WO1997000318A1 - Facteur regulateur de la croissance des osteoplastes - Google Patents

Facteur regulateur de la croissance des osteoplastes Download PDF

Info

Publication number
WO1997000318A1
WO1997000318A1 PCT/US1996/009127 US9609127W WO9700318A1 WO 1997000318 A1 WO1997000318 A1 WO 1997000318A1 US 9609127 W US9609127 W US 9609127W WO 9700318 A1 WO9700318 A1 WO 9700318A1
Authority
WO
WIPO (PCT)
Prior art keywords
dna
osf
cells
antibody
polypeptide
Prior art date
Application number
PCT/US1996/009127
Other languages
English (en)
Inventor
G. David Roodman
Sakamuri V. Reggy
Gregory R. Mundy
Original Assignee
Osteosa 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 Osteosa Inc. filed Critical Osteosa Inc.
Priority to AU60908/96A priority Critical patent/AU6090896A/en
Publication of WO1997000318A1 publication Critical patent/WO1997000318A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/22Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against growth factors ; against growth regulators
    • 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
    • C07K14/475Growth factors; Growth regulators
    • C07K14/51Bone morphogenetic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to a novel osteoclast growth cegulatory factor, "osteoclast stimulating factor (OSF),” which stimulates the growth and/or differentiation of osteoclast cells, methods of preparing OSF, therapeutic and diagnostic uses thereof, nucleic acid sequences encoding all or part of OSF, antibodies to and antagonists of OSF, and assays for OSF, and provides therapeutics for the prevention and treatment of diseases involving bone tissue including osteoporosis, Paget's disease, and osteopetrosis.
  • OSF osteoclast stimulating factor
  • Living bone tissue is continuously being replenished by the process of resorption and deposition of calcium minerals.
  • This process described as the absorption-resorption cycle, is facilitated by means of substantially two cell types, the osteoblasts and the osteoclasts.
  • the osteoclast is a multinucleated cell and is the only cell in the body known to have the capacity to degrade (or resorb) bone.
  • This resorption activity is accomplished by the osteoclast forming pits (resorption lacunae) in bone tissue, and, in fact, osteoclast activity in cell culture is measured by their capacity to form these pits on slices of mineralized tissue such as bone or sperm whale dentine.
  • the osteoclast is derived from a hematopoietic precursor which it shares with the formed elements of the blood (Mundy & Roodman, In: Bone and Mineral Research V, Elsevier, Peck WA (ed.)., Chapter 5, pp. 209-280, 1987).
  • the precursor for the osteoclast is a mononuclear cell (cell with a single nucleus) which is found in the bone marrow and which forms the mature and unique multinucleated osteoclast after undergoing replication and differentiation by means of cell fusion.
  • the mature osteoclast is distinguished from other multinucleated cells by the presence of the enzyme tartrate-resistant acid phosphatase (TRAP) which is used as an osteoclast cell marker.
  • TRIP tartrate-resistant acid phosphatase
  • cytokines Cells found in blood and bone respond to specific protein factors excreted by other cells in response to various stimuli. These factors are referred to as cytokines, many of which have been identified by their biological characteristics and their unique amino acid sequences. Each cytokine presents a unique spectrum of characteristics utilized to distinguish each specific cytokine from others. Certain cytokines stimulate the growth and/or differentiation of specific types of cells, while other cytokines target cancerous cells for destruction.
  • Exemplary cytokines include granulocyte colony stimulating factor (G-CSF), granulocyte macrophage CSF (GM-CSF), macrophage CSF (M-CSF), interleukin-1 beta, interleukin-3, interleukin-6, interferon-gamma, tumor necrosis factor, lymphotoxin, leukemia ir ⁇ ibitory factor, and transforming growth factor-alpha.
  • G-CSF granulocyte colony stimulating factor
  • GM-CSF granulocyte macrophage CSF
  • M-CSF macrophage CSF
  • interleukin-1 beta interleukin-3
  • interleukin-6 interferon-gamma
  • tumor necrosis factor lymphotoxin
  • leukemia ir ⁇ ibitory factor transforming growth factor-alpha
  • cytokines Several growth regulatory cytokines such as CSF-M, transforming growth factor alpha, interleukin-1 and tumor necrosis factor have been shown to stimulate marrow mononuclear cell proliferation. Although cytokines such as interieukin-1 (IL-1), tumor necrosis factor (TNF) and interleukin-6 (IL-6) may influence osteoclast formation and differentiation (Mundy, Trends in Endocrinol. & Metab.
  • osteoclastpoietic factor A biologically active polypeptide, osteoclastpoietic factor (OPF), was isolated from the conditioned media from cultured tumor cells (MH-85 cells). Among the biological activities of OPF was the ability to regulate the growth and/or differentiation of osteoclast cells (WO 93/01827 which is incorporated herein by reference).
  • the present invention relates to the isolation of an osteoclast stimulatory factor from an expression library derived from human marrow-derived osteoclastic cells and to the characterization of a polypeptide having biological activity including regulatory activity associated with the development of multinucleated bone cells.
  • the present invention relates to a biologically active polypeptide comprising the amino acid sequence shown in Figure 1 (SEQ ID NO: 2), or a biologically active sequence analogue thereof.
  • a biologically active polypeptide comprising the amino acid sequence shown in Figure 1 (SEQ ID NO: 2), or a biologically active sequence analogue thereof.
  • the biological properties of the polypeptide of the present invention is the capability to regulate the growth and/or differentiation of osteoclast cells.
  • the invention further provides nucleotide sequences encoding said biologically active polypeptide.
  • a preferred embodiment is the coding region (nucleotides 150-794) shown on Figure 1 (SEQ ID NO: 1).
