CA2369892A1 - Method for quantifying transforming growth factor-.beta.1 and method for detecting cancer by using same - Google Patents

Method for quantifying transforming growth factor-.beta.1 and method for detecting cancer by using same Download PDF

Info

Publication number
CA2369892A1
CA2369892A1 CA002369892A CA2369892A CA2369892A1 CA 2369892 A1 CA2369892 A1 CA 2369892A1 CA 002369892 A CA002369892 A CA 002369892A CA 2369892 A CA2369892 A CA 2369892A CA 2369892 A1 CA2369892 A1 CA 2369892A1
Authority
CA
Canada
Prior art keywords
tgf
beta
receptor
antibody
sample
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002369892A
Other languages
French (fr)
Inventor
Hoon Shin
Chang Gi Lim
Seung Won Jin
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Hanmi Pharmaceutical Co Ltd
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of CA2369892A1 publication Critical patent/CA2369892A1/en
Abandoned legal-status Critical Current

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
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • 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
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57488Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds identifable in body fluids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/475Assays involving growth factors
    • G01N2333/495Transforming growth factor [TGF]

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Urology & Nephrology (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Hematology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Medicinal Chemistry (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Microbiology (AREA)
  • Endocrinology (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The amount of TGF-.beta.1 in a sample is quantified by treating the sample with a TGF-.beta.1 specific receptor to from a complex between TGF-.beta.1 a nd the receptor and measuring the amount of the complex.

