EP0972200A1 - Criblage et traitement au moyen d'un regulateur du complement ou de proteines receptrices du complement - Google Patents

Criblage et traitement au moyen d'un regulateur du complement ou de proteines receptrices du complement

Info

Publication number
EP0972200A1
EP0972200A1 EP98910227A EP98910227A EP0972200A1 EP 0972200 A1 EP0972200 A1 EP 0972200A1 EP 98910227 A EP98910227 A EP 98910227A EP 98910227 A EP98910227 A EP 98910227A EP 0972200 A1 EP0972200 A1 EP 0972200A1
Authority
EP
European Patent Office
Prior art keywords
protein
complement
cancer
related protein
cfi
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.)
Withdrawn
Application number
EP98910227A
Other languages
German (de)
English (en)
Inventor
Robert J. Kinders
David L. Enfield
G. Michael Hass
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.)
Bion Diagnostic Sciences Inc
Original Assignee
Bion Diagnostic Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bion Diagnostic Sciences Inc filed Critical Bion Diagnostic Sciences Inc
Publication of EP0972200A1 publication Critical patent/EP0972200A1/fr
Withdrawn legal-status Critical Current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
    • G01N33/53Immunoassay; 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
    • G01N33/57496Immunoassay; 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 intracellular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/472Complement proteins, e.g. anaphylatoxin, C3a, C5a
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer

Definitions

  • the present invention is generally directed toward screening for
  • the invention is more particularly related to detecting complement C3 protein or a C3 10 related protein (or a nucleic acid molecule encoding such a protein) or a certain complement regulator or complement receptor protein or related protein (or a nucleic acid molecule encoding such a protein), or to modulating the amount or activity of such a protein.
  • cervical cancer in the U.S. is estimated at 15,700 cases for 1996. Although this disease is highly treatable, approximately 4,900 patients die annually.
  • Bladder cancer is the fifth most common cancer in the United States.
  • the American Cancer Society estimated that in 1996 a total of 52,900 new cases would be detected and that there would be 11,700 deaths resulting from this disease.
  • the incidence of bladder cancer increases with age. It is more common in men than in women by a ratio of approximately three to one and has been shown to be highly associated with smoking as well as exposure to certain dyes.
  • the most common type of bladder cancer is transitional cell carcinoma (TCC), representing greater than 90% of all cases.
  • hematuria The most common presenting symptoms are hematuria, which is observed in approximately 80% of the cases, and dysuria. Although hematuria is more often related to nonmalignant conditions, it is recommended that in the presence of such symptoms an evaluation for bladder cancer be completed. Once infection of the urinary tract has been eliminated as a possibility, a full evaluation is likely to include urine cytology, intravenous pyelography and cystoscopy. The possibility of a positive diagnosis by cytology (i.e., the identification of tumor cells in voided urine) increases with the grade of the tumor. In some cases cytological evaluation may be necessary to detect tumor in situ or tumors which are located in the upper end of the bladder.
  • Intravenous pyelography may aid in accurately determining the stage of the tumor.
  • transurethral biopsy and resection are usually performed. These enable removal of the apparent lesion and provide information regarding the clinical stage and extent of invasion of the tumor. Such information aids in the selection of appropriate therapeutic approaches and of subsequent monitoring procedures.
  • monitoring is crucial for the long-term survival of the patient.
  • colonoscopy in that it allows the removal of polyps and biopsy of suspicious lesions at the time of the procedure.
  • the decision to proceed with colonoscopy is based on the positive result of an occult blood test, a procedure with a sensitivity of approximately 37% and a specificity of approximately 97%.
  • the American Cancer Society recommends, for general screening, an annual digital rectal exam for all individuals beginning at age 40. At age 50, the Society further recommends initiation of an annual series of three double tests for occult blood by the guaiac method. Finally, the Society recommends that two initial sigmoidoscopies be performed one year apart, and that, if both are negative, further visualization by sigmoidoscopy be repeated every three to five years.
  • Patients diagnosed with colon cancer are often monitored with one or more blood tests for circulating tumor associated antigens, such as CEA or CA72-4.
  • markers are not capable of detecting early stage disease (A or BI by the
  • Autoimmune Diseases are complex and have diverse mechanisms and pathologies. For clarity, diseases that are known to be mediated by production of specific antibodies to self-antigens, with the result being the destruction of host tissue, are described.
  • Antibodies to basement membranes of lung and kidney glomeruli are produced by individuals with Goodpasture's syndrome, to skin basement membrane in individuals with pemphigoid syndrome, and to collagen in rheumatoid arthritis.
  • the pathologic progression of the disease involves the deposition of specific antibodies to normal constituents in the skin, kidneys, lungs or joints, followed by destruction of the basement membranes by complement activation and by cellular inflammatory processes. These processes result in tissue destruction that accelerates with age, and which current treatments can ameliorate but not control or cure.
  • component C3 is proteolytically activated to C3b, which attaches to the membrane target to be destroyed.
  • the present invention provides a variety of methods for screening for (detecting or monitoring) cancers or treating cancers, or autoimmune disorders, in which complement C3 protein or a C3 related protein is associated, or, in another aspect, cancers in which certain complement regulator or complement receptor proteins or related proteins are associated.
  • the diagnostic methods may be used on a one-time basis when an abnormality is suspected, or on a periodic basis, e.g., to monitor an individual with an elevated risk of acquiring or reacquiring cancer.
  • the present invention provides a method of screening for a cancer which comprises the step of detecting in a sample the presence of complement C3 protein or a C3 related protein, or a nucleic acid molecule encoding the protein (i.e., complement C3 or a C3 related protein).
  • the cancer that is screened for is a colorectal cancer, a renal cancer, a bladder cancer or a cervical cancer.
  • the present invention provides a method of treating a cancer which comprises the step of modulating complement C3 protein or a C3 related protein associated with the cancer.
  • the cancer that is treated is colorectal cancer, renal cancer, bladder cancer or cervical cancer.
  • the present invention provides a method of treating an autoimmune disorder which comprises the step of modulating complement C3 protein or a C3 related protein associated with the autoimmune disorder.
  • the present invention provides a method of screening for a cancer which comprises the step of detecting in a sample the presence of one or more of complement decay acceleration factor (DAF) protein or a DAF related protein, complement Factor I (CFI) protein or a CFI related protein, complement binding protein CR1 or a CR1 related protein, complement binding protein CR3 or a CR3 related protein, or a nucleic acid molecule encoding any one of the above proteins.
  • the cancer that is screened for is a colorectal cancer, a renal cancer, a bladder cancer or a cervical cancer.
  • the molecule detected is CFI protein or a CFI related protein, or a nucleic acid molecule encoding the protein; or CR3 protein or a CR3 related protein, or a nucleic acid encoding the protein.
  • the present invention provides a method of treating a cancer which comprises the step of modulating any one of complement decay acceleration factor (DAF) protein or a DAF related protein, complement Factor I (CFI) protein or a CFI related protein, complement binding protein CR1 or a CR1 related protein, or complement binding protein CR3 or a CR3 related protein, wherein the protein is associated with the cancer.
  • DAF complement decay acceleration factor
  • CFI complement Factor I
  • the cancer that is treated is a colorectal cancer, a renal cancer, a bladder cancer or a cervical cancer.
  • CFI protein or a CFI related protein is modulated; or CR3 protein or a CR3 related protein is modulated.
  • the present invention provides an agent that modulates complement C3 protein or a C3 related protein associated with cancer, for use as a medicament to treat a cancer in which complement C3 protein or a C3 related protein is associated.
  • a composition of the present invention comprises such an agent in combination with a pharmaceutically acceptable carrier or diluent.
  • such an agent is used for the manufacture of a medicament for the treatment of a cancer in which complement C3 protein or a C3 related protein is associated.
  • the present invention provides an agent that modulates complement C3 protein or a C3 related protein associated with an autoimmune disorder, for use as a medicament to treat an autoimmune disorder in which complement C3 protein or a C3 related protein is associated.
  • a composition of the present invention comprises such an agent in combination with a pharmaceutically acceptable carrier or diluent.
  • such an agent is used for the manufacture of a medicament for the treatment of an autoimmune disorder in which complement C3 protein or a C3 related protein is associated.
  • the present invention provides an agent that modulates any one of complement decay acceleration factor (DAF) protein or a DAF related protein, complement Factor I (CFI) protein or a CFI related protein, complement binding protein CR1 or a CR1 related protein, or complement binding protein CR3 or a CR3 related protein, for use as a medicament to treat a cancer in which the protein is associated.
  • a composition of the present invention comprises such an agent in combination with a pharmaceutically acceptable carrier or diluent.
  • such an agent is used for the manufacture of a medicament for the treatment of a cancer in which the protein is associated.
  • the agent modulates CFI protein or a CFI related protein; or modulates CR3 protein or a CR3 related protein.
  • Figure 1 illustrates an enzyme immunoassay for the detection of renal cancer that employs anti-C3 and alkaline phosphatase-conjugated MAb X46.3.
  • the presence of MAb X46.3 was detected by measuring the absorbance at 405 nm (A405) following the addition of substrate for alkaline phosphatase.
  • Figure 2 shows a photograph of an SDS-PAGE gel demonstrating the inhibition of C3b degradation by MAb X46.3.
  • Figure 3 illustrates the effect of MAbs X46.3 and X87.2 on complement- mediated hemolysis.
  • Figure 4 shows a photograph of an agarose gel demonstrating the presence of DAF mRNA in several different human cancer cell lines.
  • Figure 5 shows the results for amplification of complement receptor CR1 cDNA from a variety of cell lines.
  • Figure 6 shows the results for amplification of complement receptor CR2 cDNA from a variety of cell lines.
  • Figure 7 shows the results for amplification of complement receptor CR3 cDNA from a variety of cell lines.
  • Figure 8 shows the results for amplification of complement Factor I cDNA from a variety of cell lines.
  • Figure 9 illustrates the reactivity of MAb X67.2 with cervical specimens, as identified by a dot-blot procedure.
  • Figure 10 shows the results for amplification of cDNA for the CD l ib chain of CR3 from a variety of human cell lines by the CD1 lb 18-933RT primer set.
  • Figure 11 shows the results for amplification of cDNA from various peripheral blood leukocyte specimens using the CD1 lb 18-933RT primer set.
  • Figure 12 shows the results of amplification of cDNA for the CDl lb chain of CR3 from a variety of human cell lines by the CD1 lb 129-933 primer set.
  • Figure 13 shows the results of amplification of cDNA for the CDl lb chain of CR3 from a variety of human cell lines by the CDl lb 2665-3103RT primer set.
  • Figure 14 shows the results of amplification of cDNA for the CDl lb chain of CR3 from matched normal (Nl and N2) and cancerous (Cl and C2) colon tissue from two patients with colon cancer, using the CDl lb 129-933RT primer set.
  • the lane labeled MWS contains molecular weight markers.
  • Figure 15 shows, on the left half of the figure, the results of amplification of cDNA for the CD1 lb chain of CR3 from matched normal (Nl and N2) and cancerous (Cl and C2) colon tissue from two patients with colon cancer, using the CDl lb 2665-3103RT primer set. Amplification of cDNA from the same specimens with the MIC 148-2533RT control primer set is shown in the right half of the figure. Two lanes labeled MWS contain molecular weight markers.
  • Figure 16 shows the results of amplification of cDNA for complement Factor I from matched normal (Nl and N2) and cancerous (Cl and C2) colon tissue from two patients with colon cancer, using the CFI primer set FI7us and FIG683ds.
  • the tumor-associated protein antigens and nucleic acid sequences screened for or modulated in the present invention have been determined, by apparent molecular weights, by sequence comparisons, by amplicon size and by restriction mapping to be related to human complement proteins C3, Factor I, Decay Accelerator Factor, or Complement Receptors CR1 or CR3. Since cancer cells may produce more than one form of any of these protein, the terms "complement related protein” and "complement receptor related protein,” as used herein, refer to variants of the human complement and complement receptor proteins. The variants may be the result of mutations, alternate splicing or recombination events that alter nucleic acid molecules encoding the human complement and complement receptor proteins.
  • amino acid sequence identity between a human complement related protein or complement receptor related protein from a tumor cell and the corresponding human complement protein or human complement receptor protein will be at least about 50%. More typically, the amino acid sequence identity will be at least about any integer from (and including) 50% to 100%, such as at least about 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95% identity. Variants that are nearly identical to a human complement protein or a human complement receptor protein have at least about 85% or 90% identity.
  • amino acid sequence "identity" is determined by the alignment of amino acid sequences and establishment of identical amino acid residues using the program GeneJockey II (1993) for Macintosh (Philip L. Taylor, published by Biosoft, Cambridge, UK).
  • the program is run in the amino acid homology mode, using program default parameters. In the comparison of two sequences aligned by the program, the percent identity is calculated only for those positions where there is an amino acid residue present in both of the two sequences.
  • a nucleic acid molecule encoding a human complement related protein or complement receptor related protein will typically hybridize under moderately stringent conditions to one or more primer pairs, as described below. This reflects conservation of certain sequences (disclosed herein) for tumor-associated human complement related, or human complement receptor related, antigens.
  • a protein may generally be identified as a tumor-associated human complement related, or human complement receptor related, antigen based on the ability of a nucleic acid molecule encoding the protein to hybridize under moderately stringent conditions to one or the other or more of the primer pairs, as described below.