  • the invention provides monoclonal and polyclonal antibodies capable of specifically binding to the amino acid sequence of Figure 1 , or to a biologically active sequence analogue thereof, as well as uses of these monoclonal and polyclonal antibodies therapeutically and diagnostically.
  • the antibodies of the present invention are useful for affinity purification of the naturally occurring polypeptide as well as active fragments thereof, in assays for detecting the present polypeptide and for treating pathological conditions resulting from overproduction thereof.
  • the assays provide a method for the clinical diagnosis and assessment of those diseases in which there is excess production of the naturally occurring polypeptide, and for monitoring treatment efficacy.
  • compositions such as diagnostic and pharmaceutical compositions, containing the polypeptide of the present invention and methods of using these in treatment and diagnosis.
  • the present invention provides a method for the treatment of bone diseases characterized by abnormal osteoclast activity such as osteopetrosis, comprising administration of the present polypeptide to individuals in need of such treatment.
  • Antagonists such as the present antibodies to the present polypeptides, are useful for inhibiting bone resorption in a number of disease states where bone resorption is enhanced such as, but not limited to osteoporosis, Paget's disease, malignant diseases which affect the skeleton such as myeloma and breast cancer, and chronic inflammatory diseases which cause localized bone loss such as rheumatoid arthritis and periodontal disease. Treatment of these diseases may be accomplished by administration of antagonists such as neutralizing antibodies to this and related polypeptides to individuals in need of such treatment.
  • Figure 1 shows the cDNA sequence (SEQ ID NO: 1) and deduced amino acid sequence (SEQ ID NO: 2) of OSF.
  • Figure 2 shows a primary screening of the human osteoclast-like cell cDNA expression library. Conditioned media from 293 cells transiently transfected with pools (P1-P16) from the pcDNAI expression library were tested for the ability to enhance osteoclast-like MNC formation in human marrow cultures. Results shown are the mean ⁇ S.E. for five replicate samples and were compared by the Student's t test. Results were considered significantly different for ⁇ 0.05 ( * ) or p ⁇ 0.01 ( ** ).
  • Figure 3 shows the ability of conditioned media from 5F cDNA- transfected 293 cells to enhance formation of MNCs in human bone marrow cultures. Results shown are the mean ⁇ S.E. for five replicate samples and were compared by the Student's t test. Results were considered significantly different for p ⁇ 0.05 ( * ).
  • Figure 4 shows the effect of recombinant OSF on the formation of MNCs in human bone marrow cultures.
  • Figure 5 shows the effect of 5F conditioned media on formation of MNCs in murine marrow cultures.
  • Figure 6 shows the effect of 5F conditioned media on mouse bone marrow pit formation.
  • Figure 7 shows the effect of recombinant OSF on fetal rat bone resorption.
  • the present invention relates to a biologically active polypeptide comprising the amino acid sequence shown in Figure 1 (SEQ ID NO: 2), or a biologically active sequence analogue thereof.
  • the amino acid sequence of this polypeptide is distinct from the sequences of other proteins which have been shown to promote osteoclast formation.
  • Bio activity means one or more functions, effects of , .activities performed or caused by a molecule in a biological context (that is, in an organism or in an in vitro facsimile).
  • a characteristic biological activity of OSF is the ability to stimulate the growth and/or differentiation of osteoclast cells.
  • biologically active polypeptide means the naturally occurring polypeptide per se. as well as biologically active analogues thereof, synthetic produced polypeptides, natural and pharmaceutically acceptable salts and pharmaceutically acceptable derivatives.
  • the present invention encompasses OSF and biologically active fragments thereof, as well as biologically active sequence analogues thereof. Different alleles of OSF may exist in nature.
  • compositions of the structural gene coding for proteins of identical biological function may be characterized by differences in the nucleotide sequence of the structural gene coding for proteins of identical biological function.
  • biologically active sequence analogue includes analogues having single or multiple amino acid substitutions, deletions, additions, or replacements. All such allelic variations, modifications, and analogues resulting in derivatives of OSF which retain one or more of the biologically active properties of native OSF are included within the scope of this invention.
  • the present invention provides polypeptides in substantially homogeneous form.
  • substantially homogeneous means that the polypeptide is essentially free of other proteins normally associated with the polypeptide in its natural state.
  • substantially homogeneous is not meant to exclude artificial or synthetic mixtures of the polypeptide with other compounds.
  • a "substantially homogeneous" nucleic acid is a nucleic acid essentially free of other nucleic acids normally associated with the nucleic acid in its natural state.
  • the term “substantially homogeneous” is not meant to exclude artificial or synthetic mixtures of the nucleic acid with other compounds.
  • the present invention also provides methods for purifying OSF.
  • the present invention provides a method for purifying recombinant OSF, which is produced by transforming E. coli with a vector comprising the OSF cDNA.
  • the present invention provides a method for purifying OSF comprising contacting a medium containing OSF mixed with other proteins with an antibody which binds to at least one epitope of the OSF molecule, removing the antibody-OSF complex, releasing the OSF from the antibody and separating the OSF from the antibody.
  • the antibody is bound to a solid support.
