • 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 also be used.
  • Antibodies are generated by state of the art procedures, e.g., as described in Tijssen
  • monoclonal and polyclonal antibodies have been used.
  • Polyclonal antibodies have been raised in rabbits.
  • polyclonal antibodies from different species e.g. rats or guinea pigs can also be used.
  • Monoclonal antibodies have been produced using spleen cells from immunized mice. Since monoclonal antibodies can be produced in any amount required with constant properties, they represent ideal tools in development of an assay for clinical routine.
  • the generation and use of monoclonal antibodies to FKBP52 in a method according to the present invention is yet another preferred embodiment.
  • a synthetic peptide comprises a subsequence of SEQ ID NO:l which is specific for FKBP52, i.e., which has a comparatively low homology to other/related polypeptides. It is preferred that the synthetic peptide comprises a contiguous subsequence consisting of 5 to 25 amino acid residues of SEQ ID NO:l. More preferred, the peptide comprises a contiguous subsequence consisting of 10 to 15 amino acid residues of SEQ ID NO:l.
  • 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 FKBP52 under conditions appropriate for formation of a binding agent FKBP52-complex.
  • Such conditions need not be specified, 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 BC.
  • the skilled artisan will appreciate there are numerous methods to measure the amount of specific binding agent FKBP52-complex all described in detail in relevant textbooks (cf., e.g., Tijssen P., supra, or Diamandis et al., eds. (1996) Immunoassay,
  • FKBP52 is detected in a sandwich type assay format.
  • a first specific binding agent is used to capture FKBP52 on the one side and a second specific binding agent, which is labeled to be directly or indirectly detectable is used on the other side.
  • FKBP52 can be measured from a liquid sample obtained from an individual sample. No tissue and no biopsy sample is required to apply the marker FKBP52 in the diagnosis of BC.
  • 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. In a further preferred embodiment the method according to the present invention is practiced with whole blood as liquid sample material.
  • the method according to the present invention is practiced with nipple aspirate fluid as liquid sample material.
  • Antibodies to FKBP52 with great advantage can be used in established procedures, e.g., to detect breast cancer cells in situ, in biopsies, or in immunohistological procedures.
  • an antibody to FKBP52 is used in a qualitative (FKBP52 present or absent) or quantitative (FKBP52 amount is determined) immunoassay.
  • the present invention relates to use of protein FKBP52 as a marker molecule in the diagnosis of breast cancer from a liquid sample obtained from an individual.
  • marker molecule is used to indicate that an increased level of the analyte
  • FKBP52 as measured from a bodily fluid of an individual marks the presence of BC.
  • the use of protein FKBP52 itself represents a significant progress to the challenging field of BC diagnosis.
  • Combining measurements of FKBP52 with other known markers, e.g. CA 15-3 and CEA, or with other markers of BC presently known or yet to be discovered leads to further improvements. Therefore in a further preferred embodiment the present invention relates to the use of FKBP52 as a marker molecule for breast cancer in combination with one or more marker molecules for breast cancer in the diagnosis of breast 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 BC markers with which the measurement of FKBP52 may be combined are CEA and CA 15-3.
  • FKBP52 is used as part of a marker panel at least comprising FKBP52 and CA 15-3.
  • a further preferred embodiment of the present invention is the use of the protein FKBP52 as a marker molecule for breast cancer in combination with one or more marker molecules for breast cancer in the diagnosis of breast cancer from a liquid sample obtained from an individual, whereby the at least one other marker molecule is CA 15-3.
  • the inventive method is used with samples of patients suspected of suffering from breast cancer.
  • An individual suspected of suffering from breast cancer is an individual for which other types of cancers have been excluded.
  • Other cancers include but are not limited to cancers of the colon, lung, stomach, ovary, and prostate.
  • a preferred embodiment of the invention is therefore a method for the diagnosis of breast cancer comprising the steps of a) providing a liquid sample obtained from an individual suspected of suffering from breast cancer, b) contacting said sample with a specific binding agent for FKBP52 under conditions appropriate for formation of a complex between said binding agent and FKBP52, and c) correlating the amount of complex formed in (b) to the diagnosis of breast cancer.
  • 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 FKBP52 and auxiliary reagents for measurement of FKBP52.
  • 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/-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 FKBP52 has been assessed in comparison to and in combination with the established marker CA 15-3 using a receiver operator curve analysis (ROC; Zweig, M. H., and Campbell, G., Clin. Chem. 39 (1993) 561- 577).
  • ROC receiver operator curve analysis
  • This analysis has been based on well-defined patient cohorts consisting of 50 samples each from patients with invasive ductal or lobular carcinoma in Tl-3; NO; M0, more progressed tumor, i.e., T4 and/or various severity of metastasis (N+ and/or M+), medullary, papillary, mucinous and tubular carcinoma, ductal carcinoma in situ, 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).
  • the circle in the enlarged section of these gels indicates the position for the protein FKBP52.
  • this protein has not been detected in healthy tissue.
  • FKBP52 migrated in the 2D gel corresponding to an isoelectric point of about pH 5.4 and an apparent molecular weight of about 50 to 55 kDa.