Description

METHOD FOR QUANTIFYING TRANSFORMING GROWTH FACTOR-~31 AND
METHOD FOR DETECTING CANCER BY USING SAME
FIELD OF THE INVENTION
The present invention relates to a method for quantifying the concentration of transforming growth factor-(31 (TGF-X31) in a body fluid, a method for detecting cancer by using same, a composition for detecting cancer, and a TGF-(31-specific monoclonal antibody.
BACKGROUND OF THE INVENTION
Transforming growth factor-(3 (TGF-(3) regulates the growth and differentiation of several cells, its mode of action depending on the cell configuration and the presence of other growth factors(Sporn et al., Science, 233, 532-534 (1986); and Roberts and Sporn, Adv. Cancer Res., ~l_, 107-145 (1988)).
Three forms of TGF-(3 factor, TGF-(31, -(32 and -~i3, occur in mammals, and, among these, TGF-(31 is believed to play a key role in the physiological mechanism and disease progression. It has been reported that it acts abnormally in an invasion process, e.g., carcinogenesis.
This suggests that TGF-(31 is useful as a tumor marker in cancer diagnosis, and that a method for quantifying TGF-(31 in a body fluid with a high precision can be critical in cancer diagnosis.
EP Publication No. 0 722 773 A1 discloses a method for detecting cancer by contacting a blood sample containing TGF-(31 with an absorbent, OH-carbonated hydroxyapatite, to adsorb TGF-(31 thereto, eluting the absorbed TGF-(31 with a buffer, and determining the amount of TGF-(31 eluted with UV spectrometry. However, this method suffers from the problems of limited sensitivity and imprecision manifested by large fluctuations in measured values.
Therefore, there has existed a need to develop an improved method for quantifying the amount of TGF-(31 in plasma.
S~TMMARY OF THE INVENTION
Accordingly, it is an object of the present invention to provide a method for quantifying the amount of TGF-(31 in a sample with high precision and sensitivity.
Another object of the present invention is to provide a method for detecting cancer by using said method .
A further object of the present invention is to provide a composition for detecting cancer.
A still further object of the present invention is to provide a TGF-(31-specific monoclonal antibody and a hybridoma cell line producing the monoclonal antibody.
In accordance with one aspect of the present invention, there is provided a method for quantifying the amount of TGF-ail in a sample which comprises treating the sample with a TGF-(31-specific receptor to form a complex between TGF-(31 and the receptor and measuring the amount of the complex.
BRIEF DESCRIPTION OF THE DRAWINGS
The above objects and features of the present invention will become apparent from the following description of preferred embodiments taken in conjunction with the accompanying drawings, in which:
Fig. 1 shows the optical density-TGF-ail concentration correlations obtained in Example 1 for TGF-(31 type III and type II receptors, respectively;
Fig. 2 depicts the respective distributions of TGF-(31 concentrations in plasma samples taken from healthy persons, stomach cancer patients, hepatoma patients and breast cancer patients; and Fig. 3 provides the respective distributions of TGF-(31 concentrations in plasma samples taken from healthy persons, lung cancer patients, rectal-colic cancer patients, prostate cancer patients and uterine cervical cancer patients.
DETAILED DESCRIPTION OF THE INVENTION
The TGF-(31-specific receptors which may be used in the present invention include TGF-(31 type I, II and III
receptors (RI , RII and RIII ) , and preferred is TGF-X31 type III receptor, RIII. The TGF-(31 receptors may be obtained by expressing a TGF-(31 receptor gene in a mammal or insect cell line in accordance with a conventional method(Burand, J.P. et al., Virology 101, 286-290 (1980)). For example, the TGF-(31 receptor may be obtained by infecting an insect cell line, e.g., Sf21(Invitrogen, Netherlands), with a recombinant baculovirus containing a TGF-(31 receptor gene;
extracting a water-insoluble receptor protein expressed in the insect cell; solubilizing the water-insoluble receptor protein with guanidine HC1 or urea; refolding the solubilized receptor protein by removing the guanidine or urea to restore the affinity for TGF-(31.
The TGF-(31-specific antibody which may be used in the present invention may be prepared by immunizing a mammal with TGF-ail or a part thereof . The TGF-(31 specific antibody may be a monoclonal antibody or a polyclonal antibody having a specificity only for TGF-(31.
A preferred method for quantifying the amount of TGF-(31 in a body fluid, e.g., plasma or urine, in accordance with the present invention comprises (a) attaching a TGF-(31-specific receptor to a solid support;
(b) adding a body fluid sample to the supported receptor to form a TGF-(31-receptor complex;
(c) binding a TGF-(31-specific antibody conjugated with a label to the complex; and (d) measuring the amount of TGF-(31 using the label as a detection marker.
Representative labels which may be employed in the present invention include horseradish peroxidase, biotin and fluorescence.
A first preferred embodiment of the present invention comprises attaching a TGF-(31 receptor to a solid support, e.g., the well of a microtiter plate;
adding an appropriately diluted sample containing TGF-(31 to the TGF-(31 receptor to allow the formation of a complex between TGF-(31 and the TGF-(31 receptor; washing the support with a phosphate buffered saline(PBS);
adding thereto a chromogenic enzyme-conjugated anti-TGF-(31 antibody and developing the chromogenic enzyme; and measuring the optical density of the resulting solution to quantify the content of TGF-X31 in the sample.
In a second preferred embodiment of the present invention, a liquid containing a TGF-(31-specific receptor may be used in place of the supported TGF-(31 receptor. In this method, the amount of TGF-(31 in a sample may be quantified by adding the sample to the liquid containing a TGF-(31-specific receptor; adding a TGF-(31 specific antibody conjugated with a label thereto; precipitating an antibody-TGF-(31-receptor complex; and measuring the optical density thereof.
The inventive method is capable of detecting TGF-(31 at a very low concentration range of 30 pg/ml or below.
The above method is particularly useful in cancer diagnosis, since the TGF-(31 concentration in a cancer patient's body fluid is distinctly different from that of a healthy person. Accordingly, a cancer may be detected by repeating the above method to quantify the TGF-(31 level in a patient's body fluid sample, e.g., 5 plasma or urine; and comparing the TGF-(31 concentration with that of a healthy person.
A preferred embodiment of the inventive method for detecting a cancer comprises (a) attaching a TGF-(31-specific receptor to a solid support;
(b) adding a body fluid sample to .the supported receptor to form the TGF-X31-receptor complex;
(c) binding a TGF-ail-specific antibody conjugated with a label to the complex;
(d) measuring the amount of TGF-ail using the label as a detection marker; and (e) comparing the TGF-(31 amount with that of a healthy person.
The above method is particularly effective in detecting stomach cancer, hepatoma, breast cancer, lung cancer, rectal-colic cancer, prostate cancer and uterine cervical cancer.
A composition which may be used in the method for detecting a cancer comprises a TGF-(31 receptor, preferably RIII, and a TGF-(31 specific antibody.
In order to improve the sensitivity, the monoclonal antibody may be obtained by preparing a hybridoma cell line which produces TGF-(31-specific monoclonal antibody using TGF-(31 or an antigenic determinant part thereof as an immunogen according to a conventional cell fusion method; and isolating the monoclonal antibody from the hybridoma cell line. For example, such a hybridoma cell line may be prepared by immunizing a mouse with human TGF-(31; fusing the mouse spleen cell with myeloma cell according to the cell fusion method described by Kohler and Milstein(Eur. J.
Immunol., ~., 511-519 (1976)); selecting by way of using ELISA a hybridoma cell line having a specificity only for human TGF-(31; determining the subclass of the monoclonal antibody produced by the hybridoma cell line using an immunodiffusion method; and selecting a hybridoma cell line secreting IgGl subclass with the highest antibody titer. The hybridoma cell line thus obtained was designated hTGF-46 and deposited with Korean Collection for Type Culture(Address: #52, Oun-dong, Yusong-ku, Taejon 305-600, Republic of Korea) on April 20, 1998 under the accession number of KCTC 0460BP, in accordance with the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganism for the Purpose of Patent Procedure.
Hybridoma cell line hTGF-46 originates from (3-limphoma, and continuously divides while producing human TGF-(31-specific, IgGl subclass antibody. The hybridoma cell line may be cultured in RPMI 1640 medium(Gibco-BRL, USA) containing loo bovine fetal serum at 37 °C under an atmosphere of 5% COz and 100% humidity. The cell number doubles in 12 to 14 hours. This hybridoma cell line floats on the medium without attaching itself to the bottom of the culture flask and has a round shape having a diameter of 15 to 20 ~,m.
To produce a large amount of the TGF-ail-specific monoclonal antibody from the hybridoma cell line, the hybridoma cell line is injected to a mouse and when its abdominal cavity swells the ascites containing a high concentration of hybridoma cells is taken to isolate the monoclonal antibody therefrom.
When TGF-(31, -X32 and -(33 are subjected to electrophoresis followed by western blotting, the monoclonal antibody of the present invention recognizes only TGF-X31, but not TGF-(32 or -(33. This suggests that the present monoclonal antibody has a unique specificity for TGF-ail. The monoclonal antibody of the present invention also shows a high affinity toward human TGF-(31, and binds to the epitope region corresponding to the 5th to 80th amino acid residues of TGF-(31.
The following Examples are intended to further illustrate the present invention without limiting its scope.
Further, percentages given below for solid in solid mixture, liquid in liquid, and solid in liquid are on a wt/wt, vol/vol and wt/vol basis, respectively, unless specifically indicated otherwise.
Exam lp a 1: Sensitivity of TGF-(31 for Receptor (Step 1) Preparation of TGF-(31 type III receptor Plasmid pCEP4(Invitrogen, Netherlands) containing a full length cDNA of Human TGF-(31 type III receptor was subjected to polymerase chain reaction(PCR) using primers RIII1 and RIII2(SEQ ID NOs: 1 and 2) to obtain a DNA fragment encoding an extracellular domain of the receptor which is composed of 400 amino acid residues(1 to 400). The DNA fragment thus obtained was inserted into baculovirus vector pCRBac(Invitrogen, Netherlands) to obtain recombinant plasmid pCRBac-TGFR.
E. coli cells were transformed with the recombinant plasmid pCRBac-TGFR and the transformed E. coli cells were selected on a selective medium, LB medium containing ampicillin.
Vector pCRBac-TGFR and Bac-N-Blue DNA(Invitrogen, Netherlands) were cointroduced to insect cell line Sf 21(Invitrogen, Netherlands) using the liposome transfection method(Burand, J.P., Virology, 101, 286 290 (1980)) and cultured for 3 days to obtain a virus product. After 3 days, the virus thus obtained was subjected to plaque analysis using lacZ gene as a selective marker to select the recombinant virus. The recombinant virus thus obtained was subjected to PCR
using forward primer(SEQ ID NO: 3) and reverse primer(SEQ ID NO: 4) to confirm the presence of the TGF-(31 receptor gene. The wild vaculovirus showed a 839 by PCR product whereas the recombinant virus gave a 1.5 kbp PCR product.
Insect cell line SF21 was infected with the recombinant virus and then cultured for 5 days. The culture was centrifuged to remove cell debris and the supernatant containing virus was collected.
Insect cell line Sf21 was inoculated with the supernatant and then cultured at 27 °C for 72 days in Grace Insect medium(Invitrogen, Netherlands) containing 10% fetal bovine serum(FBS), 7.3% TC
yeastolate, and 73% lactoalbumin hydrolysate. The culture was centrifuged to collect cells and the cells were washed with PBS. Protein lysis solution(50 mM
Tris-HC1 (pH 7. 5) , 50 mM NaCl, 10 mM (3-mercaptoethanol, 1% Triton X-100 and 2 mM BMSF) was added thereto and then the resulting solution was heated at 100 °C for 10 minutes to prepare a sample.
The sample was subjected to SDS-PAGE in 12.5%
SDS-polyacrylamide gel and the resulting gel was stained with coomassie brilliant blue. The gel was subjected to western blotting which was conducted by electrically transferring the proteins separated on the gel to a filter, binding the antibody for TGF-(31 receptor obtained from R&D Systems Inc., USA to the proteins of the filter and then analyzing the TGF-X31 using horseradish peroxidase(HRP)-conjugated anti-IgG
secondary antibody(Chemicon, USA) to confirm the expression of the TGF-(31 III receptor.
Since the TGF-(31 receptor is water-insoluble, 8M
guanidine HCl (pH 8 . 0 ) was added to the sample and the resulting solution was stirred for 1 hour. The resulting solution was centrifuged at 7,000 rpm for 40 minutes and then the supernatant was adjusted to a protein concentration of 2 mg/ml. To restore the binding activity of the TGF-(31 receptor to TGF-X31, the resulting solution was added to a refolding buffer(100 mM Tris, 0.5 M arginin, 0.2 M EDTA, pH 8.0) to a protein concentration of 150 ~g/ml and kept at 10 °C
for 40 hours. The resulting solution was dialyzed with 20 mM Tris solution (pH 8 . 0) , successively in the order of twice every 4 hours, once after overnight, and twice every 2 hours thereafter, to effectively refold the TGF-(31 receptor.
(Step 2) Sensitivity of TGF-X31 type III receptor for TGF- (31 Each 2 ~g of the TGF-X31 type III receptor obtained in Step 1 was placed in the wells of a microtiter plate and the resulting plate was held at an ambient temperature for 24 hours to attach the receptor on the plate. 2 ng of purified TGF-(31(R&D
systems Inc., USA) was dissolved in PBS and diluted serially. Each dilution solution was added in an amount of 100 ~l to the well and then held at an ambient temperature for 3 hours to allow the TGF-(31 bind the receptor. Each well was washed with PBS
containing 0.050 of Tween 20(PBST) and then HRP-conjugated anti-TGF-(31 antibody (R&D systems Inc . , USA) was added thereto. The resulting plate was left at room temperature for 1.5 hours. Each well was washed with PBST. 100 ~l of TMB-ELISA(Gibco-BRL, USA), a substrate of HRP, was added thereto and the resulting plate was left at an ambient temperature for 20 minutes to develop. The development reaction was terminated by adding 25 ~l of 2N sulfuric acid. The optical density of the reaction mixture was determined at a measuring wavelength of 450 nm and a correction wavelength of 570 nm, and the result were plotted to obtain a standard optical density-concentration correlation.
Fig. 1 shows that the correlation thus obtained 5 is a straight line with a correlation coefficient of 0.999 and a slope of 0.28. The slope represents the sensitivity of the receptor used in the measurements and the TGF-(31 type III receptor is deemed to have an excellent sensitivity toward TGF-(31 binding. The 10 correlation in Fig. 1 also shows that an extremely low concentration of TGF-(31, down to 10 pg/ml, can be detected by the present method.
The above procedure was repeated using TGF-(31 type II receptor(R&D systems Inc., USA) to determine the sensitivity of the TGF-(31 type II receptor. The results which are also plotted in Fig. 1 show that the correlation obtained using TGF-(31 type II receptor is also a straight line with a correlation coefficient of 0.999 and a slope of 0.57. Accordingly, the type II
receptor may also be used in quantifying the concentration of TGF-(31 but its sensitivity is considerably lower than that of type III receptor.
Example 2: Specificity of TGF-(31 Type III Receptor for TGF- (31 The procedure of step 2 of Example 1 was repeated except that a mixture containing 2,000 pg/ml TGF-(31, 2, 000 pg/ml TGF-(32 and 2, 000 pg/ml TGF-(33 (R&D Systems Inc., USA) was used in place of TGF-(31. The procedure of step 2 of Example 1 was repeated using 2, 000 pg/ml TGF-(31 as a control. Results are shown in Table I.
Table I
TGF-(31 TGF-(31 + TGF-(32 + TGF-(33 TGF-ail III receptor 100% 91.50 As can be seen from Table I, the TGF-X31 type III
receptor binds only with TGF-(31 without the occurrence a crossreaction with TGF-(32 or TGF-(33.
Example 3: Measurement of Plasam TGF-(31 Concentration in Cancer Patients Using Monoclonal Antibody Blood samples were taken from 101 healthy persons, 111 stomach cancer patients, 100 hepatoma patients and 151 breast cancer patients. Blood samples were collected with vacuumtainer containing 0.081 ml of 15%
ethylene diamine tetraacetic acid(EDTA) as an anticoagulant, and then the resulting mixture was centrifuged at 3,000 rpm for 20 minutes to obtain a plasma sample. 0.1 ml of the plasma sample was added to 0.1 ml of 2.5 N acetic acid/10 M urea solution. The resulting mixture was kept at room temperature for 10 minutes, and neutralized with 0.1 ml of 2.7 N NaOH
containing 1M hydroxyethyl piperazine ethanesulfonic acid(HEPES). Activated plasma thus obtained was diluted 4-fold with PBST to obtain a plasma sample solution which was subj ected to the following process for measuring its TGF-X31 concentration.
0.1 ml of the plasma sample solution thus obtained, as well as 0.1 ml portions of TGF-ail standard solutions(0, 100, 1,000 and 2,000 pg/ml), were respectively added to the wells of a 96-well plate containing TGF-(31 type III receptor, kept at room temperature for 3 hours, and then, washed three times with PBST. Purified TGF-(31 monoclonal antibody-HRP complex(Sigma) was added to each well and then the plate was kept at room temperature for 1.5 hours, followed by washing the wells three times with PBST.
100 ~l of TMB ELISA(Gibco-BRL, USA), a substrate of HRP, was added to each well and the plate was kept at room temperature for 20 minutes to develop. The development reaction was terminated by adding 25 ~l of 2N sulfuric acid. The optical density of the reaction mixture was determined at a measuring wavelength of 450 nm, and a correction wavelength of 570 nm. The TGF-X31 concentration of the plasma sample was determined based on the calibration curve obtained using standard solutions, and the results are shown in Table II and Fig. 2.
Table II
Plasma Sample Group Mean Standard Range Error (ng/ml) (ng/ml) Stomach Cancer(n=111) 6.53 0.31 1.5 - 16.35 Hepatoma(n=100) 5,gg 0,3 1.77 - 14.76 Breast Before 5.49 0.32 0.87 - 13.44 Cancer Operation (n=117) After 2.15 0.42 0.46 - 9 Operation (n=34) [Healthy Person(n=101) I 1.03 0.08 0.27 - 8 As can be seen in Fig. 2 which depicts distribution patterns of plasma TGF-(31 concentrations of respective patient groups, the plasma samples of cancer patients, display TGF-(31 concentration patterns which are distinctly different from that of the healthy group. This suggests that the above-mentioned cancers can be detected by measuring plasma TGF-(31 concentration in accordance with the above procedure.
Example 4: Measurement of Plasma TGF-(31 Concentration of Cancer Patient Using Monoclonal Antibody The procedure of Example 3 was repeated using blood samples taken from 288 healthy persons, 29 lung cancer patients, 48 rectal-colic cancer patents, 50 prostate cancer patients and 88 uterine cervical cancer patients to measure respective plasma TGF-X31 concentrations and the results are shown in Table III
and Fig. 3.
Table III
Plasma Sample Mean Standard Standard Error(ng/ml) Deviation Lung Cancer(n=29) g,4g 1,2~ 4.16 Rectal-colic Cancer(n=48) 5,1g p,g~ 3.69 Prostate Cancer(n=50) 4.12 0.53 2.30 Uterine Cervical Cancer g.55 0.92 5.25 (n=88) Healthy Person(n=288) 1,17 0.05 0.55 P<0.01 As can be seen in Fig. 3 which shows distribution patterns of plasma TGF-(31 concentrations of respective patient groups, the plasma samples of cancer patients, display TGF-ail concentration patterns which are distinctly different from that of the healthy group.
This suggests that the above-mentioned cancers can be detected by measuring plasma TGF-(31 concentration in accordance with the above procedure.
Exam 1~: Measurement of Plasma TGF-ail Concentration of Cancer Patient Using Polyclonal Antibody The procedure of Example 3 was repeated using blood samples taken from 50 healthy persons, 50 hepatoma patients and 50 breast cancer patients, except that a polyclonal antibody was used in place of the monoclonal antibody, to measure respective plasma TGF-(31 concentrations and the results are shown in Table IV.
Table IV
Plasma Sample Mean Standard Standard Range Error(ng/ml) Deviation (ng/ml) Hepatoma 5.14 0.57 2.92 1.44-16.96 (n=50) Breast Cancer 5.31 0.46 1.67 2.07-10.27 (n=50) Healthy Person 1.19 0.08 0.29 0.70-1.9 (n=50) P<0.05 As can be seen in Table IV, the plasma samples of cancer patients display TGF-ail concentration patterns which are distinctly different from that of the healthy group. This suggests that the above-mentioned cancers can be detected by measuring plasma TGF-(31 concentration in accordance with the above procedure.
Example 6: Preparation of Hybridoma Cell Line Producing a Monoclonal Antibody for TGF-(31 (Step 1) Immunization of Mouse TGF-(31 was mixed with an equal volume of Complete Freund Adjuvant until the mixture became fluid and the resulting mixture was injected, in an amount of 100 ~1/mouse, to the caudal vein of a 7 weeks-old Balb/c mouse. After 2 weeks, the same amount of TGF-(31 as in the first injection, which was mixed with Freund's incomplete adjuvant, was injected to the caudal vein of the mouse . Af ter 4 to 5 days , a small amount of blood was taken from the tail and the presence of an antibody for TGF-(31 was confirmed by ELISA. 30 ~g of human TGF-(31 dissolved in 0.85% PBS was then intravenously injected 3 to 4 days before the following cell fusion procedure.