  • the choice of solid support and methods for binding the antibody to the solid support are well known to those skilled in the art.
  • the present invention also provides nucleic acids encoding the biologically active polypeptide of the present invention.
  • a preferred embodiment is the coding region (nucleotides 150-794) shown on Figure 1 (SEQ ID NO: 1).
  • the biologically active polypeptides of the present invention may be prepared utilizing recombinant technology.
  • a recombinant DNA molecule coding for any of the polypeptides of the present invention can be used to transform a host using techniques known to those of ordinary skill in the art.
  • a "plasmid” is a non chromosomal double-stranded DNA sequence comprising an intact "replicon” such that the plasmid is replicated in a host cell.
  • the characteristics of that cell may be changed (or transformed) as a result of the DNA of the plasmid.
  • a plasmid carrying the gene for tetracycline resistance (TetR) transforms a cell previously sensitive to tetracycline into one which is resistant to it.
  • a cell transformed by a plasmid is called a "transformant.”
  • a “vector” is a plasmid, phage DNA or other DNA sequence which is able to replicate in a host cell, typically characterized by one or a small number of endonuclease recognition sites at which such DNA sequences may be cut in a determinable fashion for the insertion of heterologous DNA without attendant loss of an essential biological function of the DNA, e.g., replication, production of coat proteins or loss of expression control regions such as promoters or binding sites, and which may contain a selectable gene marker suitable for use in the identification of host cells transformed therewith, e.g., tetracycline resistance or ampicillin resistance.
  • An "expression vector” is a vector which is capable of expressing a peptide encoded by heterologous DNA sequences contained in the vector, such as the cDNA encoding OSF.
  • the heterologous DNA sequences are operably linked to regulatory sequences which are capable of regulating expression of the peptide.
  • An expression vector typically contains an origin of replication, promoter(s), terminator(s), a ribosome binding site, as well as specific genes which are capable of providing phenotypic selection in transformed cells. These expression vectors must be replicable in the host organisms or systems either as episomes, bacteriophage, or as an integral part of the chromosomal DNA.
  • the transformed hosts can be fermented and cultured according to means known in the art to achieve optimal cell growth.
  • Prokaryotic cells are preferred for the cloning of DNA sequences and in the construction of vectors.
  • Cell lines derived from multicellular organisms may also be used as hosts. Examples of such hosts are the VERO, HeLa, mouse C127, Chinese hamster ovary (CHO), WI38, BHK, COS-7, and MDCK cell lines.
  • Expression vectors for such cells ordinarily include an origin of replication, a promoter Iocated in front of the gene to be expressed, RNA splice sites (if necessary), and transeriptional termination sequences.
  • control functions on the expression vectors are often provided by viral material.
  • promoters are derived from polyoma, Adenovirus 2, and most frequently, Simian Virus 40 (SV40).
  • Eukaryotic promoters such as the promoter of the murine metallothionein gene [Paulakis and Hamer, Proc. Natl. Acad. Sci. 80:397-401 (1983)], may also be used.
  • eukaryotic enhancer sequences can be obtained from a variety of animal cells or oncogenic retroviruses such as the mouse sarcoma virus.
  • An origin of replication may be provided either by construction of the vector to include an exogenous origin, such as that provided by SV40 or other viral sources, or may be provided by the host cell chromosomal replication mechanism. If the vector is integrated into the host cell chromosome, the latter is often sufficient.
  • the present invention includes any host modified according to the methods described, or modified by any other methods commonly known to those of ordinary skill in the art, which yields a prokaryote or eukaryote expressing the gene for OSF.
  • Host cells used to prepare the polypeptides of the present invention can be of a variety of chemical compositions.
  • the polypeptide may be.produced having methionine as its first amino acid. This methionine is present by virtue of the ATG start codon naturally existing at the origin of the structural gene or by being engineered before a segment of the structural gene.
  • the protein may also be intracellularly or extracellularly cleaved, giving rise to the amino acid which is found naturally at the amino terminus of the polypeptide.
  • Recombinant host cells are cells which have been transformed with vectors constructed using recombinant DNA techniques.
  • the polypeptides of the present invention or fragments thereof produced by such cells are referred to as "recombinant polypeptides of the present invention.”
  • the present invention also provides probes and methods to identify cells containing or lacking these sequences, and means to administer these sequences to cells. This will enable the establishment of systems in which the recombinant protein is produced after transfection of an expression vector into appropriate host cells. Additionally, the present invention provides a means to inhibit the expression of the novel sequences by providing an antisense RNA sequence which, when administered to a cell, or when the DNA encoding said antisense RNA is administered to a cell, said DNA sequence will produce an antisense RNA which can bind to and therefore block the expression of the RNA encoding the novel polypeptides of the present invention.
  • antibodies against any of the proteins of the present invention can be utilized to block the binding of ligands to the polypeptides and to target drugs or other agents (such as labels) to the cells expressing these polypeptides.
  • Monoclonal antibodies of the present invention may be prepared using the method of Mishell, B. B., et aL, Selected Methods ln Cellular Immunology. (W.H. Freeman, ed.) San Francisco (1980).