  • tissue specimen from 14 patients suffering from breast cancer are analyzed.
  • Tumor tissue > 80% tumor
  • N adjacent healthy tissue
  • 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 [Roche Diagnostics GmbH, Mannheim, Germany, Cat. No. 1 873 580]
  • Freeze-dried CNBr-activated Sepharose 4B (Amersham Biosciences, 17-0430-01) is reswollen and washed according to the instructions of the manufacturer.
  • Monoclonal antibody directed against human albumin is dissolved in 0.1 M NaHCO 3 , pH 8.3, 0.5 M NaCl, 10 mg/ml.
  • 1 ml antibody solution is mixed with 1 ml reswollen CNBr-activated Sepharose 4B.
  • the reaction time is 1 h. Blocking of the remaining acitve groups and washing of the gel is carried out according to the instructions of the manufacturer.
  • sample buffer 7 M urea, 2 M fhiourea, 2% CHAPS, 0.4% IPG buffer pH 4-7, 0.5% DTT
  • sample buffer 7 M urea, 2 M fhiourea, 2% CHAPS, 0.4% IPG buffer pH 4-7, 0.5% DTT
  • 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, M ⁇ nchen, Germany) following the instructions of the supplier's manual.
  • Bio-Rad ® protein assay Cat.No. 500-0006; Bio-Rad Laboratories GmbH, M ⁇ nchen, Germany
  • IPG strips pH 4-7 (Amersham Biosciences, Freiburg, Germany) overnight.
  • the IEF is performed using the following gradient protocol: (1.) 1 minute to 500 V; (2.) 2 h to 3500 V; (3.) 22 h at constant 3500 V giving rise to 82 kVh. After IEF, strips are stored at -80°C or directly used for SDS-PAGE.
  • protein FKBP52 is found to be specifically expressed or strongly overexpressed in tumor tissue and not detectable in healthy control tissue. It therefore - amongst many other proteins
  • Polyclonal antibody to the breast cancer marker protein FKBP52 is generated for further use of the antibody in the measurement of serum and plasma and blood levels of FKBP52 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-FKBP52 fusion protein Purification of His-FKBP52 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-FKBP52 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- Turrax ® . 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.
  • Synthesis is carried out using heterobifunctional chemistry (maleimide/SH- chemistry). Selected cysteine containing FKBP52-peptides are coupled to 3- maleimidohexanoyl-N-hydroxysuccinimidester (MHS) activated hemocyanin from Concholepas concholepas (Sigma, B-8556).
  • MHS maleimidohexanoyl-N-hydroxysuccinimidester
  • Hemocyanin is brought to 10 mg/ml in 100 mM NaH 2 PO 4 /NaOH, pH 7.2. Per ml hemocyanin 100 ⁇ l MHS (12.3 mg in DMSO) are added and incubated for 1 h. The sample is dialyzed over night against 100 mM NaH 2 PO /NaOH, pH 6.5 and adjusted to 6 mg/ml with dialysis buffer. A selected cysteine containing FKBP52- peptide was dissolved in DMSO (5 mg/ml for a peptide of 1500 Dalton).
  • mice 12 week old A/J mice are initially immunized intraperitoneally with 100 ⁇ g FKBP52 or hemocyanin-peptide-conjugate (see above). This is followed after 6 weeks by two further intraperitoneal immunizations at monthly intervals. In this process each mouse is administered 100 ⁇ g FKBP52 or hemocyanin-peptide-conjugate adsorbed to aluminium hydroxide and 10 9 germs of Bordetella pertussis. Subsequently the last two immunizations are carried out intravenously on the 3rd and 2nd day before fusion using 100 ⁇ g FKBP52 or hemocyanin-peptide-conjugate 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. lxl 0 8 spleen cells of the immunized mouse are mixed with 2xl0 7 myeloma cells (P3X63-Ag8-653, ATCC CRL1580) and centrifuged (10 min at
  • the cells are then washed once with RPMI 1640 medium without foetal calf serum (FCS) and centrifuged again at 400 x 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 room temperature within a period of 4-5 min. Afterwards 5 ml RPMI 1640 containing 10% FCS is added drop-wise within ca.
  • FCS foetal calf serum
  • FKBP52- 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.
  • 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). Generation of polyclonal antibodies
  • a fresh emulsion of the protein solution (100 ⁇ g/ml FKBP52 or hemocyanin-peptide-conjugate) 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
  • IgG immunoglobulin G
  • 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 /NaOH, 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 M 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 M NaH 2 PO /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.
  • biotinylated primary antibody is diluted in SuperBlock Blocking Buffer (0.01-0.2 ⁇ g/ml) and incubated with the membrane for lh. The membranes are washed 3 times in PBS/0.05 % Tween-20.
  • the specifically bound biotinylated primary antibody is labeled with a streptavidin-
  • a sandwich ELISA For detection of FKBP52 in human serum or plasma, a sandwich ELISA is developed. For capture and detection of the antigen, aliquots of the anti-FKBP52 polyclonal antibody (see Example 2) are conjugated with biotin and digoxygenin, respectively.
  • Streptavidin-coated 96-well microtiter plates are incubated with 100 ⁇ l biotinylated anti-FKBP52 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
  • 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.
  • wells are incubated with 100 ⁇ l 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.
  • Accuracy is assessed by analyzing individual liquid samples obtained from well- characterized patient cohorts, i.e., 50 patients having undergone mammography and found to be free of BC, 50 patients each diagnosed and staged as invasive ductal and invasive lobular Tl-3, NO, M0 of BC, 50 patients diagnosed with progressed
  • CA 15-3 as measured by a commercially available assay (Roche Diagnostics, CA 15-3-assay (Cat. No. 0 304 5838 for Elecsys ® Systems immunoassay analyzer) and FKBP52 measured as described above have been 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 Ti S -3, NO, M0 from healthy individuals for the combination of FKBP52 with the established marker CA 15-3 is calculated by regularized discriminant analysis (Friedman, J. H., Regularized Discriminant Analysis, Journal of the American Statistical Association 84 (1989) 165-175).