  • Moderately stringent hybridization conditions are well known to one in the art, and may be defined, for example, as those performed at the calculated melting temperature of the primer with the target. Based on the disclosure herein, in combination with the methodologies known in the art, it will be evident to one in the art whether a protein is a tumor-associated human complement related or complement receptor related antigen, or whether a nucleic acid molecule encodes such as protein. Listed below are the reference numbers for nucleic acid sequences coding for complement proteins.
  • the present invention in one aspect is directed toward methods of screening for (detecting or monitoring) or treating an abnormality (such as cancers or autoimmune disorders) in which complement C3 or a C3 related protein is associated.
  • complement C3 or C3 related protein(s), "C3rp” has been found to be associated with the presence of tumor cells and found to survive in detectable concentrations in specimens from patients with tumors.
  • the present disclosure describes, for example, the presence of elevated C3/C3rp (i.e., tumor C3 or C3rp) in the urine of bladder cancer patients and the synthesis of C3/C3rp by tumor cells in culture.
  • Monoclonal antibodies which recognize C3/C3rp were prepared by immunization with partially purified protein fractionated from the urine of bladder cancer patients by heparin agarose chromatography. Monoclonal antibodies raised against normal blood C3 do not distinguish C3rp from C3.
  • C3/C3rp may be isolated in substantially pure form. Briefly, for example, urine samples are clarified (e.g., by centrifugation) and concentrated (e.g., by hollow fiber concentrator). The concentrated sample is chromotagraphed on heparin agarose, and bound material (bound directly or indirectly to heparin agarose) eluted using a linear buffered NaCl gradient. Pooled fractions are concentrated. C3/C3rp may be further purified using an antibody against C3/C3rp. Purity can be assessed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis ("SDS-PAGE") with appropriate protein stains. Approximate molecular weights of polypeptides are estimated by comparison of their mobility to the mobility of polypeptides of known molecular weights on SDS-PAGE. C3/C3rp has substantial sequence homology to normal human blood complement C3.
  • Purified antigen C3/C3rp
  • partially purified antigen or biological samples containing antigen may be used to produce antibodies that specifically bind to the antigen.
  • Antibodies that specifically bind are those with an affinity of about 10 ⁇ liters/mol or greater. Either polyclonal antibodies or monoclonal antibodies may be generated. Polyclonal antibodies may be produced by immunization of an animal and subsequent collection of its sera. It is generally preferred to follow the initial immunization with one or more booster immunizations prior to sera collection. Monoclonal antibodies are generally produced by the method of Kohler and Milstein ⁇ Nature 256:495-491, 1975; Eur. J. Immunol. 6:511-519, 1976).
  • lymph nodes and/or spleens of an animal injected with antigen in pure or impure form are fused with myeloma cells to form hybrid cell lines ("hybridomas" or "clones").
  • hybridoma secretes a single type of immunoglobulin specific for the antigen and, like the myeloma cells, has the potential for indefinite cell division.
  • Antigen in pure or impure form (“immunogen”) is used for the immunization.
  • the animals are immunized with at least 100 ng each of the immunogen, most preferably greater than 500 ng each.
  • the immunogen may be adsorbed to a solid phase matrix, preferably to nitrocellulose paper. The paper is then introduced into the animal. Techniques for introduction of the adsorbed antigen preparation include implantation (U.S. Patent No. 4,689,220) or solubilization of the solid phase and injection of the solubilized material (Knudsen, Anal. Biochem. 47:285-288, 1985).
  • the solid phase matrix may be solubilized in an appropriate organic solvent (e.g., DMSO) and either mixed with adjuvant or saline, or injected directly.
  • the immunogen may be injected in the absence of a solid matrix and/or adjuvant. Injection or implantation may be intraperitoneal, intra-foot pad, subcutaneous, intramuscular or intravenous, but preferably intraperitoneal.
  • the animals may also be injected with antigen complexed with adjuvant, such as Freund's adjuvant. Single or multiple booster immunizations are used. Between one and seven days prior to the fusion date, preferably on days one through four, intravenous injections of the immunogen may be given daily.
  • spleens or portions thereof are harvested from the immunized animals.
  • the lymph nodes may also be harvested and included in the cell preparation.
  • the harvested organs are minced using techniques which disrupt the structure of the organ, but which are not detrimental to the lymphocytes.
  • the organs are preferably minced with scissors, passed through a mesh screen and mixed with growth medium to enrich the preparation for lymphocytes.
  • the minced and strained tissue is harvested by centrifugation, then mixed with growth medium to form a cell suspension.
  • the red blood cells may be lysed by adding a hypotonic or hypertonic solution to the cell suspension.
  • a preferred method for cell lysis is to add distilled water to the suspensions and quickly return the suspensions to an isotonic state with a hypertonic sodium chloride solution. Any remaining tissue may be removed by filtration through gauze.
  • the harvested cell suspension is then mixed with a myeloma cell line, preferably one which is syngeneic with the immunized animal.
  • Myeloma cell lines from various species are widely available through, for example, American Type Culture Collection (ATCC), Rockville, Maryland. Myeloma cell lines commonly used include P3X63Ag8 (ATCC TIB 9), SP2/0-Agl4 (ATCC CRL 1581), FO (ATCC CRL 1646) and 210-RCY-Agl (Galfre et al., Nature 277:131, 1979).
  • the myeloma cells are cultured in an appropriate mammalian cell growth medium, a variety of which are generally known in the art and available from commercial sources.
  • Mammalian cell lines are routinely grown between 36°C and 40°C under conditions which maintain an optimal pH between 6.0 and 8.0, preferably about pH 7.2. pH may be maintained through the use of a variety of buffer systems known in the art.
  • a preferred buffer system involves growing the cells in a bicarbonate buffer in a humidified incubator containing CO 2 , preferably about 7% CO 2 .
  • the fusion between the lymphocytes from the immunized animal and the myeloma cells may be carried out by a variety of methods described in the literature. These methods include the use of polyethylene glycol (PEG) (Brown et al., J. Biol. Chem. 255:4980-4983, 1980) and electro fusion (Zimmerman and Vienken, J. Membrane Biol. 67:165-182, 1982).
  • An electrofusion generator is commercially available from Biotechnologies and Experimental Research, Inc., San Diego, California. Following the fusion, the cells are plated into multi-well culture plates, preferably 96-well plates.
  • a reagent which selectively allows for the growth of the fused myeloma cells over the unfused cells is added to the culture medium.
  • a preferred selection technique uses HAT (hypoxanthine, aminopterin, thymidine) selection. Other selection techniques may also be used depending on the myeloma cell line chosen.
  • Lymphocytes may be harvested from lymphoid organs, such as spleen or lymph nodes, or from whole blood as peripheral blood lymphocytes. The lymphocytes are put into culture in the presence of the appropriate immunogen. Often immuno stimulatory polypeptides will be added to the culture medium concurrently. At various times following the culturing of the lymphocytes in vitro, the lymphocytes are harvested and fused with a myeloma cell line as described above.
  • EBV transformed cells do not require fusion with a myeloma cell to allow continued growth in culture.
  • Thymocytes may be used as a feeder layer to condition the medium for the fused cells.
  • peritoneal macrophages or non-immune spleen cells may be used as a feeder layer.
  • Another alternative is to use conditioned medium from thymocytes or macrophages.
  • Thymocytes may be prepared from juvenile mice less than 8 weeks old. The thymus glands are harvested and minced using techniques which disrupt the thymus gland but are not detrimental to the thymocytes. This procedure is preferably carried out using scissors to mince the tissue, followed by passage of the tissue through a mesh screen. The minced and strained cell material is then harvested by centrifugation. Cell suspensions are made using growth medium. Any remaining connective tissue may be removed by filtration through gauze.
  • the fused cells are then analyzed for the production of antibody against the antigen.
  • This "screening" can be done by a wide variety of techniques, including Western blot, ELISA, immunoprecipitation, effect on biological activity assays and immunocytochemical staining. These techniques and others are well described in the literature. (See, for example, J. G. R. Hurrell (ed.), Monoclonal Hybridoma Antibodies: Techniques and Applications, CRC Press Inc., Boca Raton, Fla., 1982.) Introduction of a screening procedure permits further definition of antibodies of useful reactivity.
  • antigen purified from a biological sample of a patient with a bladder cancer may be used in any of the above-named techniques to define antibodies which react, for example, to determinants which are common to patients with the disease.
  • Hybridomas which secrete antibodies of interest are maintained in culture.
  • the cells are expanded in culture and at the same time may be cloned in such a manner as to obtain colonies originating from single cells. This provides for the monoclonal nature of the antibodies obtained from the hybridomas.
  • clones of cells are obtained, they are re-assayed for the production of the antibody of interest. These cells are then expanded in culture to allow for the production of larger amounts of the antibody. Methods for expansion of the cells include maintaining the cells in culture, placement of the cells in a bioreactor or other type of large-scale cell culture environment, or culturing the cells using various agar or gelatin carrier matrices. Antibodies are then isolated from the cell culture media.
  • Antibodies may be purified from conditioned media or ascites fluid by a variety of methods known in the art. These methods include ammonium sulfate precipitation, ion exchange chromatography (see Hurrell, ibid.) and high pressure liquid chromatography using a hydroxylapatite support (Stanker et al., J. Immunol. Methods 76:157, 1985).
  • a preferred method for purifying antibodies from conditioned media or ascites fluid utilizes a commercially available Protein A-Sepharose CL-4B column or Protein G Sepharose ® (Pharmacia, Piscataway, NJ; Sigma, St. Louis, MO) or ABX mixed ion exchange resin (JT Baker, Phillipsburg, NJ). Antibodies may be purified with these columns using conditions suggested by the manufacturer.
  • C3/C3rp is found to be associated with a variety of cancers, including colorectal, renal, bladder and cervical cancers, and may be detected in a variety of ways, including by detecting C3/C3rp itself or a nucleic acid molecule encoding C3/C3rp.
  • Methods for detecting the presence (i.e., qualitative or quantitative) of C3/C3rp include those based on physical properties, immunological properties, biochemical properties and combinations thereof (e.g., physical size of the molecule, nucleic acid sequence, amino acid sequence, binding by monoclonal or polyclonal antibodies, ligand binding, enzymatic properties, and combinations thereof).
  • C3/C3rp can be degraded (e.g., proteolytically activated) to produce C3b.
  • the production of C3b may be measured directly (e.g., using an anti-C3b antibody) or indirectly (e.g., by the ability of C3b to promote the lysis of target cells).
  • a nucleic acid molecule encoding C3/C3rp can be detected.
  • Such a nucleic acid molecule may be a deoxyribonucleic acid (DNA) or a ribonucleic acid (RNA).
  • a nucleic acid molecule encoding for C3/C3rp is detected by amplification of the nucleic acid.
  • a variety of methods may be utilized in order to amplify a selected sequence, including, for example, RNA amplification (see Lizardi et al., Bio/Technology 6:1197-1202, 1988; Kramer et al., Nature 339:401-402, 1989; Lomeli et al., Clinical Chem. 35(9):1826- 1831, 1989; U.S. Patent No. 4,786,600), and DNA amplification utilizing ligase chain reaction (“LCR”) or polymerase chain reaction (“PCR”) (see U.S. Patent Nos.
  • LCR ligase chain reaction
  • PCR polymerase chain reaction
  • PCR for example, the method may be modified as known in the art. Transcriptional enhancement of PCR may be accomplished by incorporation of bacteriophage T7 RNA polymerase promoter sequences in one of the primary oligonucleotides, and immunoenzymatic detection of the products from the enhanced emitter may be effected using anti-RNA:DNA antibodies (Blais, Appl. Environ. Microbiol.
  • PCR may also be used in combination with reverse dot-blot hybridization (Iida et al., FEMS Microbiol. Lett. 114:167-172, 1993). PCR products may be quantitatively analyzed by incorporation of dUTP (Duplaa et al., Anal. Biochem. 212:229-236, 1993), and samples may be filter sampled for PCR-gene probe detection (Bej et al., Appl. Environ. Microbiol. 57:3529-3534, 1991).
  • Primers for the amplification of a selected sequence should be selected from sequences that are highly specific to C3/C3rp and form stable duplexes with the target sequence.
  • the primers should also be non-complementary, especially at the 3' end, should not form dimers with themselves or other primers, and should not form secondary structures or duplexes with other regions of DNA.
  • primers (such as those described in greater detail below) of about 20 to 30 nucleotides are preferred, and can be easily synthesized using techniques well known in the art.
  • PCR products, and other nucleic acid amplification products may be quantitated using techniques known in the art (Duplaa et al., Anal. Biochem. 212:229-236, 1993; Higuchi et al., Bio/Technology 77:1026-1030).