  • the biologically active polypeptide of the present invention is used as the antigen for the production of these antibodies.
  • OSF peptide is used to immunize spleen cells of Balb/C mice.
  • the immunized spleen cells are fused with FO myeloma cells.
  • Fused cells containing spleen and myeloma cell characteristics are isolated by growth in HAT medium, a medium which kills both parental cells, but allows the fused products to survive and grow.
  • the anti-OSF antibodies are useful in the treatment of disease states caused by increased levels of OSF in the individual.
  • Neutralizing antibodies can inhibit the activity of the excessively produced OSF in these individuals.
  • treatments include administration of anti-OSF monoclonal astibodies to individuals suffering from osteoporosis, malignant diseases which affect the skeleton such as myeloma and breast cancer, and chronic inflammatory diseases which cause localized bone loss such as rheumatoid arthritis and periodontal disease.
  • the anti-OSF antibodies are also useful in assays for detecting or quantitating levels of OSF. These assays provide a clinical diagnosis and assessment of those diseases in which excess production of these factors occurs, and a method for monitoring treatment efficacy. Synthetic antagonists to OSF have the same beneficial therapeutic effect as neutralizing antibodies in those diseases characterized by the overproduction of OSF.
  • the term "individual” is meant to include any animal, preferably a mammal, and most preferably a rodent, cat, dog, cow or human.
  • the techniques for detectably labeling the homogeneous OSF and the monoclonal antibodies thereto of the present invention with a radiolabel, an enzyme label, or a fluorescent label are well known to those of skill in the art. Reference can be made to Chard, An Introduction To Radioimmunoassay And Related Techniques. North-Holland Publishing Co., Amsterdam-NY-Oxford (1978). The Enzvme-Linked Immunoadsorbent Assav (ELISA) by Voller.
  • the purified OSF is labeled with 125 l using the Bolton/Hunter reagent which involves succinylation of the free N-terminals and lysine.
  • DNA probes may also be labeled with a detectable label. Commonly used detectable labels are radioactive labels including, but not limited to, 32 P, 1 C, 1 5 l, 3 H and 35 S.
  • Biotin labeled nucleotides can be incorporated into DNA or RNA by nick translation, enzymatic, or chemical means. The biotinylated probes are detected after hybridization using avidin/streptavidin, fluorescent, enzymatic or colloidal gold conjugates. Nucleic acids may also be labeled with other fluorescent compounds, with immunodetectable fluorescent derivatives or with biotin analogues. Nucleic acids may also be labeled by means of attaching a protein. Nucleic acids cross-linked to radioactive or fluorescent histone HI, enzymes (alkaline phosphatase and peroxidases), or single-stranded binding (ssB) protein may also be used.
  • Administration of the compounds useful in the method of present invention may be by parenteral, intravenous, intramuscular, subcutaneous, rectal or any other suitable means.
  • the dosage administered may be dependent upon the age, weight, kind of concurrent treatment, if any, and nature of the pathological state being treated.
  • the effective compound useful in the method of the present invention may be employed in such forms as capsules, liquid solutions, suspensions or elixirs for oral administration, or sterile liquid forms such as solutions or suspensions.
  • Any inert carrier is preferably used, such as saline, or phosphate-buffered saline, or any such carrier in which the compounds used in the method of the present invention have suitable solubility properties for use in the method of the present invention.
  • salts refers to both salts of carboxy groups of the polypeptide or protein chain and to acid addition salts of amino groups of the polypeptide chain.
  • Salts of the carboxy group may be formed with either in- organic or organic bases by means known in the art per se.
  • Inorganic salts include, for example, sodium, calcium, ammonium, ferric or zinc salts, and the like.
  • Salts with organic bases include those formed, for example, with amines such as triethanolamine, arginine, lysine, piperidine, caffeine, procaine and the like.
  • Acid addition salts include, for example, salts with mineral acids such as, for example, hydrochloric acid or sulfuric acid, and salts with organic acids such as, for example, acetic acid or oxalic acid.
  • Both the salts and the derivatives encompassed by the invention include those which are therapeutically or diagnostically acceptable, i.e., those which do not destroy the biologic, immunogenic, or binding activity of OSF depending on the functional activity desired to be utilized.
  • Human long term marrow cultures were prepared as described above, and after 3 weeks, the cells were washed twice with phosphate-buffered saline and treated with trypsin-EDTA for 10 min to remove the lightly adherent mononuclear cells. The remaining adherent cells were then washed with phosphate-buffered saline vigorously four times, and fresh medium was added to each culture. The cells were cultured overnight and lysed in 0.2 ml of Triton X-100 (0.05%, v/v). Aliquots of the lysate (40 ⁇ l) were incubated with 160 ⁇ l of Triton X-100 (0.05%, v/v). Aliquots of the lysate (40 ⁇ l) were incubated with 160 ⁇ l of Triton X-100 (0.05%, v/v). Aliquots of the lysate (40 ⁇ l) were incubated with 160 ⁇ l of Triton X-100 (0.05%, v
  • Human bone marrow-derived nonadherent mononuclear cells from 20 separate normal marrow donors were cultured at 10 8 cells/ml in the presence of 10" 8 M 1 ,25-(OH)2D3 in T-75 culture flasks for 3 weeks as desc ⁇ ed previously (Takahashi et al., J. Clin. Invest. 83:543-550, 1989).
  • the cells were harvested from the plastic culture flasks by treatment of the cultures with chymopapain (300 units/ml) for 30 min at 37°C, and the released cells were pelleted by centrifugation at 1500 rpm for 5 min.