Abstract

The present invention relates to the diagnosis of breast cancer. It discloses the use of protein FK506-binding protein 4 (FKBP52) in the diagnosis of breast cancer. It relates to a method for diagnosis of breast cancer from a liquid sample, derived from an individual by measuring FKBP52 in said sample. Measurement of FKBP52 can, e.g., be used in the early detection or diagnosis of breast cancer.

Description

Use of protein FKBP52 as a marker for breast cancer
The present invention relates to the diagnosis of breast cancer. It discloses the use of FK506-binding protein 4 (=FKBP52) in the diagnosis of breast cancer. Furthermore, it especially relates to a method for diagnosis of breast cancer from a liquid sample, derived from an individual by measuring FKBP52 in said sample. Measurement of FKBP52 can, e.g., be used in the early detection or diagnosis of breast cancer.
Cancer remains a major public health challenge despite progress in detection and therapy. Amongst the various types of cancer, breast cancer (=BC) is one of the most frequent cancers among women in the Western world.
The earlier cancer can be detected/diagnosed, the better is the overall survival rate. This is especially true for BC. 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 BC as a public health problem, it is essential that more effective screening and preventative measures for breast cancer will be developed.
The earliest detection procedures available at present for breast cancer involve using clinical breast examination and mammography. However, significant tumor size must typically exist before a tumor is palpable or can be detected by a mammogram. The density of the breast tissue and the age are important predictors of the accuracy of screening mammography. The sensitivity ranges from 63% in women with extremely dense breasts to 87% in women with almost entirely fatty- breasts. The sensitivity increases with age from 69% in women of about 40 years of age to 83% in women 80 years and older (Carney, P.A., et al., Ann. Intern. Med. 138 (3) (2003) 168-175). Only 20 - 25 % of mammographically detected abnormalities that are biopsied prove to be malignant. The visualization of precancerous and cancerous lesions represents the best approach to early detection, but mammography is an expensive test that requires great care and expertise both to perform and in the interpretation of results (WHO, Screening for Breast Cancer, May 10, 2002; Esserman, L., et al., I. Natl. Cancer Inst. 94 (2002) 369-375).
In the recent years a tremendous amount of so-called breast specific or even so- called breast 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 breast (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 00/60076 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 BC. 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 (Chen, G., et al., Molecular and Cellular Proteomics, 1.4 (2002) 304- 313). 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 BC. Wulfkuhle et al. Cancer Research 62 (2002) 6740-6749 have identified fifty-seven proteins which were differentially expressed between BC tissue and adjacent normal tissue. No data from liquid samples obtained from an individual are reported.
WO 02/23200 reports about twelve breast 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 BC as compared to sera obtained from healthy controls. However, the identity of the molecule(s) comprised in such spot, e.g their sequence, is not known.
Nipple aspirate fluid (NAF) has been used for many years as a potential non- invasive method to identify breast cancer-specific markers. Kuerer et al. compared bilateral matched pair nipple aspirate fluids from women with unilateral invasive breast carcinoma by 2D gel electrophoresis (Kuerer, H.M., et al, Cancer 95 (2002) 2276-2282). 30 to 202 different protein spots were detected in the NAF of breasts suffering from breast carcinoma and not in the matched NAF of the healthy breasts. These spots were detected by a gel image analysis. But the identity of the protein spots is not known.
Despite the large and ever growing list of candidate protein markers in the field of BC, 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 cancer antigen 15-
3 (CA 15-3), a tumor-associated mucin, and carcinoembryonic antigen (CEA), a tumor associated glycoprotein, are available to assist diagnosis in the field of BC. CA 15-3 is usually increased in patients with advanced breast cancer. CA 15-3 levels are rarely elevated in women with early stage breast cancer (Duffy, M.J., Critical Reviews in Clinical Laboratory Sciences 38 (2001) 225-262). Cancers of the ovary, lung and prostate may also raise CA 15-3 levels. Elevated levels of CA 15-3 may be associated with non-cancerous conditions, such as benign breast or ovary disease, endometriosis, pelvic inflammatory disease, and hepatitis. Pregnancy and lactation can also cause CA 15-3 levels to raise (National Cancer Institute, Cancer Facts, Fact Sheet 5.18 (1998) 1-5). The primary use of CEA is in monitoring colon cancer, especially when the disease has metastasized. However, a variety of cancers can produce elevated levels of CEA, including breast cancer.
Due to the lack of organ and tumor specificity, neither measurement of CA 15-3 nor measurement of CEA are recommended for screening of BC. These tumor markers are helpful diagnostic tools in follow-up care of BC patients (Untch, M., et al., J. Lab. Med. 25 (2001) 343-352). Whole blood, serum, plasma, or nipple aspirate fluid are the most widely used sources of sample in clinical routine. The identification of an early BC 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 BC from blood. It is especially important to improve the early diagnosis of BC, 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 BC diagnosis.