  • a preferred embodiment involves assaying for the presence of specific messenger RNA (mRNA) encoding C3/C3rp. More specifically, for example, as described herein, a cell sample may be lysed and the mRNA isolated, amplified and examined for the presence of mRNA specific for C3/C3rp. A variety of procedures may be used to detect the presence of antigen-specific mRNA. A particularly preferred method includes RT-PCR (Reverse Transcriptase based Polymerase Chain Reaction) amplification of mRNA.
  • RT-PCR Reverse Transcriptase based Polymerase Chain Reaction
  • Detecting the presence of C3/C3rp in a sample has a variety of uses.
  • the present invention may be used for diagnostic purposes to screen warm-blooded animals, such as humans, for cancers such as colorectal cancer, renal cancer, bladder cancer or cervical cancer (depending upon the source of the particular sample).
  • cancers such as colorectal cancer, renal cancer, bladder cancer or cervical cancer (depending upon the source of the particular sample).
  • Preferred sample sources for a particular cancer would be evident to one of ordinary skill in the art.
  • using a voided urine sample one may screen for renal or bladder cancer.
  • the present invention may be used to monitor warm-blooded animals.
  • a preferred use is to follow patients who have been previously diagnosed and treated for colorectal cancer, renal cancer, bladder cancer or cervical cancer. Patients who are in remission (or may in fact be cured) can be monitored for the reappearance of colorectal, renal, bladder or cervical cancer. It may be desirable to use the present invention in conjunction with one or more other tests for colorectal, renal, bladder or cervical cancer to confirm positive or negative results obtained from use of the present invention.
  • C3/C3rp may be serving as a "decoy" that interferes with the operation of the complement system (thus permitting tumor cells to escape surveillance by the host's immune system); or the C3/C3rp production and degradation may result in product(s) that serve as a growth factor (i.e., an autocrine function) for tumor cells.
  • a growth factor i.e., an autocrine function
  • C3/C3rp may be modulated in a variety of ways.
  • C3/C3rp may be modulated by interrupting the production of C3/C3rp by tumor cells or "inactivating" C3/C3rp (e.g., blocking C3/C3rp, its conversion or its effect) following production by tumor cells.
  • a preferred method of interrupting the production of the antigen is by use of DNA, or PNA (peptide nucleic acid), constructs with base sequence complementary to the antigen's mRNA.
  • DNA or PNA (peptide nucleic acid)
  • PNA peptide nucleic acid
  • Such an approach is generically termed antisense technology.
  • the C3/C3rp antisense DNA is inserted into an appropriate vector (virus) which delivers it to the tumor cells.
  • the antisense construct specifically binds to mRNA coding for the C3/C3rp, thereby preventing its translation.
  • Primary among other methods which may be used to interrupt production of the antigen is the use of specific molecules which block the transcription of the specific gene or genes coding for the C3/C3rp. Chemicals designed to block the ability of the tumor cell to produce antigen are preferably delivered in the vicinity of the tumor, rather than systemically.
  • reagents to inhibit the activity of, or interfere with the binding sites on, C3/C3rp.
  • One family of such reagents includes monoclonal antibodies, or fragments thereof (e.g., antigen binding fragments).
  • an appropriate concentration of an antibody which is reactive with C3/C3rp increases the rate of lysis of cancer cells by one of the complement pathways (Example III).
  • delivery is preferably administered to the tumor site, rather than systemically.
  • reagent affinities should be at least about 10 6 liters/mole and doses should be within the range of about 0.01 ⁇ g/kg body weight to 10 mg/kg body weight.
  • C3/C3rp may be modulated such that the killing of cancer cells by the complement system is promoted.
  • the present invention also provides for the modulation of C3/C3rp as a means for treating autoimmune disorders. It is well known that the lysis of normal cells by the immune system is a significant source of tissue destruction associated with autoimmune disorders and myocardial infarcts.
  • autoimmune antibodies deposit on a basement membrane or cell surface in the vicinity of a joint or bursa, resulting in the local activation of the classical complement pathway.
  • C3 is a central element for both complement pathways (i.e., classical and alternative) after its proteolytic activation to form C3b.
  • an appropriate concentration of an antibody which is reactive with C3/C3 ⁇ inhibits the conversion of C3 to C3b, thereby preventing lysis of targeted cells.
  • Alternative binding partners include small organic molecules and amino acid based molecules such as peptides. Thus, molecules which bind to C3/C3 ⁇ may be used to inhibit C3-mediated cell destruction.
  • proteins related to: decay accelerator factor (DAF) or DAF related proteins (“DAF ⁇ ”), complement Factor I (CFI) or CFI related proteins (“CFL ⁇ ”), complement binding protein CRl or CRl related proteins (“CRl ⁇ ”) and complement binding protein CR3 or CR3 related proteins (“CR3 ⁇ ”) are found to be associated with a variety of cancers, including colorectal, renal, bladder and cervical cancers.
  • DAF, DAF ⁇ , CFI, CFIrp, CRl, CRl ⁇ , CR3 or CR3 ⁇ may be detected in a variety of ways.
  • one or more of DAF, DAF ⁇ , CFI, CFI ⁇ , CRl, CRl ⁇ , CR3 and CR3 ⁇ may be detected, or a molecule encoding one or more of these proteins may be detected.
  • Methods for detecting the presence (i.e., qualitative or quantitative) of one or more of DAF, DAF ⁇ , CFI, CFI ⁇ , CRl, CRl ⁇ , CR3 or CR3 ⁇ include those based on physical properties, immunological properties, biochemical properties and combinations thereof (e.g., physical size of the molecule, nucleic acid sequence, amino acid sequence, binding by monoclonal or polyclonal antibodies, ligand binding, enzymatic properties, and combinations thereof).
  • the present invention may be used for diagnostic pu ⁇ oses to screen warm-blooded animals, such as humans, for cancers such as colorectal cancer, renal cancer, bladder cancer or cervical cancer (depending upon the source of the particular sample).
  • the present invention may be used to monitor warm-blooded animals.
  • a preferred use is to follow patients who have been previously diagnosed and treated for colorectal cancer, renal cancer, bladder cancer or cervical cancer.
  • CFI ⁇ , CRl, CRl ⁇ , CR3 or CR3 ⁇ serves a beneficial pu ⁇ ose for the cancers.
  • One or more of these molecules may be interfering with the complement system, thus permitting tumor cells to escape surveillance by the host's immune system.
  • the present invention provides for the modulation of one or more of DAF, DAF ⁇ , CFI, CFI ⁇ , CRl, CRl ⁇ , CR3 or CR3 ⁇ as a means of treating cancers. It will be evident to those of ordinary skill in the art that these proteins may be modulated in a variety of ways. For example, one or more of these proteins may be modulated by interrupting the production by tumor cells or "inactivating" following production by tumor cells.
  • an antibody that binds to one of these proteins and blocks the protein's activity by use of an antibody that binds to one of these proteins and blocks the protein's activity, the rate of lysis of cancer cells by one of the complement pathways may be increased.
  • An antibody may be replaced, or supplemented, with a small organic molecule or amino acid based molecule (such as a peptide) with similar functional properties to the antibody.
  • expression of one or more of these proteins may be inhibited by use of antisense DNA or PNA (peptide nucleic acids).
  • PNA peptide nucleic acids
  • the antigen source for immunization was a pool of Heparin-Agarose fractionated urines from clinically diagnosed bladder cancer patients. Twenty-four hour urine samples were centrifuged in a Beckman centrifuge (Fullerton, CA), Model #J2-21, S/N 5539, using a JA-10 rotor at 6,000 ⁇ m for 20 minutes. The clarified urine sample was then concentrated using an Amicon stirred cell, 76 mm, (cat# 5124) fitted with a YM30 membrane MWCO 30,000 dalton (Amicon, cat# 13742) or a Microgon hollow fiber concentrator, 50,000 MWCO (cat# M15S-260-01N) to approximately 100X concentration.
  • the concentrated sample was diluted 1:2 with 25 mM Tris-HCl pH 7.4 and loaded onto a column of Heparin- Affigel (BioRad, Richmond, CA, cat# 153-6173), equilibrated in 25 mM Tris-HCl pH 7.4, at a flow rate of 2.0 ml/min.
  • the sample was followed with equilibration buffer until the A280 elution profile returned to background.
  • Bound material was eluted with a linear NaCl gradient from 0 to 250 mM NaCl in 25 mM Tris-HCl pH 7.4. Eight ml fractions were collected and fractions from the trailing half of the elution peak were pooled.
  • mice Five female BALB/c mice, of 8-10 weeks of age, were immunized intraperitoneally with 0.2 ml of a 1 :1 emulsion of Pool II in Freund's Complete Adjuvant (Difco, Detroit, MI). Three weeks later, booster immunizations of 0.1 ml containing 10 ⁇ g of protein of an emulsion in incomplete Freund's Adjuvant was administered to the rear footpads and peritoneum. Ten days later each mouse was sampled for antibody response via retro-orbital bleeds and the sera were tested via an ELISA described below for titers. Mouse number 340 showed the highest titer and was chosen for fusion four days after boosting in the footpads and peritoneum with 15 ⁇ g of Pool II in phosphate buffered saline.
  • IMDM Iscove's Modified Dulbecco's Medium
  • HT hypoxanthine/thymidine
  • the fusion was divided into two parts, in the first part forty-eight 96 well plates were seeded at the above density in media containing 1% fetal bovine serum (FBS). The second part consisted of 49 plates seeded at the same density in media containing 10% FBS. A total of 97 plates, or 9,312 wells were used.
  • the plates were incubated at 37°C in 7% CO 2 at 100% humidity. The next day 100 ⁇ l of selective media consisting of IMDM-HT with 2x methotrexate (8 x 10 " M) and appropriate FBS concentration was added. The plates were returned to the incubator and not disturbed for six days. On day seven the plates were removed from the incubator and approximately 150 ⁇ l of media was removed via aspiration with a sterile eight place manifold. Complete IMDM with HT and appropriate FBS was added to each well using a Brinkman eight place pipette. The plates were returned to the incubator for another five to six days before screening. The fusion plates were examined each morning for wells showing growth levels suitable for screening, and were analyzed that day.
  • the plates containing the 1% FBS medium were clearly lagging in growth, and were therefore supplemented to 10% FBS. Thereafter, those wells selected from the plates initially plated in 1% FBS were designated as MOFI-followed by a number indicating the order of selection, those from the 10% FBS plates were designated with the MOFX prefix.
  • Wells selected via the screening assays were immediately transferred to 24 well plates containing 1 ml of complete IMDM containing 10% FBS.
  • a sample of cells was also used to immediately re-clone the hybridomas by a serial limiting dilution procedure. This consisted of transferring a 10 ⁇ l sample of cells from the chosen well of the 96 well plate to the first well of a fresh 96 well plate previously filled with 100 ⁇ l of complete IMDM with 10% of a cloning supplement prepared from murine macrophages and thymocytes (Condimed, Boehringer-Mannheim Co ⁇ ., Indianapolis, IN).
  • Cells from the first well were serially diluted in the first column of wells by transferring 100 ⁇ l from the first well to the second, then from the second to the third, etc. The remaining 100 ⁇ l removed from the last well of the column is transferred back to the first well.
  • the wells of the first column were then serially diluted across the plate by transfer of 50 ⁇ l of cell suspension using an 8 place pipette.
  • 100 ⁇ l of cloning media was added to each well, and the plates incubated for approximately two weeks before subclones were ready for re-screening. Following growth in the 24 well plates, the clones were transferred to six well plates with 5-6 ml of culture media, the plates were incubated until near confluent growth was observed. A sample of the cells were removed for storage in a cryogenic freezer in 5% DMSO in FBS, and the remaining cells were transferred to a T-75 flask with 10 ml media for producing spent media for further testing.
  • the titer assay was carried out by coating Pool II (described above) antigen adjusted to 4 ⁇ g/ml in 0.1 M carbonate buffer, pH 9.6, directly to polystyrene plates. Each well received 50 ⁇ l of coating solution and the plate was covered and incubated at 37°C for 2 hours, after which time it was washed twice with phosphate buffered saline (PBS) in a Denley strip well washer. The plate was blocked by the addition of 100 ⁇ l of a 1% gelatin hydrolysate, 2% sucrose solution in 50 mM Tris-HCl, pH 7.5, at 37°C for 1 1/2 hours (all reagents from Sigma).
  • PBS phosphate buffered saline
  • the plate was again washed twice with PBS, then two-fold serial dilutions of mouse serum, starting at 1 :100, into 10% normal horse serum in PBS, were added row- wise to the plate at 50 ⁇ l per well.
  • the plate was incubated at 37°C for 1 hour, washed 4 times in PBS, and 50 ⁇ l of affinity purified goat anti-mouse IgG- horseradish peroxidase (HRP) conjugate (Tago, Burlingame, CA) diluted 1 :5000 in 10% horse serum in PBS added to each well. This was allowed to incubate for 1 hour at 37°C.
  • HR horseradish peroxidase
  • the plate was washed with PBS 4 times, and 50 ⁇ l of substrate (K-Blue, ELISA Technologies, Lexington, KY) was added and the plate allowed to develop for 10 minutes at room temperature before stopping the reaction via the addition of 100 ⁇ l of 2M phosphoric acid solution in water (Sigma).
  • the optical density of the wells were read at 450 and at 410 nm in a BioTek EL311 plate reader. Readings which were off scale at 450 nm were calculated from the corresponding reading at 410 nm by the method of Madersbacher and Berger, J. Immunol. M. 73S.T21-124, 1991.
  • the fusion was screened for antibody production by use of the following fusion screen.
  • Antibody binding was tested with: (a) two clinically diagnosed bladder cancer patient urines, stages T2III and T3III, (diluted 1 :80), (b) two pools of normal human urines (diluted 1 :15), (c) human type IV collagen (diluted to 4 ⁇ g/ml), all dilutions in 25 mM Tris-HCl, pH 7.5, and (d) pooled human red blood cells (Gamma Biologicals, Houston, TX) diluted into PBS and coated onto poly-lysine coated plates.
  • alkaline phosphatase conjugate of a similar antibody was used (KPL, Gaithersburg, MD) followed by use of PNPP (p-nitrophenyl phosphate) substrate.
  • Controls were used for each assay, negative control was fresh IMDM with 10% FBS, positive controls were monoclonal anti-human collagen (Sigma C1926), and monoclonal anti-hlgA (Al.1.2.4, Bard Diagnostic Sciences, Inc., Redmond, WA), both of which showed high binding to all test antigens except the red blood cells. Criteria for selection were high binding to cancer urine plates (OD>l), low binding to normal urines and other test antigens (ODO.5). Others which showed high antibody levels in different patterns with respect to the test antigens were also selected for potential research uses.
  • Subclones were screened by several assays. First, the fusion assay was again used then, following expansion in culture of selected subclones, an abbreviated ELISA was employed using normal urine pool I and the two advanced stage urines used in the fusion assay. The testing was carried out at dilutions of 1 :10 and 1:100 for the early subclones, and an additional dilution of 1 :1000 for the later subclones. In several of the subclone assays the addition of urine from a patient with a lower grade cancer was included.
  • Urine specimens from five cancer patients and two normal urine pools were tested in an enzyme immunoassay format utilizing a commercially-available anti-C3 MAb, which was reactive, as well, with the activated C3, designated C3b. These samples were also tested with several monoclonal antibodies (MAbs) produced by immunizing Balb/c mice with a partially purified protein fraction from urines of human bladder cancer patients (see Example I) and identified as MOF MAbs.
  • the normal urine pools were each composed of equal volumes from five individuals, non-randomly selected to include males, females, Caucasians, Asians and Blacks, all over the age of 40.
  • the urine specimens were diluted in 1 mM carbonate buffer, pH 9.6.
  • the normal pools were diluted 1 :8 and the specimens from the cancer patients were each diluted (dilutions between 1:15 and 1 :80) to yield an antigen concentration of approximately 100 U/mL in the Bard BTA TRAKTM assay (Bard Diagnostic Sciences, Redmond, WA).
  • Each diluted specimen was pipetted into individual wells of a High- Binding 96-well microtiter plate (LabSystems, Helsinki, Finland) and allowed to incubate overnight at 2-8°C. Samples were removed from the plate by aspiration.
  • a blocking buffer 1% gelatin hydrolysate, 2% sucrose, 50 mM Tris buffer, pH 7.5; all reagents from Sigma, St. Louis, MO was then added and allowed to incubate for 1 hour at 37°C.
  • MAbs as noted above, were diluted in Bard ® BTA TRAKTM diluent (Bard Diagnostic Sciences) to a final concentration of 1 ⁇ g/mL. They were then added to individual wells and allowed to incubate for 1 hour at 37°C. MAb solutions were then removed by aspiration and the plates were washed four times on a plate washer (BioTek, Winooski, VT). Next, a reporter conjugate (alkaline phosphatase- labeled goat anti-mouse IgG, human serum-absorbed, Kirkegaard and Perry Labs, Gaithersburg, MD) at a concentration of 0.5 ⁇ g/mL in assay buffer was added to each well.
  • Bard ® BTA TRAKTM diluent Board Diagnostic Sciences
  • C3b which is also reactive with the activated C3 molecule (designated C3b), was tested on five cancer patient urine specimens (PES, GS3, GS4, GS5, OCK), each diluted to read 100 U/mL on the Bard BTA TRAKTM assay (approximately 1 :15 to 1 :80).
  • the commercial anti-C3 MAb reacted strongly with C3, but was unreactive with the two normal pools, unreactive with the five specimens from bladder cancer patients, and unreactive with the other complement factors tested. In fact, all of the MAbs were unreactive with the other complement proteins, except for Clq, which is known to bind the Fc region of mouse IgGl antibodies.
  • MOF MAbs X46.3, X87.2, X67.2, and X59.1 are all reactive with one or more of the five patient specimens and su ⁇ risingly the C3 antigen, but unreactive with the normal urine pools and the other complement proteins (again with the exception of Clq, to which most of the MAbs show at least some level of reactivity, presumably due to binding of the Fc region of the MAb to the Clq).
  • a sandwich EIA was constructed utilizing commercially-obtained anti-C3 (BioDesign, Kennebunk, ME) and alkaline phosphatase-conjugated MAb X46.3. Antibodies were purified by chromatography on immobilized Protein G or Protein A by standard techniques. Although antibody-enzyme conjugates could be prepared using a variety of coupling techniques (for review see Scouten, W.H., Methods in Enzymology 735:30-65, 1987), a minor variation of a method described by S. Hashida and E. Ishikawa (Anal. Lett. 18, 739:1143-1155, 1985) was used.
  • 96-Well microtiter plates were coated with anti-C3 at a concentration of 5 ⁇ g/mL in carbonate buffer and blocked, as described above in Example LA for plates coated with urine specimens and complement proteins.
  • Urine samples from healthy donors, from patients with bladder or renal cancer, or from those with non-cancerous renal diseases were diluted 1 :50 in carbonate buffer (25 mM pH 9.6) and added to the antibody-coated plates. After incubation at 37°C for 1 hour, the samples were aspirated from the wells, which were then washed four times with Bard BTA TRAKTM wash buffer using a Dynatech (Chantilly, VA) plate washer. The alkaline phosphatase-conjugated MAb X46.3, at a concentration of 1.0 ⁇ g/mL in Bard BTA TRAKTM assay diluent, was added to each well (100 ⁇ L per well).
  • Cells were grown in culture with specifications for media as defined by ATCC and were harvested by trypsin-EDTA treatment (0.25% trypsin, 10 mM EDTA; reagents from Sigma) after the cells had reached a confluence of 90% or greater. Cells were counted in a hemacytometer by standard methods using a phase contrast microscope (American Optical). After aliquots were removed for counting, the cells were pelleted at lOOOxG at room temperature. Cell pellets were prepared for staining with MAbs by quick freezing in liquid nitrogen. The frozen pellets were sectioned on a cryostat microtome (Bartles & Stout) and then fixed in cold acetone (-20°C) for 15 minutes.
  • trypsin-EDTA treatment 0.25% trypsin, 10 mM EDTA; reagents from Sigma
  • the primary MAb (X46.3; ATCC Accession No. HB- 12064, American Type Culture Collection, Rockville, MD) was diluted to a concentration of 2 ⁇ g/mL in phosphate-buffered saline (PBS), pH 7.2, and incubated on the sections for 30 minutes at room temperature.
  • PBS phosphate-buffered saline
  • the conjugated secondary MAb (goat anti-mouse IgG peroxidase conjugate, human serum absorbed, Kirkegaard and Perry Laboratories, Gaithersburg, MD), at a concentration of 0.25 ⁇ g/mL in PBS, was incubated on the slide at room temp for 30 minutes. Following three washes with PBS, the specimens were developed with 3,3'-diaminobenzidine (DAB). Unless otherwise indicated all reagents were from Sigma Chemical.
  • DAB 3,3'-diaminobenzidine
  • Results were determined by SDS-PAGE of the reaction mixtures under reducing conditions (50 mM DTT) on a 4-12% gradient gel (Novex, San Diego, CA). (Unless specified otherwise, all reagents are from Sigma, St. Louis, MO.) The gel was scanned with a BioRad GelDoc scanner. The intensities of the bands measured in this way were converted to percentage of C3b remaining. Two control lanes were included. The one containing the reaction mixture in the absence of MAb was used to represent 100 percent degradation, while the other containing the reaction mixture with no Factor H was used to represent 0 percent degradation. A photograph of the SDS-PAGE gel from the experiment described above is shown in Figure 2. The results derived from scanning the gel are summarized in Table III.
  • Lane 1 of the gel shows the molecular masses of C3b and Factor I. Note that C3b is 108 kD, and is uncleaved in the presence of Factor I alone. Lane 2 shows the result after addition of Factor H to the mixture of C3b and Factor I. Note the complete degradation of C3b and the appearance of a smaller 38 kD degradation product. Lanes 3 and 4 show the inhibition of C3b degradation by MAb X46.3. Rows 2 and 3 of the accompanying Table III summarize the effect of X46.3 inhibition, which is 83.7% when the reaction mixture contains 30 ⁇ g of MAb (Row 3, Column 3). Lanes 5-6 are negative controls containing antibody non-inhibitory to the Factor I/Factor H- mediated C3b degradation.
  • Lanes 7-8 are positive controls containing anti-Factor H antibody inhibitory to the Factor I/Factor H-mediated C3b degradation. Lanes 9-10 illustrate that Mab X87.3 is also capable of inhibiting Factor H+Factor I-mediated degradation of C3b, although not as efficiently as MAb X46.3 Rows 4 and 5 of Table III summarize the effect of this inhibition by X87.3.
  • Standard guinea pig complement was treated with EGTA to chelate calcium. Then MgCl 2 , which is required for the activity of the alternative complement pathway (ACP), was added (all reagents form Sigma). The mixture was incubated for
  • a series of ELISAs were constructed, patterned after that described in Example II. A, in order to test for the presence of proteins of the alternative complement pathway (ACP) in the urine of patients with bladder, cervical or renal cell cancer.
  • ACP alternative complement pathway
  • C3b was produced from C3 as referenced above in Example III.A. Goat polyclonal antibodies to human Clq and C2, and monoclonal antibodies to Clq, C3b, C4bp, Bb, Factor D, Factor H, Factor I and properdin were available from Quidel (San Diego, CA).
  • cells were harvested by trypsin-EDTA treatment (0.25% trypsin, 10 mM EDTA; reagents from Sigma) after the cells had reached a confluence of 90% or greater.
  • trypsin-EDTA treatment 0.25% trypsin, 10 mM EDTA; reagents from Sigma
  • cells were harvested from parallel cultures at subconfluent densities within the ranges 10-20%, 20-50%, 50-90%, as well as »100%.
  • Cells were counted with a hemacytometer by standard methods using a phase contrast microscope (American Optical). After aliquots were removed for counting, cells were pelleted at lOOOxG at room temperature.
  • RT-PCR cDNA was prepared from mRNA present in preparations of total cellular RNA from various cancer cell lines, using Reverse Transcriptase plus Random Hexamer primers (50 pmoles per 20 ⁇ L reaction), according to the protocol in a commercially-available RNA PCR kit (Perkin-Elmer, Foster City, CA). A 2-3 ⁇ g quantity of total RNA was used for each reaction, with the polymerization temperature and time set at 42°C and 90 minutes, respectively.
  • PCR of mRNA for DAF was performed with the primer pairs 797us (ATGATGAAGGAGAGTGGAGTGG) and 1269ds (CTCCTTGCTCTGTTGACATTCC), each used at a final concentration of 0.3 ⁇ M in the reaction mixture.
  • Primers were synthesized by Midland Certified Reagents (Midland, TX) and were purified by anion- exchange chromatography. Annealing conditions were 62°C for 30 seconds and extension was at 72°C for 90 seconds, with 40 cycles.
  • PCR of mRNA for the complement receptors was performed with primer pairs 302064 (CRl), 302065 (CR2) and 302066 (CR3), each used at 10 nmoles per 100 ⁇ L reaction mixture.
  • Primers were purchased from Stratagene Cloning Systems (La Jolla, CA). Annealing conditions were 60°C for 60 seconds and extension was at 70°C for 2 minutes, with 30 cycles; an additional 10 cycles were performed with the extension step set at 2 minutes and 30 seconds, followed by a final extension for 7 minutes.
  • the size of the cDNA products were determined by electrophoresis at 90 volts for 90 minutes on 1.5% agarose gels (Sigma), followed by staining with ethidium bromide (Sigma), and destaining in deionized water. Positive results were defined to be a 521 -base pair product for CRl and ad 345-based pair product for CR3.
  • Figure 5 (CRl), Figure 6 (CR2) and Figure 7 (CR3) show the results for amplification of complement receptor cDNA from a variety of cell lines. Note that all cell lines were negative for CR2. CRl cDNA of the correct size was produced by a number of cell lines. HeLaS3 was an exception, in that a second product of unexpected size (approximately 350 base pairs) was observed. RT-PCR for CR3, a complement protein coded for in the same chromosomal locus as C3, yielded extremely heterogeneous amplicons. The HeLaS3, LS174T, HL60 and HTB33 cell lines all made products which were much larger than expected size in addition to the product of the correct size for cognate CR3.
  • DAF CD55
  • complement receptors 1 and 3 CD35 and CD1 lb
  • CRl The role of CRl as a negative regulator of both complement pathways and its structural relationship to DAF and factor H, two other proteins coded within the RCA gene locus, is highly significant. Upregulation of CRl by cancer cells would augment their ability to evade lysis through either complement pathway.
  • CFI Complement Factor I
  • PCR of mRNA for complement Factor I was performed with the primer pair FI7us (GCAAGGTCACTTATACATCTCAAGAGC) and FIG683ds (CCCATTCACACACTGAAAGAAGTCATCC), each used at 0.50 nmoles per 100 ⁇ L reaction mixture.
  • Primers were synthesized by Midland Certified Reagents (Midland, TX) and were purified by anion- exchange chromatography. Annealing conditions were 62°C for 30 seconds and extension was at 72°C for 90 seconds, with 40 cycles.
  • Positive results defined to be the identification of a 472-base pair amplicon, were determined by electrophoresis at 90 volts for 90 minutes on 2% agarose gels (Sigma), followed by staining with ethidium bromide (Sigma), and destaining in deionized water.
  • one TCC line (HTB-9) and three cervical liens (HeLaS3, C4ii and HTB-33) are positive for amplicons of the correct size, while one cervical line (C4i), one colon line (LS174T), one TCC line (HTB-5), one prostate line (PC-3) and the negative controls (NHEK and myeloblastoma HL-60) are negative.
  • Reactivity of X67.2 with cervical specimens was initially identified by a dot-blot procedure.
  • Samples were tested either neat or diluted 1 :10 WITH Bard BTA TRAKTM assay diluent. All assay steps were performed at room temperature. Samples of 2 ⁇ L were applied to nitrocellulose (Millipore, Bedford, MA) and air dried for approximately 30 minutes, followed by blocking (as above in Example LA) for 2 hours. The blots were rinsed once with BTA TRAKTM assay diluent.
  • the blots were then covered with a solution of MAb X67.2, at a concentration of 1 ⁇ g/mL in assay diluent, incubated for 1 hour, and rinsed twice with assay diluent. Specimens which were reactive with the MAb were identified by incubation of the blot for 1 hour with alkaline phosphatase-conjugated goat anti -mouse IgG (Kirkegaard and Perry, human serum absorbed) at a concentration of 0.5 ⁇ g/mL in BTA TRAKTM assay diluent. Finally, development of the blot with substrate was performed as described in Example II. A.
  • MAb 67.2 discriminates patients with squamous cell cancer of the cervix from patients with infectious diseases, normal healthy individuals and individuals with dysplastic disease.
  • Human complement receptor CR3 consists of two polypeptide chains, designated CD18 and CDl lb.
  • the CD18 polypeptide is also found in the complement receptor CR4.
  • the CDl lb polypeptide chain (GenBank file accession number M18044, locus HUMLAPA) is though to be a subunit only of CR3.
  • the CD1 lb chain is also referred to as MAC1, reflecting the belief that the molecule is expressed specifically in circulating monocytic macrophage cells. These cells are a subset of the peripheral blood leukocyte fraction (PBL).
  • PBL peripheral blood leukocyte fraction
  • a set of PCR primers (Table VI) was designed from the sequence listed in GenBank file Ml 8044 and was used in RT-PCR reactions to amplify the mRNA isolated from the human myeloid leukemia cell line HL-60 and encoding the CDl lb protein ( Figure 10).
  • the PCR primers were synthesized by Midland Certified Reagents, Midland, Texas.
  • RNAzol Tel-Test, Inc., Friendswood, Texas
  • suitable cell numbers were obtained by culturing cells to 90% confluency in a size T162 flask (Corning-Costar, Cambridge, MA), in a suitable cell culture medium, as specified by the American Type Culture Collection (ATCC, Rockville, MD).
  • Total cellular RNA from peripheral blood leukocytes was obtained after fractionating fresh whole blood on a gradient of Histopaque (Sigma, Catalog #1077), according to the method provided by the manufacturer, and collecting the leukocyte fraction. Ribonuclease-free deoxyribonuclease I (Boehringer-Mannheim, Indianapolis, IN, Catalog #776785) was used, according to the manufacturer's instructions, at 1 unit per microgram of target nucleic acid, in order to remove contaminating DNA from the RNA preparations. b. Total RNA ranging between 0.5 and 5 ⁇ g, but optimally 2-3 ⁇ g, in a volume of up to 5 ⁇ L was used in each reaction.
  • RNA was mixed with a Reverse Transcription Master Mix (RTMM), formulated as described below. All reagents were from Perkin-Elmer Applied Biosystems, Foster City, CA, and may be purchased together as the Gene Amp RNA PCR Kit, Part No. N808-0017, and used as described below. Reverse Transcription Master Mix
  • RTMM Reverse Transcription Master Mix
  • RTMM The 16.5 ⁇ L volume of RTMM was mixed with the solution of total RNA and the final volume adjusted to 20 ⁇ L (up to 22 ⁇ L allowable) with molecular biology grade (ribonuclease-free) water (Catalog #2-538561, 5'-3' Inc., Boulder, CO).
  • the reaction was performed by allowing the RTMM containing the random hexamer primers to anneal with target RNA in the sample for 15 minutes at room temperature (22°C). The reaction mixture was then raised to 42°C and polymerization allowed to proceed for 90 minutes. Following this incubation, the reverse transcriptase (RT) was inactivated by heating at 99°C for 5 minutes.
  • the RT reaction can be performed on a suitable thermal cycler, such as the Perkin-Elmer Gene Amp Model 2400, or in a series of water baths or incubators.
  • PCR Amplification of CR3 (CD1 lb) cDNA a. Amplification of the cDNA resulting from the RT reaction is performed by using between 2.5 and 10 mL of the RT product. Typically, a volume of
  • PCR Master Mix 2.5 ⁇ L of the RT product is combined with 47.5 mL of a PCR Master Mix (PCRMM), as listing below.
  • PCRMM PCR Master Mix
  • PCR amplification proceeds after the samples are mixed. First, a one minute incubation at 95°C is used to melt any cDNA/mRNA pairs or cDNA/annealed primer pairs. Then 30 cycles are performed, each including a 94°C melt for 20 seconds, annealing for one minute at a temperature which is unique for each primer set, as specified below, and a two-minute extension at 70°C. As necessary, up to 10 additional cycles of amplification may be performed under the same protocol as just described, except for an increase of 30 seconds in the extension time. c. Assessment of the amplified sample is carried out by agarose gel electrophoresis.
  • TAE buffer Tris-Acetate-EDTA, Sigma, Catalog #T4038
  • the sample to be examined can then be mixed with a suitable sample buffer such as Gel Loading Solution (Sigma, Catalog #G2526).
  • the sample is then electrophoresed at 90V, using a power supply such as the BioRad PowerPac 3000, until the two tracking dyes are separated. This takes approximately 90 minutes at room temperature.
  • the electrophoresed product is then visualized by submersion of the gel into a solution prepared by adding a small amount (0.1 mL) of a saturated solution of ethidium bromide (Sigma, Catalog #E7637) in Molecular Biology Grade water to 30 mL of the TAE buffer. After staining for 15 minutes at room temperature, the gel is transferred to a clean vessel and destained with two changes TAE buffer, with a 10-minute incubation in each.
  • the stained amplicon can be viewed under a suitable UV source such as an Ultra-Lum Transilluminator (Carson, CA).
  • Downstream primers are always designated "RT"
  • Total cellular RNA from peripheral blood leukocytes also yielded product of the expected size. This product is presumed to be derived from the monocytic fraction of the blood cells, as the monocytes are known to comprise approximately 40% of the white cells and to make complement receptors and proteins.
  • Amplification of cDNA with the 18-933 primer set is expected to yield a 915 base pair product, as shown in the gel for most cell lines tested.
  • a product of approximately 830 base pairs is produced by a false priming of the 933RT oligonucleotide at position 853, and a 505 base pair product can be produced by a false prime at position 523.
  • sequence similarities to the 18M primer can allow a false prime to occur at base positions 168, 192 and 672. All of the false priming sites have at least 4 mismatches from the correct target sequence. However, point mutations in the DNA could lead to an increased probability of a false prime, resulting in a product of an unexpected size.
  • One way to maximize the fidelity of the priming event is by employing higher annealing temperatures, thus increasing the stringency of base matching requirements.
  • a 65°C annealing temperature was used with the 18-933 primer set in these amplifications, in contrast to annealing temperatures of 50-60°C more typically used in PCR.
  • point mutations that have occurred in the 18-933 priming regions would be predicted to have brought the false primed sequences into closer agreement with the cognate sequences.
  • An alternative explanation for the presence of a product of an unexpected size is a splicing variation in the messenger RNA.
  • Alternate splices occur when one or more exons are deleted from an expressed gene. The resulting mRNA is smaller than the original gene product, in some cases very much smaller.
  • a relevant example of alternate splicing occurs with the complement Factor H mRNA. Alternate splicing within factor H proteins has been reviewed by Zipfel and Skerka ⁇ Immunology Today 75:121-126, 1994).
  • the presence of an unexpected amplicon from the 18-933 primer set is illustrated for various cell lines in Figure 10, in which the material in each lane is as follows, beginning from the left: Lanes 1 and 8, molecular weight markers; Lane 2, HL60; Lane 3, HTB9; Lane 4, HeLaS3; Lane 5, LS174T; Lane 6, 33CO; Lane 7, SW480; Lane 9, MCF7; Lane 10, PC3; Lane 11, C4ii; Lane 12, HTB5, Lane 13, negative control; Lane 14, no target.
  • the unexpected amplicon is approximately 420 bases and is heavily expressed in the colon lines LS174T, 33CO, SW480; in the cervical lines HeLaS3, C4i and C4ii; and in the bladder line HTB5.
  • the size of the amplicon can be determined by reading the 100 base pair interval molecular weight standard ladder (MWS) in Figure 10. Note that only a small amount of this product is seen in the HL60 myeloid line. Note also that the 420 base product is the only amplicon produced by the breast cancer line MCF7 under the conditions used.
  • Figure 11 shows the results of amplification of cDNA from peripheral blood leukocyte specimens using the same 18-933RT primer set. The samples on the gel are as follows, beginning from the left: Lane 1, molecular weight markers; Lane 2, PBL LW; Lane 3, PBL RP; Lane 4, PBL JS; Lane 5, PBL HR; Lane 6, PBL SA; Lane 7, PBL DB; Lane 8, no target.
  • primer sets from the list shown in Table VI are also useful in detecting CR3/CDl lb product.
  • Examples of the cancer cell line surveys are shown in Figures 12 and 13 using the primer sets 129-933RT and 2665-3103RT, respectively.
  • the amplification conditions were the same as those described above, with the annealing temperature set at 65°C for the 129-933 RT primers and at 70°C for the 2665-3103RT primers.
  • Lane 2 the cell lines tested, listed from left to right, beginning with Lane 2, are as follows: Lane 2, HTB9; Lane 3, HeLaS3; Lane 4, LS174T; Lane 5, 33-CO; Lane 6, SW480; Lane 7, MCF-7; Lane 9, PC3; Lane 10, C4ii; Lane 11, COLO320; Lane 12, 841CON; Lane 13, COLO201. Lanes 1 and 8 contain molecular weight standards (in 100 base increments) and Lane 13 shows a control in which target was absent. The survey of cell lines reproduced in these figures show that four of the six colon cell lines possess CR3 mRNA.
  • 84 ICON is derived from normal colon tissue and LS174T is from a mucin-producing adenocarcinoma.
  • the cervical cancer line HeLaS3 and breast cancer line MCF-7 were also positive for CR3 expression with these two primer sets.
  • the bladder cancer line HTB9, the prostate cancer line PC3, and the cervical cancer line C4ii did not produce levels of CDl lb mRNA detectable in these assays.
  • the tissue was then homogenized by multiple passes in the tissue grinder with a tight-fitting pestle, while immersed in ice.
  • the lysate was extracted with equal volumes of phenol and chloroform/isoamyl alcohol.
  • the aqueous phase was aspirated and re-extracted with an equal volume of chloroform/isoamyl alcohol.
  • the aqueous phase was then precipitated with 1/10 volume of 3M sodium acetate (Cat #16-019B, BioWhittaker, Walkersville, MD) and one volume of isopropanol (Sigma, Molecular Biology Grade).
  • RNA pellet obtained after centrifugation at 10,000 X G for 30 minutes was washed one time with 70% isopropanol before being redissolved in Molecular Biology Grade water for further use. 2.
  • RT-PCR was performed with primer sets 129-933RT and 2665-
  • Figure 14 shows the results for amplification of normal colon specimen 1 (Nl, Lane 2), matched colon cancer specimen 1 (Cl, Lane 3), normal colon specimen 2 (N2, Lane 5) and colon cancer specimen 2 (C2, Lane 6) with the 129-933RT primer set.
  • Lane 1 contains molecular weight standards
  • Lane 4 is blank
  • Lane 7 contains a control with no target.
  • Figure 15 shows the same specimens amplified with the 2665-3103RT primer set on the left half of the gel.
  • Figure 15 also shows the amplification of a control sequence, ⁇ 2- microglobulin (MIC), on the right half of the gel, amplified according to the protocol and with primers specified in Corey, E., et al. ⁇ Clinical Chemistry 43:443-452, 1997).
  • the samples are (beginning from the left) as follows: Lanes 1 and 8, molecular weight markers; Lanes 2 and 9, Nl ; Lanes 3 and 10, Cl ; Lanes 4 and 11, blank; Lanes 5 and 12, N2; Lanes 6 and 13, C2; Lanes 7 and 14, no target.
  • the amplification of the MIC target demonstrates that the quantity and quality of total RNA containing the CDl lb target mRNA was comparable for both normal and cancerous tissues for both specimens tested.
  • the results of the two CDl lb primer amplifications clearly show an upregulation of CR3 in the colon cancer specimens.
  • RT-PCR was also performed with the CFI primer pair FI7us and FIG683ds described above in Example IV. FI7us and FIG683ds were each used at 0.50 nmoles per 100 ⁇ L reaction mixture. Annealing conditions were 62°C for 30 seconds and extension was at 72°C for 90 seconds, with 40 cycles. Positive results, defined to be the identification of a 472-base pair amplicon, were determined by electrophoresis as described above. When the matched normal and cancerous colon tissues from patients 1 and 2 in the CDllb study were examined, as shown in Figure 16, complement Factor I mRNA expression was clearly upregulated in the tumors.
  • the samples on the gel are as follows: Lane 1, molecular weight markers; Lane 2, Nl; Lane 3, Cl; Lane 4, N2; Lane 5, C2; Lane 6, no target.
  • the result, as shown in this figure, is unexpected and significant since Factor I has not previously been described as being expressed in colon tumors, and since Factor I with Factor H as a cofactor is capable of hydrolyzing C3b, resulting in failure of the Membrane Attack Complex (MAC) to assemble on the cell surface. This in turn would enable a colon cancer cells to evade lysis by the immune system.
  • All publications and patent applications mentioned in this specification are herein inco ⁇ orated by reference to the same extent as if each individual publication or patent application was specifically and individually inco ⁇ orated by reference.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Cell Biology (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Biomedical Technology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Zoology (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Oncology (AREA)
  • General Physics & Mathematics (AREA)
  • Pathology (AREA)
  • Toxicology (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Hospice & Palliative Care (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Abstract

L'invention concerne des procédés de criblage ou de traitement du cancer ou de troubles auto-immuns. Dans un aspect de la présente invention, lesdits procédés de criblage se basent sur la détection du composant C3 du complément ou d'une protéine apparentée au C3, ou sur la détection d'une molécule d'acide nucléique codant ledit C3, lequel est connu pour être lié à la présence du cancer. D'autres procédés de criblage se basent sur l'utilisation des régulateurs du complément, les facteurs I ou DAF, ou de récepteurs des compléments 1 ou 3. Des modes de réalisation préférés de ces procédés consistent en une détection basée sur des propriétés immunitaires, des propriétés physiques, des propriétés enzymatiques, et des combinaisons de ces propriétés, ou en une détection d'une molécule d'acide nucléique codant un antigène, sur la base d'une amplification d'acide nucléique.
EP98910227A 1997-03-06 1998-03-06 Criblage et traitement au moyen d'un regulateur du complement ou de proteines receptrices du complement Withdrawn EP0972200A1 (fr)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US3861597P 1997-03-06 1997-03-06
US81248197A 1997-03-06 1997-03-06
US38615P 1997-03-06
US812481 1997-03-06
PCT/US1998/004492 WO1998039659A1 (fr) 1997-03-06 1998-03-06 Criblage et traitement au moyen d'un regulateur du complement ou de proteines receptrices du complement

Publications (1)

Publication Number Publication Date
EP0972200A1 true EP0972200A1 (fr) 2000-01-19

Family

ID=26715383

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98910227A Withdrawn EP0972200A1 (fr) 1997-03-06 1998-03-06 Criblage et traitement au moyen d'un regulateur du complement ou de proteines receptrices du complement

Country Status (4)

Country Link
EP (1) EP0972200A1 (fr)
JP (1) JP2001515589A (fr)
AU (1) AU6451998A (fr)
WO (1) WO1998039659A1 (fr)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL141936A0 (en) * 1998-09-10 2002-03-10 Univ Virginia Antibodies to c3b(1) for delivery of diagnostic and therapeutic agents to cancer cells
US6572856B1 (en) 1998-09-10 2003-06-03 The University Of Virginia Patent Foundation Methods for the prevention and treatment of cancer using anti-C3b(i) antibodies
DE60020529T2 (de) 1999-03-01 2006-04-27 Genentech Inc., San Francisco Antikörper zur krebsbehandlung und -diagnose
JP2004524002A (ja) * 2000-06-29 2004-08-12 デルタジェン インコーポレイテッド 標的化された遺伝子破壊を含むトランスジェニックマウス
JP2018128408A (ja) * 2017-02-10 2018-08-16 東ソー株式会社 抗体凝集体形成予測方法
CN115261401B (zh) * 2022-08-25 2023-10-20 杭州博岳生物技术有限公司 一种利用酵母细胞表面展示技术开发膀胱肿瘤抗原单克隆抗体的方法

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4342566A (en) * 1980-02-22 1982-08-03 Scripps Clinic & Research Foundation Solid phase anti-C3 assay for detection of immune complexes
IL82390A0 (en) * 1986-05-02 1987-10-30 Genentech Inc Nucleic acid and methods for the synthesis of novel daf compositions
GB8726230D0 (en) * 1987-11-10 1987-12-16 Rosen H Antibodies
US5256642A (en) * 1988-04-01 1993-10-26 The Johns Hopkins University Compositions of soluble complement receptor 1 (CR1) and a thrombolytic agent, and the methods of use thereof
DE3830271A1 (de) * 1988-09-06 1990-03-15 Goetze Otto Mittel mit immunsuppressiver wirkung
US5322838A (en) * 1988-11-16 1994-06-21 Brigham & Women's Hospital Use of INHIB (the C3 β-chain) in the detection and inhibition of inflammation
EP0512733A2 (fr) * 1991-05-03 1992-11-11 Washington University Régulateur modifié du système complémentaire
JP3562834B2 (ja) * 1994-06-01 2004-09-08 孝夫 辻 便中daf分子の検査方法
NZ333949A (en) * 1994-09-08 2000-06-23 Imutran Ltd Modified human C3 proteins as therapeutic agents

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9839659A1 *

Also Published As

Publication number Publication date
JP2001515589A (ja) 2001-09-18
AU6451998A (en) 1998-09-22
WO1998039659A1 (fr) 1998-09-11

Similar Documents

Publication Publication Date Title
CA2741641C (fr) Sequence d'adn et proteine codee du cancer du sein specifique de la glande mammaire
AU2003200722C1 (en) Compositions and methods for the treatment of tumor
US20050176104A1 (en) Compositions and methods for the treatment of tumor
US6723506B2 (en) Method of identifying PAX8-PPAR gamma-nucleic acid molecules
US6221621B1 (en) Methods of screening for colorectal cancers in which a complement Factor I or related protein is associated
JPH11502404A (ja) 細胞表面発現分子および形質転換関連遺伝子に特異的なdnaプローブおよび免疫学的試薬の開発
AU776600B2 (en) Compositions and methods for the treatment of tumors
EP1185642A2 (fr) Compositions et methodes de traitement de tumeur
AU773055C (en) Compositions and methods for the treatment of tumors
AU660001B2 (en) Development of DNA probes and immunological reagents of human tumor associated antigens
AU2192800A (en) Compositions and methods for the treatment of tumor
EP0972200A1 (fr) Criblage et traitement au moyen d'un regulateur du complement ou de proteines receptrices du complement
WO1998039659A9 (fr) Criblage et traitement au moyen d'un regulateur du complement ou de proteines receptrices du complement
AU718842B2 (en) Methods and compositions for screening for or modulating a tumor associated antigen
US6150110A (en) HMGI(Y)-LAMA4* fusion oncogene, oncoprotein and methods of use
WO1999035159A1 (fr) Oncogene et oncoproteine de lymphome/leucemie, ainsi que leurs procedes d'utilisation
WO2001005836A1 (fr) Compositions polypeptidiques et methodes de traitement des tumeurs
US20060089493A1 (en) Novel gene targets and ligands that bind thereto for treatment and diagnosis of colon carcinomas
AU756400B2 (en) Compositions and methods for the treatment of tumor
JPWO2005118811A1 (ja) 癌の診断と治療において有用な新規ポリペプチド
NZ513423A (en) Compositions and methods for the treatment of tumors

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19990910

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FI FR GB GR IE IT LI LU MC NL PT SE

17Q First examination report despatched

Effective date: 20020619

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20021231