  • the cells were resuspended in serum-free ⁇ -minimum essential medium ( ⁇ -MEM), and the osteoclast-like multinucleated cells (MNC) were purified by immune panning with the 23c6 monoclonal antibody (Ohsaki et al., Endocrinology 131 : 2229-2234, 1992).
  • the 23c6 monoclonal antibody was generously provided by Dr. Michael Horton (St. Bartholomew's Hospital, London, UK).
  • Dr. Michael Horton St. Bartholomew's Hospital, London, UK.
  • We have previously shown that MNC that react with the 23c6 antibody are the osteoclast-like cells in these cultures (Kurihara et al., Endocrinology 126: 2733-2741 , 1990).
  • RNA was isolated from the 23c6 + cell fraction using oiigo(dT)-cellulose spin columns.
  • a cDNA library was made in the ⁇ gtl 1 vector using a cDNA synthesis kit (Pharmacia) following the manufacturer's protocol.
  • the cDNA inserts were excised from the ⁇ gtl 1 vectors by digestion at 37°C for 2 h with 30 units of EcoRI.
  • the cDNAs were then size-fractionated, and the 0.5-8 kilobase cDNAs were cloned into the pcDNAI mammalian expression vector (Invitrogen, San Diego, CA).
  • the pcDNAI expression vector contains enhancer-promoter sequences from the immediate early gene of the human cytomegalovirus (CMV), which can be transactivated by the adenovirus EIA protein. This results in high levels of gene expression when genes are transiently transfected into mammalian cells such as human 293 cells.
  • CMV human cytomegalovirus
  • the pcDNAI vector also contains a Col E1 origin for growth in E. coli, and a gene encoding the tRNA suppressor F gene (sup F).
  • the pcDNAI vector is efficiently replicated when transformed into a strain of E. coli such as MC1061/P3, which harbors the plasmid P3.
  • P3 is a low-copy 60 kb episome which encodes a kanamycin resistance gene as well as amber mutants of the tetracycline and ampicillin resistance genes.
  • E. coli carrying the P3 plasmid such as MC1061/P3 are transformed with su ⁇ F vectors such as pcDNAI, they are rendered both tetracycline and ampicillin resistant by suppression of the amber mutations.
  • the cDNAs in pcDNAI were transformed into MCIO6I/P3 Escherichia coli, and transformants were selected with tetracycline (10 ⁇ g/ml) and ampicillin (40 ⁇ g/ml).
  • the resulting pcDNAI expression library contained 4x10 5 cDNA clones. This library was divided into 200 pools containing 2000 clones each. Replicate plate lifts of each pool were performed using nitrocellulose filters, and the plates were stored at -20°C as descried (Hanahan and Meselson, Gene 10: 63-67, 1980).
  • the cDNA expression library was screened by testing the effects of conditioned media from transfected mammalian 293 cells for the capacity to enhance osteoclast-like MNC formation in human marrow cultures.
  • Cesium chloride gradient purified plasmid DNA (10 ⁇ g) from each of the 200 expression library pools was transiently transfected into 293 cells grown in individual 35-mm wells (4x10 5 cells/well) using a calcium phosphate method (using a kit from Stratagene according to the manufacturer's protocol). Twelve hours after the start of the DNA transfection process, the cells were fed with 1.5 ml of serum-free Dulbecco's modified Eagle's medium. Conditioned media from each pool were collected after 48 h and tested at different concentrations (0.1-10%, v/v) for the capacity to enhance MNC formation in human marrow cultures.
  • Conditioned media were added to normal human bone marrow long term culture in the presence or absence of 10" 8 M 1 ,25-(OH)2D3.
  • the cells were stained for 23c6 reactivity, or the MNC were lysed for determining tartrate-resistant acid phosphatase (TRAP) activity as described above. Pools that increased TRAP activity were retested to confirm that they enhanced osteoclast-like MNC formation by counting the number of 23c6 antibody-reactive MNC formed in the marrow cultures. The number of 23c6 + MNC were counted and compared with cultures not containing conditioned medium or with cultures treated with conditioned medium from 293 cells that had not been transfected. A typical screening experiment is shown in Figure 2.
  • the remaining six positive cDNA pools were separated into 12 subpools each, containing 100-200 clones/subpool.
  • the individual pools were transfected into 293 cells, and the conditioned media were tested for their effects on MNC formation.
  • the initial assay was for TRAP activity in cell lysates from the human bone marrow cultures. Because there are many TRAP+ mononuclear cells in human bone marrow cultures, the cultures were first treated with trypsin-EDTA to remove the majority of the mononuclear cells. Greater than 90% of the MNC were not removed, and few ( ⁇ 5%) of the mononuclear cells remained.
  • TRAP activity in the cell lysate was determined.
  • TRAP activity was significantly increased in cultures treated with conditioned media from six positive pools compared to cultures treated with conditioned media from nontransfected 293 cells or cultures not treated with conditioned media.
  • One of the positive pools (no. 12) was further subdivided into several subpools and screened by the TRAP assay. Each positive subpool, containing 100-200 clones, was separated into 96 or 192 subpools that had one clone per pool. Conditioned medium from each of these clones was treated for its capacity to increase MNC formation.
  • the first positive clone identified was 5F.
  • the effects of conditioned media from 5F cDNA-transfected 293 cells was tested on osteoclast-like MNC formation in mouse (see Example 5, below) and in human bone marrow cultures.
  • the 5F conditioned medium increased MNC formation in human bone marrow cultures, as shown in Figure 3.
  • An increase in MNC formation was seen at concentrations as low as 0.05% (v/v), in the absence or presence of 1 ,25-(OH) 2 D 3
  • the 5F cDNA was sequenced using a dideoxyoligonucleotide chain termination method (using a kit from USB according to the manufacturer's directions).
  • the 5F cDNA encoded a 23kd peptide.
  • the nucleotide sequence and deduced amino acid sequence are shown in Figure 1. The sequence was compared to sequences in GenBank and was found to be unique, with no other sequence having complete homology.