Surprisingly, it has been found that use of the marker FKBP52 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 breast cancer comprising the steps of a) providing a liquid sample obtained from an individual, b) contacting said sample with a specific binding agent for FKBP52 under conditions appropriate for formation of a complex between said binding agent and FKBP52, and c) correlating the amount of complex formed in (b) to the diagnosis of breast cancer.
Another preferred embodiment of the invention is a method for the diagnosis of breast cancer comprising the steps of a) contacting a liquid sample obtained from an individual with a specific binding agent for FKBP52 under conditions appropriate for formation of a complex between said binding agent and FKBP52, and b) correlating the amount of complex formed in (a) to the diagnosis of breast cancer.
As the skilled artisan will appreciate, any such diagnosis is made in vitro. The patient sample is discarded afterwards. The patient sample is merely used for the in vitro diagnostic method of the invention and the material of the patient sample is not transferred back into the patient's body. Typically, the sample is a liquid sample. The FK506-binding protein 4 (FKBP52; Swiss-PROT: Q02790) is characterized by the sequence given in SEQ ID NO:l. This sequence translates to a theoretical molecular weight of 51,673 Da and to an isoelectric point at pH 5.23.
Ward et al. examined breast carcinoma cell lines for mRNA expression of FKBP52. The mRNA expression correlated strongly with protein expression and was significantly higher in estrogen receptor (ER) alpha-positive compared with ER alpha-negative cell lines. FKBP52 was overexpressed in breast tumors compared to matched normal controls (Ward, B.K., et al., Breast Cancer Research and Treatment 58 (1999) 267-280).
The immunophilin FKBP52 is associated with the unactivated estrogen receptor in mutually exclusive heterocomplexes and may differentially modulate receptor activity. Kumar et al. showed that FKBP52 mRNA is differentially elevated in breast carcinomas compared with normal breast tissue. Other studies suggest that such alterations in the ratio of immunophilines might potentially influence steroid receptor functions. Therefore, Kumar et al. investigated the influence of estradiol on FKBP52 expression in MCF-7 breast cancer cell lines and found out that estradiol regulates the FKBP52 mRNA expression through both transscriptional and post-transcriptional mechanisms (Kumar, P., et al., Biochemical and Biophysical Research Communications 284 (2001) 219-225).
The unactivated steroid receptors are chaperoned into a confirmation that is optimal for binding hormones by a number of heat shock proteins and the immunophilin FKBP52. Together with its partner co-chaperones, cyclophilin 40 and FKBP51, FKBP52 belongs to a distinct group of structurally related immunophilins that modulate steroid receptor function through their association with heat shock protein 90 (Mark, P.J., et al., Cell Stress & Chaperones 6 (2001) 59-
70).
Cheung-Flynn et al. identified the structural features that differentiate binding of FKBP51 and FKBP52 to heat shock protein 90 (Hsp 90). They found out that the final C-terminal 30 amino acids of FKBP51 enhance binding to Hsp 90, whereas the corresponding region of FKBP52 moderates binding to Hsp 90 (Cheung-Flynn, J. et al., Journal of Biological Chemistry 278 (2003) 17388-17294). Li et al. determined the crystal structure of the N-terminal domain of FKBP52. This domain is responsible for the peptidyl-prolyl isomerase activity and binding of the immunosuppressive drug FK 506 (Li, P., et al., Acta Crystallographica 59 (2003) 16- 22).
As obvious to the skilled artisan, the present invention shall not be construed to be limited to the full-length protein FKBP52 of SEQ ID NO:l. Physiological or artificial fragments of FKBP52, secondary modifications of FKBP52, as well as allelic variants of FKBP52 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 up a sandwich immunoassay.
In preferred embodiments, the novel marker FKBP52 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 FKBP52 are directly correlated to tumor burden. After chemotherapy a short term (few hours to 14 days) increase in FKBP52 may serve as an indicator of tumor cell death. In the follow-up of patients (from 3 months to 10 years) an increase of FKBP52 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 BC by measuring the level of FKBP52 and correlating the level measured to the presence or absence of BC.
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). The staging system for breast cancer has recently been revised (Singletary, S.E., et al., Journal of Clinical Oncology 20 (2002) 3628-3636).
What is especially important is, that early diagnosis of BC translates to a much better prognosis. Therefore, best prognosis have those patients 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 18% for patients diagnosed when distant metastases are already present.
In the sense of the present invention early diagnosis of BC refers to a diagnosis at a pre-cancerous state (DCIS) or at a tumor stage where no metastases at all (neither proximal nor distal), i.e., Tιs, NO, M0 or Tl-4; NO; M0 are present. T;s denotes carcinoma in situ.
In a preferred embodiment FKBP52 is used to diagnose BC in a non-metastatic stage, i.e., that diagnosis is made at stage Tis, NO, M0 or Tl-3; NO; M0 (=Tis-3; NO; M0).
The diagnostic method according to the present invention is based on a liquid sample which is derived from an individual. Unlike to methods known from the art FKBP52 is specifically measured from this liquid sample by use of a specific binding agent.
A specific binding agent is, e.g., a receptor for FKBP52, a lectin binding to FKBP52 or an antibody to FKBP52. 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 target 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 FKBP52. Preferably, the level of binding to a biomolecule 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 FKBP52. 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 also 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. (Tijssen, P., supra, pages 108-115).
For the achievements as disclosed in the present invention monoclonal and polyclonal antibodies have been used. Polyclonal antibodies have been raised in rabbits. However, clearly also polyclonal antibodies from different species , e.g. rats or guinea pigs can also be used. Monoclonal antibodies have been produced using spleen cells from immunized mice. Since monoclonal antibodies can be produced in any amount required with constant properties, they represent ideal tools in development of an assay for clinical routine. The generation and use of monoclonal antibodies to FKBP52 in a method according to the present invention is yet another preferred embodiment.
As the skilled artisan will appreciate now, that FKBP52 has been identified as a marker which is useful in the diagnosis of BC, alternative ways maybe 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 FKBP52 for immunization. Preferably, a synthetic peptide comprises a subsequence of SEQ ID NO:l which is specific for FKBP52, i.e., which has a comparatively low homology to other/related polypeptides. It is preferred that the synthetic peptide comprises a contiguous subsequence consisting of 5 to 25 amino acid residues of SEQ ID NO:l. More preferred, the peptide comprises a contiguous subsequence consisting of 10 to 15 amino acid residues of SEQ ID NO:l.
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 FKBP52 under conditions appropriate for formation of a binding agent FKBP52-complex. Such conditions need not be specified, 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 invention the amount of complex is measured and correlated to the diagnosis of BC. As the skilled artisan will appreciate there are numerous methods to measure the amount of specific binding agent FKBP52-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 FKBP52 is detected in a sandwich type assay format. In such assay a first specific binding agent is used to capture FKBP52 on the one side and a second specific binding agent, which is labeled to be directly or indirectly detectable is used on the other side.
As mentioned above, it has surprisingly been found that FKBP52 can be measured from a liquid sample obtained from an individual sample. No tissue and no biopsy sample is required to apply the marker FKBP52 in the diagnosis of BC.
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.
In a further preferred embodiment the method according to the present invention is practiced with nipple aspirate fluid as liquid sample material.
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 FKBP52 in a bodily fluid sample. Even more surprising they have been able to demonstrate that the presence of FKBP52 in such liquid sample obtained from an individual can be correlated to the diagnosis of breast cancer.
Antibodies to FKBP52 with great advantage can be used in established procedures, e.g., to detect breast cancer cells in situ, in biopsies, or in immunohistological procedures.
Preferably, an antibody to FKBP52 is used in a qualitative (FKBP52 present or absent) or quantitative (FKBP52 amount is determined) immunoassay.
Measuring the level of protein FKBP52 has proven very advantageous in the field of BC. Therefore, in a further preferred embodiment, the present invention relates to use of protein FKBP52 as a marker molecule in the diagnosis of breast cancer from a liquid sample obtained from an individual.
The term marker molecule is used to indicate that an increased level of the analyte
FKBP52 as measured from a bodily fluid of an individual marks the presence of BC.
It is especially preferred to use the novel marker FKBP52 in the early diagnosis of breast cancer.
The use of protein FKBP52 itself, represents a significant progress to the challenging field of BC diagnosis. Combining measurements of FKBP52 with other known markers, e.g. CA 15-3 and CEA, or with other markers of BC presently known or yet to be discovered, leads to further improvements. Therefore in a further preferred embodiment the present invention relates to the use of FKBP52 as a marker molecule for breast cancer in combination with one or more marker molecules for breast cancer in the diagnosis of breast 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 BC markers with which the measurement of FKBP52 may be combined are CEA and CA 15-3. Most preferred, FKBP52 is used as part of a marker panel at least comprising FKBP52 and CA 15-3. Thus, a further preferred embodiment of the present invention is the use of the protein FKBP52 as a marker molecule for breast cancer in combination with one or more marker molecules for breast cancer in the diagnosis of breast cancer from a liquid sample obtained from an individual, whereby the at least one other marker molecule is CA 15-3.
Preferably, the inventive method is used with samples of patients suspected of suffering from breast cancer. An individual suspected of suffering from breast cancer is an individual for which other types of cancers have been excluded. Other cancers include but are not limited to cancers of the colon, lung, stomach, ovary, and prostate. A preferred embodiment of the invention is therefore a method for the diagnosis of breast cancer comprising the steps of a) providing a liquid sample obtained from an individual suspected of suffering from breast cancer, b) contacting said sample with a specific binding agent for FKBP52 under conditions appropriate for formation of a complex between said binding agent and FKBP52, and c) correlating the amount of complex formed in (b) to the diagnosis of breast cancer.
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 FKBP52 and auxiliary reagents for measurement of FKBP52.
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/-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 FKBP52 has been assessed in comparison to and in combination with the established marker CA 15-3 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 with invasive ductal or lobular carcinoma in Tl-3; NO; M0, more progressed tumor, i.e., T4 and/or various severity of metastasis (N+ and/or M+), medullary, papillary, mucinous and tubular carcinoma, ductal carcinoma in situ, and healthy controls, respectively.
The following examples, references, sequence listing and figure 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 Figure
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). The circle in the enlarged section of these gels indicates the position for the protein FKBP52. Using the same method this protein has not been detected in healthy tissue. FKBP52 migrated in the 2D gel corresponding to an isoelectric point of about pH 5.4 and an apparent molecular weight of about 50 to 55 kDa.
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
UICC International Union Against Cancer
Example 1
Identification of FKBP52 as a potential breast cancer marker
Sources of tissue
In order to identify tumor-specific proteins as potential diagnostic markers for breast cancer, analysis of two different kinds of tissue is performed using proteomics methods.
In total, tissue specimen from 14 patients suffering from breast cancer are analyzed.
From each patient two different tissue types are collected from therapeutic resections: Tumor tissue (> 80% tumor) (T), and adjacent healthy tissue (N). The latter tissue type serves as matched healthy control sample. 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.
Immobilization of monoclonal antibody anti-human albumin on CNBr-activated Sepharose 4B
Freeze-dried CNBr-activated Sepharose 4B (Amersham Biosciences, 17-0430-01) is reswollen and washed according to the instructions of the manufacturer. Monoclonal antibody directed against human albumin is dissolved in 0.1 M NaHCO3, pH 8.3, 0.5 M NaCl, 10 mg/ml. 1 ml antibody solution is mixed with 1 ml reswollen CNBr-activated Sepharose 4B. The reaction time is 1 h. Blocking of the remaining acitve groups and washing of the gel is carried out according to the instructions of the manufacturer.
Depletion of serum albumin
7 ml anti-albumin gel is equilibrated in lysis buffer without Genapol X-080. 7 ml of the upper fraction of the sucrose-density centrifugation (see above, tissue preparation) are applied onto the column and washed through with lysis buffer without Genalpol X-080. The combined effluent is used for the isoelectric focussing experiments. Isoelectric focussing (IFF) and SDS-PAGE
For IEF, 3 ml of the HSA-depleted tissue preparation are mixed with 12 ml sample buffer (7 M urea, 2 M fhiourea, 2% CHAPS, 0.4% IPG buffer pH 4-7, 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 g of protein sample buffer is added to a final volume of 350 μl. This solution is used to rehydrate IPG strips pH 4-7 (Amersham Biosciences, Freiburg, Germany) overnight. The IEF is performed using the following gradient protocol: (1.) 1 minute to 500 V; (2.) 2 h to 3500 V; (3.) 22 h at constant 3500 V giving rise to 82 kVh. 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 alle iation 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 and thereafter 1 h at 17 W/gel. Subsequently, the gels are fixed (50% methanol, 10% acetate) and stained overnight with Novex Colloidal Blue Staining Kit (Invitrogen, Karlsruhe, Germany, Cat No. LC6025, 45-7101)
Detection of FKBP52 as a potential marker for breast 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 tissue and analyzed for distinctive spots corresponding to differentially expressed proteins. By this means, protein FKBP52 is found to be specifically expressed or strongly overexpressed in tumor tissue and not detectable in healthy control tissue. It therefore - amongst many other proteins
- qualifies as a candidate marker for use in the diagnosis of breast cancer. Example 2
Generation of antibodies to the breast cancer marker protein FKBP52
Polyclonal antibody to the breast cancer marker protein FKBP52 is generated for further use of the antibody in the measurement of serum and plasma and blood levels of FKBP52 by immunodetection assays, e.g. Western Blotting and ELISA
Recombinant protein expression and purification
In order to generate antibodies to FKBP52, 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 for in-vitro transcription and expression of the nucleotide sequence coding for the FKBP52 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-FKBP52 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-FKBP52 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- Turrax®. 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.
Synthesis of hemocyanin-peptide-conjugates for the generation of antibodies
Synthesis is carried out using heterobifunctional chemistry (maleimide/SH- chemistry). Selected cysteine containing FKBP52-peptides are coupled to 3- maleimidohexanoyl-N-hydroxysuccinimidester (MHS) activated hemocyanin from Concholepas concholepas (Sigma, B-8556).
Hemocyanin is brought to 10 mg/ml in 100 mM NaH2PO4/NaOH, pH 7.2. Per ml hemocyanin 100 μl MHS (12.3 mg in DMSO) are added and incubated for 1 h. The sample is dialyzed over night against 100 mM NaH2PO /NaOH, pH 6.5 and adjusted to 6 mg/ml with dialysis buffer. A selected cysteine containing FKBP52- peptide was dissolved in DMSO (5 mg/ml for a peptide of 1500 Dalton). Per ml MHS-activated hemocyanin (6 mg/ml) 20 μl of 100 mM EDTA, pH 7.0 and 100 μl of the selected cysteine containing FKBP52 -peptide are added. After 1 h the remaining maleimide groups are blocked by the addition of 10 μl 0.5 M cysteine/HCl per ml reaction mixture. This preparation is used for immunization without further purification.
Production of monoclonal antibodies against FKBP52
a) Immunization of mice
12 week old A/J mice are initially immunized intraperitoneally with 100 μg FKBP52 or hemocyanin-peptide-conjugate (see above). This is followed after 6 weeks by two further intraperitoneal immunizations at monthly intervals. In this process each mouse is administered 100 μg FKBP52 or hemocyanin-peptide-conjugate adsorbed to aluminium hydroxide and 109 germs of Bordetella pertussis. Subsequently the last two immunizations are carried out intravenously on the 3rd and 2nd day before fusion using 100 μg FKBP52 or hemocyanin-peptide-conjugate 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. lxl 08 spleen cells of the immunized mouse are mixed with 2xl07 myeloma cells (P3X63-Ag8-653, ATCC CRL1580) and centrifuged (10 min at
300 x g and 4°C). The cells are then washed once with RPMI 1640 medium without foetal calf serum (FCS) and centrifuged again at 400 x 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 room 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 (lOO 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. FKBP52- 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 FKBP52 or hemocyanin-peptide-conjugate) 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-FKBP52 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 /NaOH, 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 M 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 M NaH2PO /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 blot for the detection of FKBP52 in human serum and plasma samples.
SDS-PAGE and Western Blotting are carried out using reagents and equipment of Invitrogen, Karlsruhe, Germany. Human plasma samples are diluted 1:20 in reducing NuPAGE® (Invitrogen) LDS sample buffer and heated for 5 min at 95°C. 10 μl aliquots are run on 4-12 % NuPAGE® gels (Bis-Tris) 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 3 times in
PBS/0.05 % Tween-20 and blocked with SuperBlock Blocking Buffer (Pierce Biotechnology, Inc., Rockford, IL, USA). The biotinylated primary antibody is diluted in SuperBlock Blocking Buffer (0.01-0.2 μg/ml) and incubated with the membrane for lh. The membranes are washed 3 times in PBS/0.05 % Tween-20. The specifically bound biotinylated primary antibody is labeled with a streptavidin-
HRP-conjugate (20 mUABTs ml in SuperBlock Blocking Buffer). After incubation for 1 h, the membranes are washed 3 times in PBS/0.05 % Tween-20. The bound streptavidin-HRP-conjugate is detected using a chemiluminescent substrate (SuperSignal West Femto Substrate, Pierce Biotechnology, Inc., Rockford, IL, USA) and autoradiographic film. Exposure times varies from 10 min to over night. Example 4
ELISA for the measurement of FKBP52 in human serum and plasma samples.
For detection of FKBP52 in human serum or plasma, a sandwich ELISA is developed. For capture and detection of the antigen, aliquots of the anti-FKBP52 polyclonal antibody (see Example 2) are conjugated with biotin and digoxygenin, respectively.
Streptavidin-coated 96-well microtiter plates are incubated with 100 μl biotinylated anti-FKBP52 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 FKBP52, plates are washed three times with 0.9% NaCl , 0.1% Tween-20. For specific detection of bound FKBP52, wells are incubated with 100 μl of digoxygenylated anti-FKBP52 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 mammography and found to be free of BC, 50 patients each diagnosed and staged as invasive ductal and invasive lobular Tl-3, NO, M0 of BC, 50 patients diagnosed with progressed
BC, having at least tumor infiltration in at least one proximal lymph node or more severe forms of metastasis, 50 patients each diagnosed with medullary, mucinous, tubular, or papillary breast carcinoma, and 50 patients diagnosed with DCIS, respectively. CA 15-3 as measured by a commercially available assay (Roche Diagnostics, CA 15-3-assay (Cat. No. 0 304 5838 for Elecsys® Systems immunoassay analyzer) and FKBP52 measured as described above have been 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 TiS-3, NO, M0 from healthy individuals for the combination of FKBP52 with the established marker CA 15-3 is calculated by regularized discriminant analysis (Friedman, J. H., Regularized Discriminant Analysis, Journal of the American Statistical Association 84 (1989) 165-175).
Preliminary data indicate that FKBP52 may also be very helpful in the follow-up of patients after surgery.
List of References
Bruck, C, and Chen, G., et al., Methods Enzymol. 121 (1986) 587-695
Carney, P.A., et al., Ann. Intern. Med. 138 (2003) 168-175 Chen, G., et al., Molecular and Cellular Proteomics, 1.4 (2002) 304-313
Cheung-Flynn, J. et al., Journal of Biological Chemistry 278 (2003) 17388-17294
Diamandis, et al., eds. (1996) Immunoassay, Academic Press, Boston
Duffy, M.J., Critical Reviews in Clinical Laboratory Sciences 38 (2001) 225-262
Essermann, L., et al., J. Natl. Cancer Inst. 94 (2002) 369-375 Galfre, G., and Milstein, C, Methods Enzymol. 73 (1981) 3-46
Kuerer, H.M., et al., Cancer 95 (2002) 2276-2282
Kumar, P., et al., Biochemical and Biophysical Research Communications 284 (2001) 219-225
Li, P., et al., Acta Crystallographica 59 (2003) 16-22 Mark, P.J., et al., Cell Stress & Chaperones 6 (2001) 59-70
National Cancer Institute, Cancer Facts, Fact Sheet 5.18 (1998) 1-5
Singletary, S.E., et al. Journal of Clinical Oncology 20 (2002) 3628-3636
Studier, F.W., et al., Methods Enzymol. 185 (1990) 60-89
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
Untch, M., et al., J. Lab. Med. 25 (2001) 343-352
Ward, B. K., et al., Breast Cancer Research and Treatment 58 (1999) 267-280 WHO, Screening for Breast Cancer, May 10, 2002
WO 00/60076
WO 02/23200
Wulfkuhle, J.D., et al., Cancer Research 62 (2002) 6740-6749
Zweig, M. H., and Campbell, G., Clin. Chem. 39 (1993) 561-577

Claims

Patent Claims
1. A method for the diagnosis of breast cancer comprising the steps of a) providing a liquid sample obtained from an individual, b) contacting said sample with a specific binding agent for FK506-binding protein 4 (FKBP52) under conditions appropriate for formation of a complex between said binding agent and FKBP52, and c) correlating the amount of complex formed in (b) to the diagnosis of breast 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. The method according to claim 1, further characterized in that said sample is nipple aspirate fluid.
6. Use of protein FKBP52 as a marker molecule in the diagnosis of breast cancer from a liquid sample obtained from an individual.
7. Use of protein FKBP52 as a marker molecule in the early diagnosis of breast cancer from a liquid sample obtained from an individual.
8. Use according to claim 7, wherein the early diagnosis is made with a sample derived from BC patients in stage Tιs -3; NO; M0.
9. Use of protein FKBP52 as a marker molecule for breast cancer in combination with one or more marker molecules for breast cancer in the diagnosis of breast cancer from a liquid sample obtained from an individual.
10. Use according to claim 9, wherein the at least one other marker molecule is CA 15-3.
11. An immunological kit comprising at least one specific binding agent for FKBP52 and auxiliary reagents for measurement of FKBP52.
PCT/EP2004/011602 2003-10-15 2004-10-15 Use of protein fkbp52 as a marker for breast cancer WO2005040808A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP03023502 2003-10-15
EP03023502.2 2003-10-15

Publications (1)

Publication Number Publication Date
WO2005040808A1 true WO2005040808A1 (en) 2005-05-06

Family

ID=34486064

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2004/011602 WO2005040808A1 (en) 2003-10-15 2004-10-15 Use of protein fkbp52 as a marker for breast cancer

Country Status (1)

Country Link
WO (1) WO2005040808A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015092186A (en) * 2009-09-24 2015-05-14 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Fkbp52-tau interaction as novel therapeutical target for treating neurological disorders involving tau dysfunction

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DUFFY M J: "Clinical uses of tumor markers: A critical review", CRITICAL REVIEWS IN CLINICAL LABORATORY SCIENCES, vol. 38, no. 3, June 2001 (2001-06-01), pages 225 - 262, XP009042383, ISSN: 1040-8363 *
LE BIHAN S ET AL: "Calcium/calmodulin kinase inhibitors and immunosuppressant macrolides rapamycin and FK506 inhibit progestin- and glucocorticosteroid receptor-mediated transcription in human breast cancer T47D cells.", MOLECULAR ENDOCRINOLOGY (BALTIMORE, MD.) JUL 1998, vol. 12, no. 7, July 1998 (1998-07-01), pages 986 - 1001, XP001204597, ISSN: 0888-8809 *
MILAD M ET AL: "Interaction of the progesterone receptor with binding proteins for FK506 and cyclosporin A.", MOLECULAR ENDOCRINOLOGY (BALTIMORE, MD.) JUL 1995, vol. 9, no. 7, July 1995 (1995-07-01), pages 838 - 847, XP001204592, ISSN: 0888-8809 *
WARD B K ET AL: "Expression of the estrogen receptor-associated immunophilins, cyclophilin 40 and FKBP52, in breast cancer.", BREAST CANCER RESEARCH AND TREATMENT. DEC 1999, vol. 58, no. 3, December 1999 (1999-12-01), pages 267 - 280, XP001204594, ISSN: 0167-6806 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015092186A (en) * 2009-09-24 2015-05-14 アンスティチュ ナショナル ドゥ ラ サンテ エ ドゥ ラ ルシェルシュ メディカル Fkbp52-tau interaction as novel therapeutical target for treating neurological disorders involving tau dysfunction

Similar Documents

Publication Publication Date Title
US7566542B2 (en) Use of protein ASC as a marker for breast cancer
EP1579220A2 (en) Use of nicotinamide n-methyltransferase as a marker for colorectal cancer
US20070196844A1 (en) Protein PDX1 as a marker for breast cancer
US20060188949A1 (en) Use of protein PLST as a marker for colorectal cancer
US7579158B2 (en) Cellular retinoic acid binding protein II as a marker for breast cancer
US20060121540A1 (en) Use of protein MASP as a marker for colorectal cancer
US20060257951A1 (en) Use of protein spee as a marker for breast cancer
US20060257950A1 (en) Use of protein UBC13 as a marker for breast cancer
WO2005040809A1 (en) Use of protein hnrnp-f as a marker for breast cancer
US20060188950A1 (en) Use of protein spee as a marker for colorectal cancer
WO2005040808A1 (en) Use of protein fkbp52 as a marker for breast cancer
WO2005040805A1 (en) Use of protein masp as a marker for breast cancer
WO2005040810A1 (en) Use of protein hnrnp-k as a marker for breast cancer
WO2005040811A1 (en) Use of protein tip47 as a marker for breast cancer
WO2005015220A1 (en) Use of protein crabp-i as a marker for breast cancer
WO2005050217A1 (en) Use of protein pspase as a marker for breast cancer
WO2005040807A2 (en) Swiprosin-2 as a breast cancer marker
US20070218510A1 (en) Use of protein PSA3 as a marker for colorectal cancer
WO2005124357A1 (en) Use of protein elongation factor-1-alpha-1 as a marker for breast cancer
WO2005124360A2 (en) Use of protein tebp as a marker for breast cancer
WO2005015222A1 (en) Use of the far upstream element (fuse) binding protein (fubp) as a marker for colorectal cancer
WO2005015230A1 (en) Use of protein psa3 as a marker for colorectal cancer
WO2005124354A2 (en) Use of protein ch10 as a marker for breast 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 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): BW GH 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 IT 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
122 Ep: pct application non-entry in european phase