(Step 2) Cell Fusion Myeloma cell SP2/O~Agl4 (ATCC CRL 1581) was used as a mother cell in the cell fusion procedure. The mother 10 cell was cultured in RPMI medium containing 10% FBS
while maintaining a maximum cell density of 5x105/ml.
The immunized mouse obtained in Step 1 was anesthetized using ether and its spleen was removed to be homogenized with a tissue homogenizer. The resulting 15 homogenate was suspended in HBSS(Gibco-BRL, USA) and the resulting suspension was placed in a 15 ml centrifugal tube and centrifuged. This procedure was repeated twice to wash the spleen cells thoroughly. The mother cells, SP2/O~Agl4, were suspended in HBSS and centrifuged.
This procedure was repeated twice . The spleen cells and the SP2/O~Agl4 cells were respectively resuspended in 10 ml of HBSS to count the cell number in each suspension.
108 spleen cells and 10' SP2/O~Agl4 cells taken from respective suspensions were mixed in a centrifugal tube and then centrifuged to precipitate the cells. The centrifugal tube was tapped with fingers to disperse the precipitated cells and, then, kept at 37 °C for 1 minute.
1 ml of HBSS containing 45% PEG(w/v) and 5o DMSO were added thereto over a period of 1 minute, followed by shaking the tube for 1 minute. 9 ml of RPMI medium was added thereto over a period of 3 minute and then RPMI
medium was added thereto until the total volume of the cell suspension became 50 ml while shaking the tube.
The resulting suspension was centrifuged and the cell pellet thus obtained was resuspended at a concentration of 1 to 2 x 105/ml in HAT medium(Gibco-BRL, USA). 0.2 ml portions of the resulting resuspension were placed in the wells of a 96-well microtiter plate and then cultured for several days in an incubator, maintaining the condition of 37 °C, 5% C02 and 100 % humidity.
(Step 3) Selection of Hybridoma cell Producing Monoclonal antibody The hybridoma cells obtained in Step 2 were subjected to ELISA using human TGF-(31 antigen to obtain cells which specifically react with TGF-(31, as described below.
Human TGF-(31 antigen was added to the wells of a microtiter plate in an amount of 50 x,1(2 ~g/ml)/well and kept at room temperature for 12 hours to attach the antigen to the well surface. The wells were washed with PBST to remove unattached antigen.
The hybridoma cell culture obtained in Step 2 was added in an amount of 50 ~1/cell to each well and kept at 37 °C for 1 hour. The wells were washed with PBST to remove the culture. Goat anti-mouse IgG-HRP(sigma, USA) was added thereto, held at room temperature for 1 hour and washed with PBST. 100 ~l of Substrate solution(OPD, Sigma) was added thereto, held at room temperature for 20 minutes, and the optical density of the resulting reaction mixture was measured at 492 nm.
Hybridoma cell lines secreting antibodies having high specificity for human TGF-(31 antigen were first selected, and each of these hybridoma cell lines was subjected to ELISA using human TGF-(31, -(32 and -X33 to screen hybridoma cells which have specificity only for human TGF-(31 antigen. Each of the hybridoma cells thus obtained was subjected to limiting dilution to obtain 7 hybridoma cell line clones producing a monoclonal antibody, hTGF-7, -8, -31, -46, -70, -119 and -207.
Each clone was freeze-dried.
The hybridoma cell culture was centrifuged and the supernatant was subjected to ELISA to determine the antibody titer and then subjected to immunotype kit(Sigma, USA) to determine the subclass type of the antibody. The results are shown in Table V.
Table V
Clone No. Optical Density(492 nm) Subclass Type hTGF-46 2.125 IgGl HTGF-7 1.644 IgGl HTGF-70 2.590 IgGl HTGF-8 2.395 IgGl HTGF-207 1.735 IgGl HTGF-119 2.462 IgGl HTGF-31 2.282 IgGl As can be seen from Table V, all of the 7 clones were IgGl.
Among the 7 clones, the clone having the highest titer, hTGF-46, was selected and injected intraperitoneally to a mouse. Then its ascites was collected and subjected to western blotting. The results showed that the hybridoma cell line clone hTGF-46 secrets a monoclonal antibody having a high specificity for human TGF-(31. The hybridoma cell line hTGF-46 was deposited with Korean Collection for Type Culture(Address: #52, Oun-dong, Yusong-ku, Taejon 305-600, Republic of Korea) on April 20, 1998 under accession number of KCTC 0460BP, in accordance with the terms of the Budapest Treaty on the International Recognition of the Deposit of Microorganism for the Purpose of Patent Procedure.
Examx~le 7: Production of Monoclonal Antibody for TGF-(31 To produce monoclonal antibody for TGF-(31 using the hybridoma cell line obtained in Example 6, 0.5 ml of pristane was injected intraperitoneally to Balb/c mice, and after 1 week, 5x106 hybridoma cells were injected to each mouse. From the mice having swollen abdominal cavity, ascites containing a high concentration of hybridoma cells was taken and centrifuged at 10,000 rpm to remove the hybridoma cells. The supernatant was stored at -2 0 °C .
Column was filled with protein G beads and then washed four times with lxPBS. 2 ml of the supernatant was applied dropwise at a rate of 5 drops/minute to the column, and 0.1 M glycine-HC1 solution was introduced at a rate of 1 drop/10 minutes to the column to elute IgG.
HRP was activated in 0.1 M sodium phosphate buffer(pH 6.5) containing 1.25 % glutaraldehyde and the activated HRP was dialyzed against carbonate buffer(pH
9.2). The dialyzed HRP was reacted with the IgG to obtain a HRP-conjugated IgG. After completion of the reaction, the RZ value (A4oa/Azeo) was determined by measuring the optical density of the reaction mixture at 280nm and 403 nm. In order to determine the activity of the enzyme-conjugated antibody, each well of a microtiter plate was coated with 1 ~.g of TGF-(31 and then reacted with the HRP-conjugated IgG to determine the activity. Futher, the HRP-conjugated IgG was subjected to Western blotting to confirm the activity thereof.
Example 8: Western Blotting To examine whether the TGF-(31-specific monoclonal antibody obtained in Example 7 reacts with TGF-~i2 and TFG-X33, the monoclonal antibody was subjected to SDS-PAGE and western blotting as follows.
Human TGF-(31, -(32 and -(33 proteins were subjected to SDS-PAGE on a 10% SDS-polyacrylamide gel and then transferred electrically to a nitrocellulose filter membrane. The membrane was reacted with the monoclonal hours. The resulting membrane was treated with 3%
bovine serum albumin at room temperature for 12 to 14 hours to block nonspecific reactions of the proteins.
The membrane was washed three times with PBS containing 0.5% Tween 20 and then reacted with HRP-conjugated anti-mouse IgG(Sigma, USA) at room temperature. The membrane was washed with PBS containing 0.5% Tween 20, and then, a substrate solution(TMB, Gibco-BRL, USA) was added to the membrane to develop.
The result showed that the monoclonal antibody of the present invention reacts only with human TGF-(31 and did not reacted with human TGF-~i2 and -(33. Therefore, the present monoclonal antibody has specificity only for human TGF-X31.
While the invention has been described with respect to the above specific embodiments, it should be recognized that various modifications and changes may be made to the invention by those skilled in the art which also fall within the scope of the invention as defined by the appended claims.

~ ~~oPtw t o ~t I~NAT1~NAL P~tM
~~c~~~r rrr ~ ~~.s~ a~ err o~r~u~r~, r~~posrr tta~~e~.~
To: Choc, Yox~-Iipung 3hindongA .Apt 1-2G3, i~ roan t Yot~oa~-dons, Doug l~u. Tacwu $00=200, Ftepubltc of I~osen z zbcA~ort of ~ ~oo~~arusM
_ .~. .-Identltlcation refs given by ttm Atllutiber given by the bT'I'c~R INTERNATIONAL DEPt7SITARY
AZTTI~O~RTi'Y:
hT(~ln - 4s ~CZC O~a~P
II. SCIENTIFTC DESCLtI~ON ANb/dRP.~p0~8D TA~CONC~C DfiSTGNATION
'Ihe m3nism ideatlfl0d rmdaar I abovo arcs mecompa~od bar:
I x 1 a ~tcientifyc dosCtiption I 1 a ~oposed tsxaasoomic doai,Baa~oa (Mm3c wlth a at~ss arhere a~tpliC3We) 1IL REGh>~ AND ACC~YTANC6 This Int~c~onsl bepasiraiy Anthaaity aooepts the mitsnargantsm Idaatisied m~dat I above, which waR receivad by it oa I~pel1 3Q 1098.
IV. RFC»pT' aFRBQtJ~STPQk SIGN
'11~e mi~a~gaaism iclenttfiad ~mder I ebonra vas roceiv~od by this Intomational Dapoeitary Acthacity on and a zto convert tha original dapoQit to a depp~it artder tha B~apest ~enty ~aaa iecelvod tr~ritvn V. INTERNATiI~NAt.. DEP(!S!"L"ARY' AZI7:~IOR1TY
Name: K area Bessarch Instltutti of 5igdt0at~c(a)uEDc~cacx~(s) having thepower to B ins ci et1 C ti o t1d B ~ tee b aOln g d ~temtit the IntornntioaeI
Dcpositary Korean Coltecfmn tofTgpe Cuttan~ Aut6cxityorofautha~fzcdotfrcia6(s);
Adcfiroas: KC'PC. KR~SB
X52 4W~.-dung, ~s6i~ ku.
1'ae~bn g05-FOCI, Xyurrg Sooic Bata, Ctuxtor Rcpubtlc of Korea. Dao~: qty ~ 1888 Sequence Listing <110> Hanmi Pllann. Co., Ltd <120> METHOD FOR QUANTIFYING TRANSFORMING GROWTH FACTOR-(31 AND METHOD FOR DETECTING CANCER BY USING SAME
<130> PCA00418/HMY
<150> KR 1999-12568 < 151 > 1999-04-09 <150> KR 1999-43935 <151> 1999-10-12 <160> 4 <170> KOPATIN 1.5 <210> 1 <211> 18 <212> DNA

<213> Artificial Sequence <220>
<223> Primer RIII1 <400> 1 atggcagtga catcccac 1 g <210> 2 <211> 12 <212> DNA

<213> Artificial Sequence <220>
<223> Primer RIII2 <400> 2 atttgggctt cc 12 <210>3 <211>24 <212>DNA

<213>Artificial Sequence <220>
<223> Forward primer <400> 3 tttactgttt tcgtaacagt tttg 24 <210>4 <211>21 <212>DNA

<213>Artificial Sequence <220>
<223> Reverse primer <400> 4 caacaacgca cagaatctag c 21

Claims (22)

What is claimed is:
1. A method for quantifying the amount of TGF-.beta.1 in a sample which comprises treating the sample with a TGF-.beta.1-specific receptor to form a complex between TGF-.beta.1 and the receptor and measuring the amount of the complex.
2. The method of claim 1, which comprises (a) attaching the TGF-.beta.1-specific receptor to a solid support;
(b) adding the sample to the supported receptor to form the TGF-.beta.1-receptor complex;
(c) binding a TGF-.beta.1-specific antibody conjugated with a label to the complex; and (d) measuring the amount of TGF-.beta.1 using the label as a detection marker.
3. The method of claim 1 or 2, wherein the TGF-.beta.1-specific receptor is TGF-.beta.1 type III receptor.
4. The method of claim 2, wherein the TGF-.beta.1-specific antibody is an antibody prepared by immunizing a mammal with TGF-.beta.1 or a part thereof.
5. The method of claim 4, wherein the antibody is a monoclonal antibody or polyclonal antibody.
6. The method of claim 5, wherein the monoclonal antibody is produced from a hybridoma cell line hTGF-46(KCTC 0460BP).
7. The method of claim 1 or 2, wherein the concentration of TGF-.beta.1 in the sample is 30 pg/ml or lower.
8. The method of claim 2, wherein the label is horseradish peroxidase, biotin or fluorescence.
9. A method for detecting a cancer in a patient which comprises treating a body fluid sample taken from the patient with a TGF-.beta.1-specific receptor to form a complex between the receptor and TGF-.beta.1 present in the sample; measuring the amount of the complex to quantify the concentration of TGF-.beta.1 in the sample; and comparing the TGF-.beta.1 concentration with that of a healthy person.
10. The method of claim 9, which comprises (a) attaching the TGF-.beta.1-specific receptor to a solid support;
(b) adding the sample to the supported receptor to form the TGF-.beta.1-receptor complex;
(c) binding a TGF-.beta.1-specific antibody conjugated with a label to the complex;
(d) measuring the amount of TGF-.beta.1 using the label as a detection marker to determine the TGF-.beta.1 concentration in the sample; and (e) comparing the TGF-.beta.1 concentration with that of a healthy person.
11. The method of claim 9 or 10, wherein the TGF-.beta.1 specific receptor is TGF-.beta.1 type III receptor.
12. The method of claim 10, wherein the TGF-.beta.1-specific antibody is an antibody prepared by immunizing a mammal with TGF-.beta.1 or a part thereof.
13. The method of claim 12, wherein the antibody is a monoclonal antibody or a polyclonal antibody.
14. The method of claim 13, wherein the monoclonal antibody is produced from a hybridoma cell line hTGF-46(KCTC 0460BP).
15. The method of claim 9 or 10, wherein the concentration of TGF-.beta.1 in the sample is 30 pg/ml or lower.
16. The method of claim 10, wherein the lable is horseradish peroxidase, biotin or fluorescence.
17. The method of claim 9 or 10, wherein the cancer is selected from the group consisting of stomach cancer, hepatoma, breast cancer, lung cancer, rectal-colic cancer, prostate cancer and uterine cervical cancer.
18. The method of claim 9 or 10, wherein the body fluid is plasma or urine.
19. A composition for detecting a cancer which comprises a TGF-.beta.1-specific receptor and a TGF-.beta.1-specific antibody.
20. The composition of claim 19, wherein the TGF-.beta.1 specific receptor is TGF-.beta.1 type III receptor.
21. A hybridoma cell line which is hTGF-46(KCTC
0460BP) producing a human TGF-.beta.1-specific monoclonal antibody.
22. A TGF-.beta.1-specific monoclonal antibody produced by hybridoma cell line hTGF-46(KCTC 0460BP).
CA002369892A 1999-04-09 2000-04-10 Method for quantifying transforming growth factor-.beta.1 and method for detecting cancer by using same Abandoned CA2369892A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
KR19990012568 1999-04-09
KR1999/12568 1999-04-09
KR1999/43935 1999-10-12
KR19990043935 1999-10-12
PCT/KR2000/000329 WO2000062062A1 (en) 1999-04-09 2000-04-10 METHOD FOR QUANTIFYING TRANSFORMING GROWTH FACTOR-β1 AND METHOD FOR DETECTING CANCER BY USING SAME

Publications (1)

Publication Number Publication Date
CA2369892A1 true CA2369892A1 (en) 2000-10-19

Family

ID=26634921

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002369892A Abandoned CA2369892A1 (en) 1999-04-09 2000-04-10 Method for quantifying transforming growth factor-.beta.1 and method for detecting cancer by using same

Country Status (11)

Country Link
EP (1) EP1166112A4 (en)
JP (1) JP2002541479A (en)
CN (1) CN1355882A (en)
AU (1) AU768029B2 (en)
BR (1) BR0009607A (en)
CA (1) CA2369892A1 (en)
ID (1) ID30316A (en)
MX (1) MXPA01009942A (en)
NZ (1) NZ514596A (en)
TR (1) TR200102917T2 (en)
WO (1) WO2000062062A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2002219947A1 (en) * 2000-11-28 2002-06-11 Amgen Inc. Transforming growth factor-beta-related molecules and uses thereof
JP4834835B2 (en) * 2006-07-27 2011-12-14 国立大学法人浜松医科大学 Diagnostic agent for autism

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS59195162A (en) * 1983-04-21 1984-11-06 Toyo Jozo Co Ltd Reagent for assay of transforming gross factor
JPS59216058A (en) * 1983-05-23 1984-12-06 Toyo Jozo Co Ltd Determining method of transforming gross factor
JPS59226864A (en) * 1983-06-07 1984-12-20 Toyo Jozo Co Ltd Method for measuring enzyme immune of transforming gross factor
JPS6018764A (en) * 1983-07-12 1985-01-30 Toyo Jozo Co Ltd Immunological measurement of transforming growth factor
JPH02126157A (en) * 1988-11-04 1990-05-15 Tosoh Corp Method for immunologically measuring human transforming growth factor-beta
WO1993010215A1 (en) * 1991-11-15 1993-05-27 Memorial Sloan-Kettering Cancer Center Purified proteoglycan betaglycan, compositions, and methods
GB9601081D0 (en) * 1995-10-06 1996-03-20 Cambridge Antibody Tech Specific binding members for human transforming growth factor beta;materials and methods

Also Published As

Publication number Publication date
BR0009607A (en) 2002-01-08
ID30316A (en) 2001-11-22
EP1166112A4 (en) 2004-11-10
AU768029B2 (en) 2003-11-27
AU4147100A (en) 2000-11-14
NZ514596A (en) 2003-03-28
MXPA01009942A (en) 2003-07-14
WO2000062062A1 (en) 2000-10-19
EP1166112A1 (en) 2002-01-02
JP2002541479A (en) 2002-12-03
TR200102917T2 (en) 2002-03-21
CN1355882A (en) 2002-06-26

Similar Documents

Publication Publication Date Title
KR101282811B1 (en) Antibody directed against gastrin-releasing peptide precursor and use thereof
US9823251B2 (en) Anti-Uroplakin II antibodies systems and methods
CA2519105A1 (en) Monoclonal antibody and hybridoma producing the same
EP0550561B1 (en) Purification of cytokeratin fragments
US20010046686A1 (en) Sensitive detection of wild-type and mutant EGFR by specific ELISA assays in any biological sample
KR102029394B1 (en) Monoclonal antibody for diagnosis of chikungunya virus infection, hybribodma producing the monoclonal antibody, and diagnosis method using the same
CA2369892A1 (en) Method for quantifying transforming growth factor-.beta.1 and method for detecting cancer by using same
JP2001122900A (en) ANTI-DNASE gamma ANTIBODY AND ITS PREPARATION AND USE
JP3920556B2 (en) Abbreviated midkine (tMK) protein-specific monoclonal antibody and use thereof
JP2008515871A (en) Novel cancer-related antibodies and antigens and their use in cancer diagnosis
KR100378746B1 (en) Method for quantifying active transforming growth factor-beta1 in body fluid and method for detecting cancer by using same
JP2925479B2 (en) Drug for detecting small cell lung cancer and use thereof
US5786451A (en) 23 Kd human retinal CAR antigen
CN113480646B (en) Preparation and application of monoclonal antibody specifically binding H1.4 frameshift mutant protein
CN109776679B (en) Antibody of serine protease inhibitor SPINK1, and preparation method and application thereof
CN111349170B (en) Monoclonal antibody of immune related GTPase family M (IRGM) and application thereof
JP4829961B2 (en) Prostasin partial peptide and anti-prostasin antibody
WO2006057623A1 (en) Runx3 antibodies and method of obtaining the antibodies
WO2012077643A1 (en) IMMUNOLOGICAL cofilin-1 PROTEIN MEASUREMENT METHOD
JP3107248B2 (en) Antibodies and immunochemical assays
JPH06125784A (en) Monoclonal antibody, hybridoma, its production and use
KR20040085749A (en) Monoclonal antibody recognizing resistin, production method and use thereof
JP2006271390A (en) METHOD FOR PRODUCING MONOCLONAL ANTIBODY SPECIFIC TO TRUNCATED MIDKINE (tMK) PROTEIN
WO2006121047A1 (en) Method for determination of megsin in biological sample
JP2011160696A (en) Antibody against modified human igf-1/e peptide

Legal Events

Date Code Title Description
EEER Examination request
FZDE Discontinued