  • the 5F cDNA was introduced into a pET vector (Novagen Inc., Madison, Wl), and the 5F clone was expressed in the BL21 (DE3)pLysS strain of E. coli.
  • the pET vector is designed for expression of target cDNA inserts as stable fusion proteins under the control of the bacteriophage T7 promoter.
  • the E. coli host BL21 (DE3)pLysS contains a chromosomal copy of the T7 RNA polymerase gene under LAC UV5 control.
  • Expression vectors containing a target cDNA in pET are transformed into BL21 (DE3)pLysS, and cDNA expression is then induced by the addition of IPTG.
  • the pET vector has the advantage of placing a six histidine tag sequence at the N-terminal end of the recombinant protein, which allows easy purification of the recombinant protein. The histidine tag can then be removed.
  • the 5F cDNA was cloned into the pET vector, resulting in the expression vector 5FpET2.
  • the 5FpET2 expression vector was transformed into the BL21 (DE3)pLysS strain of E. coli using a calcium phosphate method. Expression was induced by IPTG.
  • the recombinant OSF protein was affinity purified using a metal chelation resin containing immobilized divalent Nickel cations (His. Bind Resin, Novagen Inc.), which efficiently binds the histidine tag sequence.
  • Bone marrow from CB57 black mouse tibiae that had been aseptically removed was obtained by flushing the tibiae with 1 ml of ⁇ -MEM using a tuberculin syringe fitted with a 25-gauge needle.
  • the bone marrow-derived cells were washed twice and resuspended in ⁇ -MEM, 20% fetal calf serum, and the cells depleted of cells adherent to plastic by incubating the marrow cell suspension in sterile 10-cm tissue culture dishes for 2 h.
  • the nonadherent marrow cells were collected and cultured for 6 days in 24-well plates at a density of 1.0x10 6 cells/well in ⁇ -MEM, 10% fetal calf serum supplemented with 1 ,25-(OH) 2 D3 (10" 8 or 10 "9 M) as described by Takahashi et al.,
  • the cultures were fixed with 4.5 mM citric acid, 2.25 mM sodium citrate, 3 mM sodium chloride, 3% formaldehyde, and acetone and were washed twice in distilled water. The cultures were then stained for TRAP using an acid phosphatase staining kit (Sigma) and counterstained with methyl green. The TRAP-positive multinucleated cells containing three or more nuclei were then scored. In mouse bone marrow cultures, the 5F conditioned media increased
  • TRAP+ MNC formation two-fold at concentrations of 0.1-5% (v/v) in the presence of 10" 8 or 10" 9 M of vitamin D3 ( Figure 5).
  • the 5F conditioned medium also enhanced TRAP+ MNC formation in mouse bone marrow cultures in the absence of 1 ,25-(OH)2D3.
  • Selected murine marrow cultures were overlaid with dentine at the start of the cultures. After 6 days, the dentine slices were removed and stained for TRAP. The number of TRAP+ MNC on each dentine slice was counted manually, and the dentine slices were washed in distilled water. The area of the slice resorbed was determined in a blinded fashion with an inverted microscope using bright field reflected microscopy at 50x magnification with a Bioquant image analysis system (R and M Biometrics, Arlington, TN). The area resorbed was determined with Java image analysis software (Jandel Scientific, Corte Madona, CA).
  • Timed-pregnant rats were injected with 250 ⁇ Ci of 45 CaCl2 at day 18 of gestation, and 1 day later the rats were killed by cervical dislocation, and the embryos were removed.
  • the explanted radii and ulnae were cultured on circles of membrane filter, mixed ester, 0.45 ⁇ m (Whatman, Hillsboro, OR) on stainless steel grids in 0.5 ml of chemically defined medium (Sigma) supplemented with 1 mg/ml bovine serum albumin (Sigma) and penicillin-streptomycin (50 units/ml and 50 mg/ml), in a controlled atmosphere of 5% CO2 in air at 37°C, as modified from Raisz and Niemann (Endocrinology
  • RT-PCR analysis of a variety of tissues demonstrated the expression of OSF in peripheral blood mononuclear cells and highly purified osteoclasts derived from human giant cell tumors of bone and pagetic bone.
  • Monoclonal antibodies (MAb) to OPF are made using the in vitro immunization technique of Van Ness et al., (Nature 301 : 425-427, 1983).
  • Spleen cells of 8 to 12 week-old female Balb/C mice are immunized with 100 ng recombinant OSF in the presence of 100 ⁇ g N-acetylmuramyl-L-alanyl-D- isoglutamine (MDP, Sigma), 125 ⁇ g lipopolysaccharide (LPS, Difco) and 500 ⁇ l culture supematants of concanavalin A (50 ⁇ g/ml)-stimulated spleen cells (5 x 10 6 /ml) in 5 ml IMDM supplemented with 20% FBS in 6-well plates for 4 days.
  • MDP N-acetylmuramyl-L-alanyl-D- isoglutamine
  • LPS lipopolysaccharide
  • the immunized spleen cells are then hybridized with mouse myeloma FO cells (kindly provided by Dr. Eguchi, Kaneka, Japan) at a ratio of 2:1 in the presence of 50% polyethylene glycol (1500, Boehringer-Manheim).
  • the cells are suspended in 120 ml IMDM supplemented with 10% FBS and 0.5 ml cell suspensions are inoculated onto each well in 48-well plates.
  • 5 x 10 5 /well thymocytes in 0.5 ml HAT medium are plated onto each well as a feeder layer.
  • the plates are cultured for 14 days in HAT medium (Flow) and then for 7 to 10 days in HT medium (Flow).
  • the cells are fed fresh medium every 2 days.
  • the culture supematants harvested from the wells in which growing hybridoma cells cover 50% of the surface area are screened for their cross reactivity with OSF by enzyme-linked immunosorbent assay (ELISA).
  • ELISA enzyme-linked immunosorbent
  • Rabbits male, 6 to 8 week-old are subcutaneously injected (10 sites/animal, 100 ⁇ l/site) with 1 ⁇ g recombinant OSF in complete Freund's adjuvant.
  • animals are boosted intramuscularly with 1 ⁇ g recombinant OSF in incomplete Freund's adjuvant. The animals are boosted three times.
  • blood is drawn from an ear vein and tested for its reactivity against OSF by Western blot or by ELISA.
  • GCT GGA AGC ACT GCC TTA TAC TGG GCT TGC CAC GGG GGC CAC AAA GAT 509 Ala Gly Ser Thr Ala Leu Tyr Trp Ala Cys His Gly Gly His Lys Asp 105 110 115 120

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Immunology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

La présente invention concerne un nouveau facteur régulateur de la croissance des ostéoplastes, 'facteur de stimulation des ostéoplastes' (OSF), qui stimule la croissance et/ou la différenciation des ostéoplastes, ainsi que des procédés de préparation de l'OSF, des utilisations thérapeutiques et diagnostiques de celui-ci, des séquences nucléotidiques codant complètement ou partiellement l'OSF, des anticorps dirigés contre l'OSF et des antagonistes de celui-ci, et des dosages permettant la détection de l'OSF. L'invention concerne également des agents permettant la prévention et le traitement de maladies touchant le tissu osseux, y compris l'ostéoporose, la maladie osseuse de Paget et la maladie d'Albers-Schönberg.
PCT/US1996/009127 1995-06-07 1996-06-04 Facteur regulateur de la croissance des osteoplastes WO1997000318A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU60908/96A AU6090896A (en) 1995-06-07 1996-06-04 Osteoclast growth regulatory factor

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US47457895A 1995-06-07 1995-06-07
US08/474,578 1995-06-07

Publications (1)

Publication Number Publication Date
WO1997000318A1 true WO1997000318A1 (fr) 1997-01-03

Family

ID=23884149

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1996/009127 WO1997000318A1 (fr) 1995-06-07 1996-06-04 Facteur regulateur de la croissance des osteoplastes

Country Status (2)

Country Link
AU (1) AU6090896A (fr)
WO (1) WO1997000318A1 (fr)

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0974671A1 (fr) * 1997-09-24 2000-01-26 Snow Brand Milk Products, Co., Ltd. Procede permettant de diagnostiquer un dysfonctionnement du metabolisme osseux
US6855808B2 (en) 1995-02-20 2005-02-15 Sankyo Co., Ltd. Proteins and methods for producing the proteins
US6919434B1 (en) 1995-02-20 2005-07-19 Sankyo Co., Ltd. Monoclonal antibodies that bind OCIF
US7192718B2 (en) 1997-04-15 2007-03-20 Sankyo Co. Ltd Methods for identifying a substance which specifically binds to an osteoclastogenesis inhibitory factor-binding molecule and modulates the biological activity thereof
US7411050B2 (en) 1996-12-23 2008-08-12 Immunex Corporation Monoclonal blocking antibody to human RANKL
US7718776B2 (en) 2002-04-05 2010-05-18 Amgen Inc. Human anti-OPGL neutralizing antibodies as selective OPGL pathway inhibitors
US7807795B2 (en) 1997-04-16 2010-10-05 Amgen Inc. Antibodies to osteoprotegerin binding proteins
US7923008B2 (en) 1997-04-16 2011-04-12 Amgen Inc. Methods for decreasing osteoclast formation or bone resorption using an antibody to osteoprotegerin binding protein

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5169837A (en) * 1991-03-28 1992-12-08 Allelix Biopharmaceuticals Inc. Isolated osteogenic factor

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5169837A (en) * 1991-03-28 1992-12-08 Allelix Biopharmaceuticals Inc. Isolated osteogenic factor

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
JOURNAL OF CLINICAL INVESTIGATION, Volume 67, issued May 1981, JOSSE et al., "Observations on the Mechanism of Bone Resorption Induced by Multiple Myeloma Marrow Culture Fluids and Partially Purified Osteoclast-Activating Factor", pages 1472-1481. *
PROC. NATL. ACAD. SCI. U.S.A., Volume 88, issued October 1991, LEE et al., "Isolation of a Murine Osteoclast Colony-Stimulating Factor", pages 8500-8504. *
THE JOURNAL OF BIOLOGICAL CHEMISTRY, Volume 269, Number 46, issued 18 November 1994, TAKAHASHI et al., "Cloning and Identification of Annexin II as an Autocrine/Paracrine Factor that Increases Osteoclast Formation and Bone Resorption", pages 28696-28701. *
TRENDS IN ENDOCRINOLOGY AND METABOLISM, issued July/August 1990, G.R. MUNDY, "Immune System and Bone Remodeling", pages 307-311. *

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7125686B1 (en) 1995-02-20 2006-10-24 Sankyo Co., Ltd. Proteins and methods for producing the proteins
US7468268B2 (en) 1995-02-20 2008-12-23 Daiichi Sankyo Co., Ltd. Nucleic acid molecules encoding osteoclastogenesis inhibitory factor proteins
US7276344B2 (en) 1995-02-20 2007-10-02 Sankyo Co., Ltd. Methods for using the osteoclastogenesis inhibitory factor (OCIF) protein
US6855808B2 (en) 1995-02-20 2005-02-15 Sankyo Co., Ltd. Proteins and methods for producing the proteins
US6919434B1 (en) 1995-02-20 2005-07-19 Sankyo Co., Ltd. Monoclonal antibodies that bind OCIF
US7205397B2 (en) 1995-02-20 2007-04-17 Sankyo, Co., Ltd. Proteins and methods for producing the proteins
US7744886B2 (en) 1996-12-23 2010-06-29 Immunex Corporation Methods for interfering with rank signaling
US8715683B2 (en) 1996-12-23 2014-05-06 Immunex Corporation RANK ligand polypeptides
US8377690B2 (en) 1996-12-23 2013-02-19 Immunex Corporation Cells and methods for producing blocking antibodies to human RANKL
US7411050B2 (en) 1996-12-23 2008-08-12 Immunex Corporation Monoclonal blocking antibody to human RANKL
US7932375B2 (en) 1996-12-23 2011-04-26 Immunex Corporation Kits for detecting rank nucleic acids
US7449185B2 (en) 1997-04-15 2008-11-11 Daiichi Sankyo Company, Limited Antibodies to OCIF-binding molecules
US7527790B2 (en) 1997-04-15 2009-05-05 Daiichi Sankyo Company, Limited Methods of reducing bone absorption comprising administering an antibody that binds OBM
US7192718B2 (en) 1997-04-15 2007-03-20 Sankyo Co. Ltd Methods for identifying a substance which specifically binds to an osteoclastogenesis inhibitory factor-binding molecule and modulates the biological activity thereof
US7807795B2 (en) 1997-04-16 2010-10-05 Amgen Inc. Antibodies to osteoprotegerin binding proteins
US7923008B2 (en) 1997-04-16 2011-04-12 Amgen Inc. Methods for decreasing osteoclast formation or bone resorption using an antibody to osteoprotegerin binding protein
EP0974671A1 (fr) * 1997-09-24 2000-01-26 Snow Brand Milk Products, Co., Ltd. Procede permettant de diagnostiquer un dysfonctionnement du metabolisme osseux
US6693175B2 (en) 1997-09-24 2004-02-17 Sankyo Co., Ltd. Method for diagnosing bone dysbolism
EP0974671A4 (fr) * 1997-09-24 2004-10-22 Sankyo Co Procede permettant de diagnostiquer un dysfonctionnement du metabolisme osseux
US6998242B2 (en) 1997-09-24 2006-02-14 Sankyo Co., Ltd. Method of diagnosing metabolic bone diseases
US7718776B2 (en) 2002-04-05 2010-05-18 Amgen Inc. Human anti-OPGL neutralizing antibodies as selective OPGL pathway inhibitors
US8367063B2 (en) 2002-04-05 2013-02-05 Amgen, Inc. Human anti-OPGL neutralizing antibodies as selective OPGL pathway inhibitors
US8455629B2 (en) 2002-04-05 2013-06-04 Amgen Inc. Human anti-OPGL neutralizing antibodies as selective OPGL pathway inhibitors

Also Published As

Publication number Publication date
AU6090896A (en) 1997-01-15

Similar Documents

Publication Publication Date Title
KR100554186B1 (ko) 악성종양의예방또는치료용의HER-2/neu단백질의세포내도메인
KR100467998B1 (ko) Il-17수용체
US5608035A (en) Peptides and compounds that bind to the IL-1 receptor
US5880096A (en) Peptides and compounds that bind to the IL-1 receptor
US5599708A (en) Osteoclast growth regulating factors and antibodies
CZ302262B6 (cs) Izolovaná nukleová kyselina, polypeptid kódovaný touto nukleovou kyselinou, expresní vektor obsahující tuto nukleovou kyselinu, hostitelská bunka transfekovaná tímto expresním vektorem, izolovaný protein vázající osteoprotegerin, protilátka vázající
EP0567575B1 (fr) Poteine induite par la cytokine, adn tsg-6 codant pour cette proteine et ses utilisations
WO1997000317A1 (fr) Facteur regulateur de la croissance d'osteoclastes
US6040426A (en) Human Th2 specific protein
AU4027799A (en) Osteoporosis treatment
US5408041A (en) Process of purifying antler-derived bone growth factors
WO1997000318A1 (fr) Facteur regulateur de la croissance des osteoplastes
WO1997030157A1 (fr) Nouveau gene de reponse a la differenciation terminale myeloide d'origine humaine
JPH10500850A (ja) インターロイキン−6 スプライス変異体
WO1995033499A1 (fr) ADN ISOLES CODANT LE PROTO-ONCOGENE C-$i(MER)
EP0860447B1 (fr) Facteur de croissance des cellules souches hematopoietiques (scgf)
JP2003525634A (ja) Fctrxと命名されたタンパク質およびこれをコードする核酸
KR20010043088A (ko) 신규인 폴리펩티드, 그 폴리펩티드를 코드화하는 cDNA및 그 용도
EP2264059A1 (fr) Polypeptide, l'ADNc qui les code et leur utilisation
EP1652924A2 (fr) Polypeptide, l'ADNc qui la code et leur utilisation
IE912281A1 (en) New protein with cell-differentiation or cell-formation¹ability and recombinant process for the production
US20020123613A1 (en) Endoderm, cardiac and neural inducing factors - oligonucleotides for expressing xenopus frazzled (frzb-1) protein
US20100285527A1 (en) Novel Polypeptide, cDNA Encoding the Same, and Use Thereof
JP2000300275A (ja) 新規タンパク質hsgl及びその遺伝子
EP0894137A1 (fr) Nouveau gene de reponse a la differenciation terminale myeloide d'origine humaine

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IL IS JP KE KG KP KR KZ LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA