CA2274803C - Receptor activator of nf-kappa b, receptor is member of tnf receptor superfamily - Google Patents

Receptor activator of nf-kappa b, receptor is member of tnf receptor superfamily Download PDF

Info

Publication number
CA2274803C
CA2274803C CA2274803A CA2274803A CA2274803C CA 2274803 C CA2274803 C CA 2274803C CA 2274803 A CA2274803 A CA 2274803A CA 2274803 A CA2274803 A CA 2274803A CA 2274803 C CA2274803 C CA 2274803C
Authority
CA
Canada
Prior art keywords
rank
polypeptide
seq
amino acid
ser
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.)
Expired - Fee Related
Application number
CA2274803A
Other languages
French (fr)
Other versions
CA2274803A1 (en
Inventor
Dirk M. Anderson
Laurent J. Galibert
Eugene Maraskovsky
William C. Dougall
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.)
Immunex Corp
Original Assignee
Immunex Corp
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 Immunex Corp filed Critical Immunex Corp
Priority claimed from PCT/US1997/023866 external-priority patent/WO1998028424A2/en
Publication of CA2274803A1 publication Critical patent/CA2274803A1/en
Application granted granted Critical
Publication of CA2274803C publication Critical patent/CA2274803C/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70575NGF/TNF-superfamily, e.g. CD70, CD95L, CD153, CD154
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/02Fusion polypeptide containing a localisation/targetting motif containing a signal sequence

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Toxicology (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

Isolated receptors, DNAs encoding such receptors, and pharmaceutical compositions made therefrom, are disclosed. The isolated receptors can be used to regulate an immune response. The receptors are also useful in screening for inhibitors thereof.

Description

TITLE
RECEPTOR ACTIVATOR OF NF-KAPPA B, RECEPTOR IS MEMBER OF TNF RECEPTOR
SUPERFAMILY

TECHNICAL FIELD OF THE INVENTION
The present invention relates generally to the field of cytokine receptors, and more specifically to cytokine receptor/ligand pairs having immunoregulatory activity.
BACKGROUND OF THE INVENTION
Efficient functioning of the immune system requires a fine balance between cell proliferation and differentiation and cell death, to ensure that the immune system is capable of reacting to foreign, but not self antigens. Integral to the process of regulating the immune and inflammatory response are various members of the Tumor Necrosis Factor (TNF) Receptor/Nerve Growth Factor Receptor superfamily (Smith et al., Science 248:1019; 1990). This family of receptors includes two different TNF receptors (Type I
and Type II; Smith et al., supra; and Schall et al., Cell 61:361, 1990), nerve growth factor receptor (Johnson et al., Cell 47:545, 1986), B cell antigen CD40 (Stamenkovic et al., EMBO J. 8:1403, 1989), CD27 (Camerini et al., J. Immunol. 147:3165, 1991), (Durkop et al., Cell 68:421, 1992), T cell antigen OX40 (Mallett et al., EMBO
J. 9:1063, 1990), human Fas antigen (Itoh et al., Cell 66:233, 1991), murine 4-1BB
receptor (Kwon et al., Proc. Natl. Acad. Sci. USA 86:1963, 1989) and a receptor referred to as Apoptosis-Inducing Receptor (AIR; USSN 08/720,864, filed October 4, 1996).
CD40 is a receptor present on B lymphocytes, epithelial cells and some carcinoma cell lines that interacts with a ligand found on activated T cells, CD40L
(USSN
08/249,189, filed May 24, 1994). The interaction of this ligand/receptor pair is essential for both the cellular and humoral immune response. Signal transduction via CD40 is mediated through the association of the cytoplasmic domain of this molecule with members of the TNF receptor-associated factors (TRAFs; Baker and Reddy, Oncogene 12:1, 1996).
It has recently been found that mice that are defective in TRAF3 expression due to a targeted disruption in the gene encoding TRAF3 appear normal at birth but develop progressive hypoglycemia and depletion of peripheral white cells, and die by about ten days of age (Xu et al., Immunity 5:407, 1996). The immune responses of chimeric mice reconstituted with TRAF3-/- fetal liver cells resemble those of CD40-deficient mice, although TRAF34- B cells appear to be functionally normal.
The critical role of TRAF3 in signal transduction may be in its interaction with one of the other members of the TNF receptor superfamily, for example, CD30 or CD27, which are present on T cells. Alternatively, there may be other, as yet unidentified SUBSTITUTE SHEET (RULE 26) members of this family of receptors that interact with TRAF3 and play an important role in postnatal development as well as in the development of a competent immune system.
Identifying additional members of the TNF receptor superfamily would provide an additional means of regulating the immune and inflammatory response, as well as potentially providing further insight into post-natal development in mammals.

SUMMARY OF THE INVENTION
The present invention provides a novel receptor, referred to as RANK (for receptor activator of NF-KB), that is a member of the TNF receptor superfamily. RANK is a Type I
transmembrane protein having 616 amino acid residues that interacts with TRAF3.
Triggering of RANK by over-expression, co-expression of RANK and membrane bound RANK ligand (RANKL), and with addition of soluble RANKL or agonistic antibodies to RANK results in the upregulation of the transcription factor NF-KB, a ubiquitous transcription factor that is most extensively utilized in cells of the immune system.
Soluble forms of the receptor can be prepared and used to interfere with signal transduction through membrane-bound RANK, and hence upregulation of NF-KB;
accordingly, pharmaceutical compositions comprising soluble forms of the novel receptor are also provided. Inhibition of NF-xB by RANK antagonists may be useful in ameliorating negative effects of an inflammatory response that result from triggering of RANK, for example in treating toxic shock or sepsis, graft-versus-host reactions, or acute inflammatory reactions. Soluble forms of the receptor will also be useful in vitro to screen for agonists or antagonists of RANK activity.
The cytoplasmic domain of RANK will be useful in developing assays for inhibitors of signal transduction, for example, for screening for molecules that inhibit interaction of RANK with TRAF2 or TRAF3. Deleted forms and fusion proteins comprising the novel receptor are also disclosed.
The present invention also identifies a counterstructure, or ligand, for RANK, referred to as RANKL. RANKL is a Type 2 transmembrane protein with an intracellular domain of less than about 50 amino acids, a transmembrane domain and an extracellular domain of from about 240 to 250 amino acids. Similar to other members of the TNF
family to which it belongs, RANKL has a `spacer' region between the transmembrane domain and the receptor binding domain that is not necessary for receptor binding.
Accordingly, soluble forms of RANKL can comprise the entire extracellular domain or fragments thereof that include the receptor binding region.
One specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide capable of binding receptor activator of NF-KB ligand (RANKL) or binding a Tumor Necrosis Factor receptor-associated factor (TRAF) selected from the group consisting of (a) a polypeptide comprising the amino acid sequence as set forth in SEQ ID NO:6; and (b) a polypeptide comprising the amino acid sequence x to y of SEQ ID NO:6, wherein x is any one of amino acids 1-33 and y is any one of amino acids 196-616; wherein RANKL consists of the amino acid sequence of SEQ ID NO:13.

Another specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide encoded by a DNA capable of hybridizing to the complement of nucleotides 33-213 of SEQ ID NO:5 under the conditions of hybridizing at 6XSSC at 63 C and washing in 3XSSC at 55 C, wherein the RANK polypeptide is capable of binding receptor activator of NF-KB ligand (RANKL) that comprises the amino acid sequence of SEQ ID NO:13 or binding a Tumor Necrosis Factor receptor-associated factor (TRAF).
Another specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide comprising amino acids 339-421, inclusive, of SEQ ID NO:6, wherein said polypeptide is capable of binding TRAF6.

Another specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide comprising amino acids 544-616, inclusive, of SEQ ID NO:6, wherein said polypeptide is capable of binding a TRAF selected from the group consisting of TRAF1, TRAF2, TRAF3, TRAF5 and TRAF6.

2a Another specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide consisting of the amino acid sequence according to SEQ ID NO:6 or a fragment thereof that is capable of binding a receptor activator of NF-KB ligand (RANKL) protein that consists of the amino acid sequence shown in SEQ ID
NO:13, wherein said polypeptide or fragment is capable of inducing an antibody that binds specifically with a polypeptide consisting of the amino acid sequence set forth in SEQ ID NO:6.

Another specific aspect of the invention relates to a receptor activator of NF-KB (RANK) fusion polypeptide capable of binding a receptor activator of NF-KB ligand (RANKL) polypeptide consisting of the amino acid sequence of SEQ ID NO:13, said fusion polypeptide comprising the RANK
polypeptide according to any one of claims 1 to 19 fused to an Fc region of a human immunoglobulin protein or a GST
polypeptide.

Another specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide capable of binding receptor activator of NF-KB
ligand (RANKL) or binding a Tumor Necrosis Factor receptor-associated factor (TRAF) selected from the group consisting of: (a) a polypeptide comprising the amino acid sequence as set forth in SEQ ID NO:15; and (b) a polypeptide comprising the amino acid sequence x to y of SEQ ID NO: 15, wherein x is amino acid 1 or any one of amino acids 25-35 and y is any one of amino acids 197-214, wherein RANKL consists of the amino acid sequence of SEQ ID NO:11 or 13.

2b Another specific aspect of the invention relates to a receptor activator of NF-KB (RANK) fusion polypeptide comprising amino acids x to 213 of SEQ ID NO:15, wherein x is amino acid 1 or 35 of SEQ ID NO:15, and amino acids 3-232 of SEQ ID NO:8, wherein the RANK fusion polypeptide is capable of binding a receptor activator of NF-KB ligand (RANKL) polypeptide consisting of the amino acid sequence of SEQ ID NO:13 or a TRAF protein.

Another specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide encoded by a DNA capable of hybridizing to the complement of the nucleotide sequence set forth in SEQ ID
N0:5 under stringent conditions of hybridizing at 6XSSC at 63 C and washing in 3XSSC at 55 C, and wherein the polypeptide is capable of binding a TRAF selected from the group consisting of TRAF1, TRAF2, TRAF3, TRAF5 and TRAF6.
Another specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide produced by a process comprising culturing a host cell transformed or transfected with a recombinant expression vector containing the nucleotide sequence set forth in SEQ ID NO:5 or containing a nucleotide sequence capable of hybridizing to the complement of the nucleotide sequence set forth in SEQ ID NO:5 under the stringent conditions of hybridizing in 6XSSC at 63 C and washing in 3XSSC at 55 C, and isolating the polypeptide resulting from the expression of said nucleotide sequence, said polypeptide being capable of binding a receptor activator of NF-KB

ligand (RANKL) protein that consists of the amino acid sequence shown in SEQ ID NO:13.

2c Another specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide which is at least 80% identical in sequence to amino acids 544-616 of SEQ ID NO:6, wherein said polypeptide is capable of binding a TRAF selected from the group consisting of TRAF1, TRAF2, TRAF3, TRAF5 and TRAF6.
Another specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide that is at least 70% identical in amino acid sequence to a polypeptide with the amino acid sequence x to y of SEQ ID N0:6, wherein x is any one of amino acids 1-33 and y is any one of amino acids 213-616, wherein said isolated polypeptide is capable of binding a receptor activator of NF-KB ligand (RANKL) polypeptide that consists of the amino acid sequence shown in SEQ ID NO:13.

Another specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide comprising an amino acid sequence with at least 80% sequence identity to SEQ ID NO:6, wherein said isolated polypeptide is capable of binding a receptor activator of NF-KB ligand (RANKL) polypeptide that consists of the amino acid sequence shown in SEQ ID NO:13.

Another specific aspect of the invention relates to an isolated receptor activator of NF-KB (RANK) polypeptide comprising an amino acid sequence with at least 80% sequence identity to SEQ ID NO:15, wherein said isolated polypeptide is capable of binding to a receptor activator of NF-KB ligand (RANKL) polypeptide consisting of SEQ ID NO:11.
2d Another specific aspect of the invention relates to a soluble receptor activator of NF-KB (RANK) polypeptide comprising the amino acid sequence x to y of SEQ ID NO:6, wherein x is any one of amino acids 1-33 and y is any one of amino acids 196-213 or fragments thereof, wherein the soluble RANK is capable of binding to a receptor activator of NF-KB ligand (RANKL) protein that consists of the amino acid sequence of SEQ ID NO:13.

Another specific aspect of the invention relates to a primer comprising at least 17 contiguous nucleotides of SEQ ID NO:5 and which binds specifically to the complement of the nucleotide sequence set forth as SEQ ID NO:5.

Another specific aspect of the invention relates to a primer which comprises at least 17 contiguous nucleotides of SEQ ID NO:5 and which binds specifically to the complement of the nucleotide sequence set forth as SEQ
ID NO:5.

Another specific aspect of the invention relates to a primer which comprises at least 17 contiguous nucleotides of SEQ ID NO:14 and which binds specifically to the complement of the nucleotide sequence set forth as SEQ
ID NO:5.

Another specific aspect of the invention relates to a DNA molecule capable of hybridization to the complement of the nucleotide sequence set forth in SEQ ID NO:5 under stringent conditions of hybridizing at 6XSSC at 63 C and washing in 3XSSC at 55 C, and which encodes receptor activator of NF-KB (RANK) that is capable of binding to 2e receptor activator of NF-KB ligand (RANKL) consisting of the amino acid sequence of SEQ ID NO:13.

Another specific aspect of the invention relates to a DNA molecule capable of hybridizing to the complement of nucleotides 33-213 of SEQ ID NO:5 under stringent conditions of hybridizing at 6XSSC at 63 C and washing in 3XSSC at 55 C, and which encodes receptor activator of NF-KB
(RANK) that is capable of binding to receptor activator of NF-KB ligand (RANKL) consisting of the amino acid sequence of SEQ ID NO:13.

Another specific aspect of the invention relates to an isolated antibody that specifically binds a RANK
polypeptide of amino acids 1-616 of SEQ ID NO:6; or amino acids 33-213 of SEQ ID NO:6.

Another specific aspect of the invention relates to a method for preparing an antibody, comprising immunizing a non-human mammal with a polypeptide comprising amino acids 544-616 of SEQ ID NO:6 in an amount effective to produce antibodies which bind specifically to the polypeptide and harvesting the antibodies.

Another specific aspect of the invention relates to a method for preparing an antibody, comprising immunizing a non-human mammal with a polypeptide comprising amino acids 339-421 of SEQ ID NO:6 in an amount effective to produce antibodies which bind specifically to the polypeptide and harvesting the antibodies.

Another specific aspect of the invention relates to a method for preparing an antibody, comprising immunizing a non-human mammal with a polypeptide comprising amino acids 33-213 of SEQ ID NO:6 in an amount effective to produce 2f antibodies which bind specifically to the polypeptide and harvesting the antibodies.

Another specific aspect of the invention relates to use of a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, of SEQ ID NO:61 and an Fc from a human IgG1 immunoglobulin for inhibiting RANK activity in a subject having a tumor or neoplastic disease, wherein RANK
activity is the ability of RANK to bind RANK ligand, andwherein said RANK:FC polypeptide is in an amount sufficient to inhibit RANK signal transduction in said subject, wherein RANK comprises SEQ ID NO:6.

Another specific aspect of the invention relates to use of a fusion protein comprising a receptor activator of NF-KB (RANK) polypeptide linked to a human immunoglobulin Fc region, wherein said RANK polypeptide has at least 90%
amino acid sequence identity with amino acids 33 to 213 of SEQ ID NO:6 and is capable of binding a RANKL protein consisting of the amino acid sequence shown in SEQ ID NO:13, and further wherein said RANK polypeptide is in an amount sufficient to inhibit RANK signal transduction in a subject having a tumor or neoplastic disease.

Another specific aspect of the invention relates to use of a composition that comprises a receptor activator of NF-KB (RANK) polypeptide and a physiologically acceptable carrier, excipient or diluent for treating a subject having a tumor or neoplastic disease, wherein the RANK polypeptide has an at least 90% amino acid sequence identity with amino acids 33-213 of SEQ ID NO:6; is capable of binding a receptor activator of NF-KB ligand (RANKL) polypeptide that 2g consists of the amino acid sequence shown in SEQ ID NO:13;
is in an amount sufficient to inhibit RANK signal transduction in said subject.

Another specific aspect of the invention relates to a kit comprising a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, of SEQ ID NO:6, and an Fc from a human immunoglobulin; together with instructions for inhibiting RANK activity in a subject having a tumor or neoplastic disease, wherein said RANK:Fc polypeptide is in an amount sufficient to inhibit RANK
signal transduction in said subject.

Another specific aspect of the invention relates to a kit comprising a fusion protein comprising a RANK

polypeptide linked to a human immunoglobulin Fc region, wherein said RANK polypeptide has at least 90% amino acid sequence identity with amino acids 33 to 213 of SEQ ID NO:6 and is capable of binding a RANKL protein consisting of the amino acid sequence shown in SEQ ID NO:13, and further wherein said RANK polypeptide is in an amount sufficient to inhibit RANK signal transduction in said subject, together with instructions for inhibiting RANK activity in a subject having a tumor or neoplastic disease.

Another specific aspect of the invention relates to a kit comprising a composition that comprises a receptor activator of NF-KB (RANK) polypeptide capable of binding a RANKL polypeptide that consists of the amino acid sequence shown in SEQ ID NO:13, wherein the RANK polypeptide has an at least 90% amino acid sequence identity with amino acids 33-213 of SEQ ID NO:6 and a physiologically acceptable 2h carrier, excipient or diluent together with instructions for treating a subject having a tumor or neoplastic disease.
Another specific aspect of the invention relates to a method of screening a molecule for its capacity to antagonize or agonize receptor activator of NF-KB (RANK), which comprises SEQ ID NO:6, said method comprising: a) contacting a RANK polypeptide or RANK fusion polypeptide as described herein with a receptor activator of NF-KB ligand (RANKL) polypeptide, which comprises SEQ ID NO: 13, or a fragment thereof that binds said RANK polypeptide, in the presence of the molecule under conditions that permit binding of the RANK and RANKL polypeptides; (b) detecting the binding of the RANK and RANKL polypeptides; (c) determining that the molecule is a RANK antagonist if the amount of RANK-RANKL binding detected is decreased when the molecule is present; and (d) determining that the molecule is a RANK agonist if the amount of RANK-RANKL binding detected is increased when the molecule is present.

Another specific aspect of the invention relates to a method of screening a molecule for its capacity to agonize or antagonize RANK, said method comprising the steps of: (a) incubating the molecule with a cell that expresses a RANK polypeptide on its surface, wherein said RANK
polypeptide has an amino acid sequence selected from the group consisting of: (i) amino acids x to y of SEQ ID NO:6, wherein x is any one of amino acids 1-33 and y is amino acid 616; and (ii) a polypeptide that is at least 80% identical to amino acids 33-616 of SEQ ID NO:6 and that is capable of binding a RANKL polypeptide consisting of amino acids 1-317 of SEQ ID NO:13; (b) measuring a biological activity 2i associated with the RANK polypeptide, wherein said biological activity is selected from the group consisting of: (i) binding a RANKL polypeptide, which comprises SEQ ID
NO: 13, or a RANK-binding fragment thereof; and (ii) ability of the RANK polypeptide to bind TRAF1, TRAF2, TRAF3, TRAF5 or TRAF6; and (c) determining that the molecule is a RANK
agonist if the biological activity is increased in the presence of the molecule or that the molecule is a RANK
antagonist if the biological activity is decreased in the presence of the molecule.

2j BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 demonstrates the influence of RANK.Fc and hRANKL on activated T cell growth. Human peripheral blood T cells were cultured as described in Example 12; viable T cell recovery was determined by triplicate trypan blue countings.
Figure 2 illustrates the ability of RANKL to induce human DC cluster formation.
Functionally mature dendritic cells (DC) were generated in vitro from CD34+
bone marrow (BM) progenitors and cultured as described in Example 13. CD 1 a+ DC were cultured in a cytokine cocktail alone (Figure 2A), in cocktail plus CD40L (Figure 2B), RANKL
(Figure 2C), or heat inactivated (oH) RANKL (Figure 2D), and then photographed using an inversion microscope.
Figure 3 demonstrates that RANKL enhances DC allo-stimulatory capacity.
Allogeneic T cells were incubated with varying numbers of irradiated DC
cultured as described in Example 13. The cultures were pulsed with [3H]-thymidine and the cells harvested onto glass fiber sheets for counting. Values represent the mean standard deviation (SD) of triplicate cultures.
Figure 4 presents an alignment of human RANK with other TNFR family members in the region of structurally conserved extracellular cysteine-rich pseudorepeats. Predicted disulfide linkages (DS 1-DS3) are indicated. RANK and CD40 contain identical amino acid substitutions (CAH, CAG) eliminating DS2 in the second pseudorepeat.
Figure 5 presents an alignment of human RANKL with other TNF family members.
DETAILED DESCRIPTION OF THE INVENTION
A novel partial cDNA insert with a predicted open reading frame having some similarity to CD40 was identified in a database containing sequence information from cDNAs generated from human bone marrow-derived dendritic cells (DC). The insert was used to hybridize to colony blots generated from a DC cDNA library containing full-length cDNAs. Several colony hybridizations were performed, and two clones (SEQ ID
NOs: I
and 3) were isolated. SEQ ID NO:5 shows the nucleotide and amino acid sequence of a predicted full-length protein based on alignment of the overlapping sequences of SEQ ID
NOs:I and 3.
RANK is a member of the TNF receptor superfamily; it most closely resembles CD40 in the extracellular region. Similar to CD40, RANK associates with TRAF2 and TRAF3 (as determined by co-immunoprecipitation assays substantially as described by Rothe et al., Cell 83:1243, 1995). TRAFs are critically important in the regulation of the immune and inflammatory response. Through their association with various members of the TNF receptor superfamily, a signal is transduced to a cell. That signal results in the proliferation, differentiation or apoptosis of the cell, depending on which receptor(s) is/are triggered and which TRAF(s) associate with the receptor(s); different signals can be SUBSTITUTE SHEET (RULE 26) transduced to a cell via coordination of various signaling events. Thus, a signal transduced through one member of this family may be proliferative, differentiative or apoptotic, depending on other signals being transduced to the cell, and/or the state of differentiation of the cell. Such exquisite regulation of this proliferative/apoptotic pathway is necessary to develop and maintain protection against pathogens; imbalances can result in autoimmune disease.
RANK is expressed on epithelial cells, some B cell lines, and on activated T
cells.
However, its expression on activated T cells is late, about four days after activation. This time course of expression coincides with the expression of Fas, a known agent of apoptosis. RANK may act as an anti-apoptotic signal, rescuing cells that express RANK
from apoptosis as CD40 is known to do. Alternatively, RANK may confirm an apoptotic signal under the appropriate circumstances, again similar to CD40. RANK and its ligand are likely to play an integral role in regulation of the immune and inflammatory response.
Moreover, the post-natal lethality of mice having a targeted disruption of the gene demonstrates the importance of this molecule not only in the immune response but in development. The isolation of RANK, as a protein that associates with TRAF3, and its ligand will allow further definition of this signaling pathway, and development of diagnostic and therapeutic modalities for use in the area of autoimmune and/or inflammatory disease.
DNAs. Proteins and Analogs The present invention provides isolated RANK polypeptides and analogs (or muteins) thereof having an activity exhibited by the native molecule (i.e, RANK muteins that bind specifically to a RANK ligand expressed on cells or immobilized on a surface or to RANK-specific antibodies; soluble forms thereof that inhibit RANK ligand-induced signaling through RANK). Such proteins are substantially free of contaminating endogenous materials and, optionally, without associated native-pattern glycosylation.
Derivatives of RANK within the scope of the invention also include various structural forms of the primary proteins which retain biological activity. Due to the presence of ionizable amino and carboxyl groups, for example, a RANK protein may be in the form of acidic or basic salts, or may be in neutral form. Individual amino acid residues may also be modified by oxidation or reduction. The primary amino acid structure may be modified by forming covalent or aggregative conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like, or by creating amino acid sequence mutants. Covalent derivatives are prepared by linking particular functional groups to amino acid side chains or at the N- or C-termini.
Derivatives of RANK may also be obtained by the action of cross-linking agents, such as M-maleimidobenzoyl succinimide ester and N-hydroxysuccinimide, at cysteine and lysine residues. The inventive proteins may also be covalently bound through reactive side groups to various insoluble substrates, such as cyanogen bromide-activated, bisoxirane-activated, carbonyldiimidazole-activated or tosyl-activated agarose structures, or by adsorbing to polyolefin surfaces (with or without glutaraldehyde cross-linking). Once bound to a substrate, the proteins may be used to selectively bind (for purposes of assay or purification) antibodies raised against the proteins or against other proteins which are similar to RANK or RANKL, as well as other proteins that bind RANK or RANKL or homologs thereof.
Soluble forms of RANK are also within the scope of the invention. The nucleotide and predicted amino acid sequence of the RANK is shown in SEQ ID NOs:I through 6.
Computer analysis indicated that the protein has an N-terminal signal peptide;
the predicted cleavage site follows residue 24. Those skilled in the art will recognize that the actual cleavage site may be different than that predicted by computer analysis. Thus, the N-terminal amino acid of the cleaved peptide is expected to be within about five amino acids on either side of the predicted, preferred cleavage site following residue 24.
Moreover a soluble form beginning with amino acid 33 was prepared; this soluble form bound RANKL. The signal peptide is predicted to be followed by a 188 amino acid extracellular domain, a 21 amino acid transmembrane domain, and a 383 amino acid cytoplasmic tail.
Soluble RANK comprises the signal peptide and the extracellular domain (residues 1 to 213 of SEQ ID NO:6) or a fragment thereof. Alternatively, a different signal peptide can be substituted for the native leader, beginning with residue 1 and continuing through a residue selected from the group consisting of amino acids 24 through 33 (inclusive) of SEQ
ID NO:6. Moreover, fragments of the extracellular domain will also provide soluble forms of RANK. Fragments can be prepared using known techniques to isolate a desired portion of the extracellular region, and can be prepared, for example, by comparing the extracellular region with those of other members of the TNFR family and selecting forms similar to those prepared for other family members. Alternatively, unique restriction sites or PCR
techniques that are known in the art can be used to prepare numerous truncated forms which can be expressed and analyzed for activity.
Fragments can be prepared using known techniques to isolate a desired portion of the extracellular region, and can be prepared, for example, by comparing the extracellular region with those of other members of the TNFR family (of which RANK is a member) and selecting forms similar to those prepared for other family members.
Alternatively, unique restriction sites or PCR techniques that are known in the art can be used to prepare numerous truncated forms which can be expressed and analyzed for activity.
Other derivatives of the RANK proteins within the scope of this invention include covalent or aggregative conjugates of the proteins or their fragments with other proteins or polypeptides, such as by synthesis in recombinant culture as N-terminal or C-terminal fusions. For example, the conjugated peptide may be a signal (or leader) polypeptide sequence at the N-terminal region of the protein which co-translationally or post-translationally directs transfer of the protein-from its site of synthesis to its site of function inside or outside of the cell membrane or wall (e.g., the yeast (X-factor leader).
Protein fusions can comprise peptides added to facilitate purification or identification of RANK proteins and homologs (e.g., poly-His). The amino acid sequence of the inventive proteins can also be linked to an identification peptide such as that described by Hopp et al., BiolTechnology 6:1204 (1988). Such a highly antigenic peptide provides an epitope reversibly bound by a specific monoclonal antibody, enabling rapid assay and facile purification of expressed recombinant protein. The sequence of Hopp et al. is also specifically cleaved by bovine mucosal enterokinase, allowing removal of the peptide from the purified protein. Fusion proteins capped with such peptides may also be resistant to intracellular degradation in E. coli.
Fusion proteins further comprise the amino acid sequence of a RANK linked to an immunoglobulin Fc region. An exemplary Fc region is a human IgG, having a nucleotide an amino acid sequence set forth in SEQ ID NO:8. Fragments of an Fc region may also be used, as can Fc muteins. For example, certain residues within the hinge region of an Fc region are critical for high affinity binding to FcyRI. Canfield and Morrison (J. Exp. Med.
173:1483; 1991) reported that Leu(234) and Leu(235)were critical to high affinity binding of IgG3 to FcyRI present on U937 cells. Similar results were obtained by Lund et al. (J.
Immunol. 147:2657, 1991; Molecular Immunol. 29:53, 1991). Such mutations, alone or in combination, can be made in an IgG, Fc region to decrease the affinity of IgG, for FcR.
Depending on the portion of the Fc region used, a fusion protein may be expressed as a dimer, through formation of interchain disulfide bonds. If the fusion proteins are made with both heavy and light chains of an antibody, it is possible to form a protein oligomer with as many as four RANK regions.
In another embodiment, RANK proteins further comprise an oligomerizing peptide such as a leucine zipper domain. Leucine zippers were originally identified in several DNA-binding proteins (Landschulz et al., Science 240:1759, 1988). Leucine zipper domain is a term used to refer to a conserved peptide domain present in these (and other) proteins, which is responsible for dimerization of the proteins. The leucine zipper domain (also referred to herein as an oligomerizing, or oligomer-forming, domain) comprises a repetitive heptad repeat, with four or five leucine residues interspersed with other amino acids. Examples of leucine zipper domains are those found in the yeast transcription factor GCN4 and a heat-stable DNA-binding protein found in rat liver (C/EBP;
Landschulz et al., Science 243:1681, 1989). Two nuclear transforming proteins, fos and jun, also exhibit leucine zipper domains, as does the gene product of the murine proto-oncogene, c-myc (Landschulz et al., Science 240:1759, 1988). The products of the nuclear oncogenes fos and jun comprise leucine zipper domains preferentially form a heterodimer (O'Shea et al., Science 245:646, 1989; Turner and Tjian, Science 243:1689, 1989). The leucine zipper domain is necessary for biological activity (DNA binding) in these proteins.
The fusogenic proteins of several different viruses, including paramyxovirus, coronavirus, measles virus and many retroviruses, also possess leucine zipper domains (Buckland and Wild, Nature 338:547,1989; Britton, Nature 353:394, 1991;
Delwart and Mosialos, AIDS Research and Human Retroviruses 6:703, 1990). The leucine zipper domains in these fusogenic viral proteins are near the transmembrane region of the proteins;
it has been suggested that the leucine zipper domains could contribute to the oligomeric structure of the fusogenic proteins. Oligomerization of fusogenic viral proteins is involved in fusion pore formation (Spruce et al, Proc. Natl. Acad. Sci. U.S.A. 88:3523, 1991).
Leucine zipper domains have also been recently reported to play a role in oigomerization of heat-shock transcription factors (Rabindran et al., Science 259:230, 1993).
Leucine zipper domains fold as short, parallel coiled coils. (O'Shea et al., Science 254:539; 1991) The general architecture of the parallel coiled coil has been well characterized, with a "knobs-into-holes" packing as proposed by Crick in 1953 (Acta Crystallogr. 6:689). The dimer formed by a leucine zipper domain is stabilized by the heptad repeat, designated (abcdefg)õ according to the notation of McLachlan and Stewart (J. Mol. Biol. 98:293; 1975), in which residues a and d are generally hydrophobic residues, with d being a leucine, which line up on the same face of a helix.
Oppositely-charged residues commonly occur at positions g and e. Thus, in a parallel coiled coil formed from two helical leucine zipper domains, the "knobs" formed by the hydrophobic side chains of the first helix are packed into the "holes" formed between the side chains of the second helix.
The leucine residues at position d contribute large hydrophobic stabilization energies, and are important for dimer formation (Krystek et al., Int. J.
Peptide Res.
38:229, 1991). Lovejoy et al. recently reported the synthesis of a triple-stranded a-helical bundle in which the helices run up-up-down (Science 259:1288, 1993). Their studies confirmed that hydrophobic stabilization energy provides the main driving force for the formation of coiled coils from helical monomers. These studies also indicate that electrostatic interactions contribute to the stoichiometry and geometry of coiled coils.
Several studies have indicated that conservative amino acids may be substituted for individual leucine residues with minimal decrease in the ability to dimerize;
multiple changes, however, usually result in loss of this ability (Landschulz et al., Science 243:1681, 1989; Turner and Tjian, Science 243:1689, 1989; Hu et al., Science 250:1400, 1990). van Heekeren et al. reported that a number of different amino residues can be substituted for the leucine residues in the leucine zipper domain of GCN4, and further found that some GCN4 proteins containing two leucine substitutions were weakly active (Nucl. Acids Res. 20:3721, 1992). Mutation of the first and second heptadic leucines of the leucine zipper domain of the measles virus fusion protein (MVF) did not affect syncytium formation (a measure of virally-induced cell fusion); however,-mutation of all four leucine residues prevented fusion completely (Buckland et al., J. Gen.
Virol. 73:1703, 1992). None of the mutations affected the ability of MVF to form a tetramer.
Amino acid substitutions in the a and d residues of a synthetic peptide representing the GCN4 leucine zipper domain have been found to change the oligomerization properties of the leucine zipper domain (Alber, Sixth Symposium of the Protein Society, San Diego, CA). When all residues at position a are changed to isoleucine, the leucine zipper still forms a parallel dimer. When, in addition to this change, all leucine residues at position d are also'changed to isoleucine, the resultant peptide spontaneously forms a trimeric parallel' coiled coil in solution. Substituting all amino acids at position d with isoleucine and at position a with leucine results in a peptide that tetramerizes. Peptides containing these substitutions are still referred to as leucine zipper domains.
Also included within the scope of the invention are fragments or derivatives of the intracellular domain of RANK. Such fragments are prepared by any of the herein-mentioned techniques, and include peptides that are identical to the cytoplasmic domain of RANK as shown in SEQ ID NO:6, or of murine RANK as shown in SEQ ID NO:15, and those that comprise a portion of the cytoplasmic region. All techniques used in preparing soluble forms may also be used in preparing fragments or analogs of the cytoplasmic domain (i.e., RT-PCR techniques or use of selected restriction enzymes to prepare truncations). DNAs encoding all or a fragment of the intracytoplasmic domain will be useful in identifying other proteins that are associated with RANK signalling, for example using the immunoprecipitation techniques described herein, or another technique such as a yeast two-hybrid system (Rothe et al., supra).
The present invention also includes RANK with or without associated native-pattern glycosylation. Proteins expressed in yeast or mammalian expression systems, e.g., COS-7 cells, may be similar or slightly different in molecular weight and glycosylation pattern than the native molecules, depending upon the expression system. Expression of DNAs encoding the inventive proteins in bacteria such as E. coli provides non-glycosylated molecules. Functional mutant analogs of RANK protein having inactivated N-glycosylation sites can be produced by oligonucleotide synthesis and ligation or by site-specific mutagenesis techniques. These analog proteins can be produced in a homogeneous, reduced-carbohydrate form in good yield using yeast expression systems.
N-glycosylation sites in eukaryotic proteins are characterized by the amino acid triplet Asn-A,-Z, where A, is any amino acid except Pro, and Z is Ser or Thr. In this sequence, asparagine provides a side chain amino group for covalent attachment of carbohydrate.
Such a site can be eliminated by substituting another amino acid for Asti or for residue Z, deleting Asn or Z, or inserting a non-Z amino acid between Al and Z, or an amino acid other than Asn between Asn and A,.

RANK protein derivatives may also be obtained by mutations of the native RANK
or subunits thereof. A RANK mutated protein, as referred to herein, is a polypeptide homologous to a native RANK protein, respectively, but which has an amino acid sequence different from the native protein because of one or a plurality of deletions, insertions or substitutions. The effect of any mutation made in a DNA encoding a mutated peptide may be easily determined by analyzing the ability of the mutated peptide to bind its counterstructure in a specific manner. Moreover, activity of RANK analogs, muteins or derivatives can be determined by any of the assays described herein (for example, inhibition of the ability of RANK to activate transcription).
Analogs of the inventive proteins may be constructed by, for example, making various substitutions of residues or sequences or deleting terminal or internal residues or sequences not needed for biological activity. For example, cysteine residues can be deleted or replaced with other amino acids to prevent formation of incorrect intramolecular disulfide bridges upon renaturation. Other approaches to mutagenesis involve modification of adjacent dibasic amino acid residues to enhance expression in yeast systems in which KEX2 protease activity is present.
When a deletion or insertion strategy is adopted, the potential effect of the deletion or insertion on biological activity should be considered. Subunits of the inventive proteins may be constructed by deleting terminal or internal residues or sequences.
Soluble forms of RANK can be readily prepared and tested for their ability to inhibit RANK-induced NF-KB activation. Polypeptides corresponding to the cytoplasmic regions, and fragments thereof (for example, a death domain) can be prepared by similar techniques.
Additional guidance as to the types of mutations that can be made is provided by a comparison of the sequence of RANK to proteins that have similar structures, as well as by performing structural analysis of the inventive RANK proteins.
Generally, substitutions should be made conservatively; i.e., the most preferred substitute amino acids are those which do not affect the biological activity of RANK (i.e., ability of the inventive proteins to bind antibodies to the corresponding native protein in substantially equivalent a manner, the ability to bind the counterstructure in substantially the same manner as the native protein, the ability to transduce a RANK signal, or ability to induce NF-KB activation upon overexpression in transient transfection systems.
for example). Examples of conservative substitutions include substitution of amino acids outside of the binding domain(s) (either ligand/receptor or antibody binding areas for the extracellular domain, or regions that interact with other, intracellular proteins for the cytoplasmic domain), and substitution of amino acids that do not alter the secondary and/or tertiary structure of the native protein. Additional examples include;
substituting one aliphatic residue for another, such as Ile, Val, Leu, or Ala for one another, or substitutions of one polar residue for another, such as between Lys and Arg; Glu and Asp; or Gin and Asn. Other such conservative substitutions, for example, substitutions of entire regions having similar hydrophobicity characteristics, are well known.
Mutations in nucleotide sequences constructed for expression of analog proteins or fragments thereof must, of course, preserve the reading frame phase of the coding sequences and preferably will not create complementary regions that could hybridize to produce secondary mRNA structures such as loops or hairpins which would adversely affect translation of the mRNA.
Not all mutations in the nucleotide sequence which encodes a RANK protein or fragments thereof will be expressed in the final product, for example, nucleotide substitutions may be made to enhance expression, primarily to avoid secondary structure loops in the transcribed mRNA (see EPA 75,444A) or to provide codons that are more readily translated by the selected host, e.g., the well-known E. coli preference codons for E. coli expression.
Although a mutation site may be predetermined, it is not necessary that the nature of the mutation per se be predetermined. For example, in order to select for optimum characteristics of mutants, random mutagenesis may be conducted and the expressed mutated proteins screened for the desired activity. Mutations can be introduced at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes an analog having the desired amino acid insertion, substitution, or deletion.
Alternatively, oligonucleotide-directed site-specific mutagenesis procedures can be employed to provide an altered gene having particular codons altered according to the substitution, deletion, or insertion required. Exemplary methods of making the alterations set forth above are disclosed by Walder et al. (Gene 42:133, 1986); Bauer et al. (,Gene 37:73, 1985); Craik (BioTechniques, January 1985, 12-19); Smith et al.
(Genetic Engineering: Principles and Methods, Plenum Press, 1981); and U.S. Patent NOs.
4,518,584 and 4,737,462 disclose suitable techniques.
Other embodiments of the inventive proteins include RANK polypeptides encoded by DNAs capable of hybridizing to the DNA of SEQ ID NO:5, under moderately stringent conditions (prewashing solution of 5 X SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0) and hybridization conditions of 50 C, 5 X SSC, overnight) to the DNA sequences encoding RANK, or more preferably under, stringent conditions (for example, hybridization in 6 X
SSC at 63 C overnight; washing in 3 X SSC at 55 C), and other sequences which are degenerate to those which encode the RANK. In one embodiment, RANK
polypeptides are at least about 70% identical in amino acid sequence to the amino acid sequence of native RANK protein as set forth in SEQ ID NO.6. In a preferred embodiment, RANK
polypeptides are at least about 80% identical in amino acid sequence to the native form of RANK; most preferred polypeptides are those that are at least about 90%
identical to native RANK.
Percent identity may be determined using a computer program, for example, the GAP computer program described by Devereux et al. (Nucl. Acids Res. 12:387, 1984) and available from the University of Wisconsin Genetics Computer Group (UWGCG).
For fragments derived from the RANK protein, the identity is calculated based on that portion of the RANK protein that is present in the fragment -The biological activity of RANK analogs or muteins can be determined by testing the ability of the analogs or muteins to inhibit activation of transcription, for example as described in the Examples herein. Alternatively, suitable assays, for example, an enzyme immunoassay or a dot blot, employing an antibody that binds native RANK, or a soluble form of RANKL, can be used to assess the activity of RANK analogs or muteins, as can assays that employ cells expressing RANKL. Suitable assays also include, for example, signal transduction assays and methods that evaluate the ability of the cytoplasmic region of RANK to associate with other intracellular proteins (i.e., TRAFs 2 and 3) involved in signal transduction will also be useful to assess the activity of RANK analogs or muteins.
Such methods are well known in the art.
Fragments of the RANK nucleotide sequences are also useful. In one embodiment, such fragments comprise at least about 17 consecutive nucleotides, preferably at least about nucleotides, more preferably at least 30 consecutive nucleotides, of the RANK
DNA
25 disclosed herein. DNA and RNA complements of such fragments are provided herein, along with both single-stranded and double-stranded forms of the RANK DNA of SEQ ID
NO:5, and those encoding the aforementioned polypeptides. A fragment of RANK
DNA
generally comprises at least about 17 nucleotides, preferably from about 17 to about 30 nucleotides. Such nucleic acid fragments (for example, a probe corresponding to the extracellular domain of RANK) are used as a probe or as primers in a polymerase chain reaction (PCR).
The probes also find use in detecting the presence of RANK nucleic acids in in vitro assays and in such procedures as Northern and Southern blots. Cell types expressing RANK can be identified as well. Such procedures are well known, and the skilled artisan can choose a probe of suitable length, depending on the particular intended application. For PCR, 5' and 3' primers corresponding to the termini of a desired RANK
DNA sequence are employed to amplify that sequence, using conventional techniques.

Other useful fragments of the RANK nucleic acids are antisense or sense oligonucleotides comprising a single-stranded nucleic acid sequence (either RNA or DNA) capable of binding to target RANK mRNA (sense) or RANK DNA (antisense) sequences.
The ability to create an antisense or a sense oligonucleotide, based upon a cDNA sequence for a given protein is described in, for example, Stein and Cohen, Cancer Res.
48:2659, 1988 and van der Krol et al., BioTechniques 6:958, 1988.

Uses of DNAs, Proteins and Analogs The RANK DNAs, proteins and analogs described herein will have numerous uses, including the preparation of pharmaceutical compositions. For example, soluble forms of RANK will be useful as antagonists of RANK-mediated NF-KB activation, as well as to inhibit transduction of a signal via RANK. RANK compositions (both protein and DNAs) will also be useful in development of both agonistic and antagonistic antibodies to RANK.
The inventive DNAs are useful for the expression of recombinant proteins, and as probes for analysis (either quantitative or qualitative) of the presence or distribution of RANK
transcripts.
The inventive proteins will also be useful in preparing kits that are used to detect soluble RANK or RANKL, or monitor RANK-related activity, for example, in patient specimens. RANK proteins will also find uses in monitoring RANK-related activity in other samples or compositions, as is necessary when screening for antagonists or mimetics of this activity (for example, peptides or small molecules that inhibit or mimic, respectively, the interaction). A variety of assay formats are useful in such kits, including (but not limited to) ELISA, dot blot, solid phase binding assays (such as those using a biosensor), rapid format assays and bioassays.
The purified RANK according to the invention will facilitate the discovery of inhibitors of RANK, and thus, inhibitors of an inflammatory response (via inhibition of NF-KB activation). The use of a purified RANK polypeptide in the screening for potential inhibitors is important and can virtually eliminate the possibility of interfering reactions with contaminants. Such a screening assay can utilize either the extracellular domain of RANK, the intracellular domain, or a fragment of either of these polypeptides.
Detecting the inhibiting activity of a molecule would typically involve use of a soluble form of RANK
derived from the extracellular domain in a screening assay to detect molecules capable of binding RANK and inhibiting binding of, for example, an agonistic antibody or RANKL, or using a polypeptide derived from the intracellular domain in an assay to detect inhibition of the interaction of RANK and other, intracellular proteins involved in signal transduction.
Moreover, in vitro systems can be used to ascertain the ability of molecules to antagonize or agonize RANK activity. Included in such methods are uses of RANK
chimeras, for example, a chimera of the RANK intracellular domain and an extracellular *rB

domain derived from a protein having a known ligand. The effects on signal transduction of various molecule can then be monitored by utilizing the known ligand to transduce a signal.
In addition, RANK polypeptides can also be used for structure-based design of RANK-inhibitors. Such structure-based design is also known as "rational drug design."
The RANK polypeptides can be three-dimensionally analyzed by, for example, X-ray crystallography, nuclear magnetic resonance or homology modeling, all of which are well-known methods. The use of RANK structural information in molecular modeling software systems to assist in inhibitor design is also encompassed by the invention.
Such computer-assisted modeling and drug design may utilize information such as chemical conformational analysis, electrostatic potential of the molecules, protein folding, etc. A
particular method of the invention comprises analyzing the three dimensional structure of RANK
for likely binding sites of substrates, synthesizing a new molecule that incorporates a predictive reactive site, and assaying the new molecule as described above.
Expression of Recombinant RANK
The proteins of the present invention are preferably produced by recombinant DNA
methods by inserting a DNA sequence encoding RANK protein or an analog thereof into a recombinant expression vector and expressing the DNA sequence in a recombinant expression system under conditions promoting expression. DNA sequences encoding the proteins provided by this invention can be assembled from cDNA fragments and short oligonucleotide linkers, or from a series of oligonucleotides, to provide a synthetic gene which is capable of being inserted in a recombinant expression vector and expressed in a recombinant transcriptional unit.
Recombinant expression vectors include synthetic or cDNA-derived DNA
fragments encoding RANK, or homologs, muteins or bioequivalent analogs thereof, operably linked to suitable transcriptional or translational regulatory elements derived from mammalian, microbial, viral or insect genes. Such regulatory elements include a transcriptional promoter, an optional operator sequence to control transcription, a sequence encoding suitable mRNA ribosomal binding sites, and sequences which control the termination of transcription and translation, as described in detail below.
The ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants may additionally be incorporated.
DNA regions are operably linked when they are functionally related to each other.
For example, DNA for a signal peptide (secretory leader) is operably linked to DNA for a polypeptide if it is expressed as a precursor which participates in the secretion of the polypeptide; a promoter is operably linked to a coding sequence if it controls the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to permit translation. Generally, operably linked means contiguous and, in the case of secretory leaders, contiguous and in reading frame. DNA
sequences encoding RANK, or homologs or analogs thereof which are to be expressed in a microorganism will preferably contain no introns that could prematurely terminate transcription of DNA into mRNA.
Useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017). Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and pGEMI (Promega Biotec, Madison, WI, USA). These pBR322 "backbone"
sections are combined with an appropriate promoter and the structural sequence to be expressed. E. coli is typically transformed using derivatives of pBR322, a plasmid derived from an E. coli species (Bolivar et al., Gene 2:95, 1977). pBR322 contains genes for ampicillin and tetracycline resistance and thus provides simple means for identifying transformed cells.
Promoters commonly used in recombinant microbial expression vectors include the P-lactamase (penicillinase) and lactose promoter system (Chang et al., Nature 275:615, 1978; and Goeddel et al., Nature 281:544, 1979), the tryptophan (trp) promoter system (Goeddel et al., Nucl. Acids Res. 8:4057, 1980; and EPA 36,776) and tac promoter (Maniatis, Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory, p.
412, 1982). A particularly useful bacterial expression system employs the phage ~PL
promoter and c1857ts thermolabile repressor. Plasmid vectors available from the American Type Culture Collection which incorporate derivatives of the k PL promoter include plasmid pHUB2, resident in E. coli strain JMB9 (ATCC 37092) and pPLc28, resident in E. coli RR1 (ATCC 53082).
Suitable promoter sequences in yeast vectors include the promoters for metallothionein, 3-phosphoglycerate kinase (Hitzeman et al., J. Biol. Chem.
255:2073, 1980) or other glycolytic enzymes (Hess et al., J. Adv. Enzyme Reg. 7:149, 1968; and Holland et al., Biochem. 17:4900, 1978), such as enolase, glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, 3-phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase. Suitable vectors and promoters for use in yeast expression are further described in R. Hitzeman et al., EPA
73,657.
Preferred yeast vectors can be assembled using DNA sequences from pBR322 for selection and replication in E. coli (Ampr gene and origin of replication) and yeast DNA
sequences including a glucose-repressible ADH2 promoter and a-factor secretion leader.
The ADH2 promoter has been described by Russell et al. (J. Biol. Chem.
258:2674, 1982) and Beier et al. (Nature 300:724, 1982). The yeast a-factor leader, which directs secretion *rB

of heterologous proteins, can be inserted between the promoter and the structural gene to be expressed. See, e.g., Kurjan et al., Cell 30:933, 1982; and Bitter et al., Proc. Natl. Acad.
Sci. USA 81:5330, 1984. The leader sequence may be modified to contain, near its 3' end, one or more useful restriction sites to facilitate fusion of the leader sequence to foreign genes.
The transcriptional and translational control sequences in expression vectors to be used in transforming vertebrate cells may be provided by viral sources. For example, commonly used promoters and enhancers are derived from Polyoma, Adenovirus 2, Simian Virus 40 (SV40), and human cytomegalovirus. DNA sequences derived from the viral genome, for example, SV40 origin, early and late promoter, enhancer, splice, and polyadenylation sites may be used to provide the other genetic elements required for expression of a heterologous DNA sequence. The early and late promoters are particularly useful because both are obtained easily from the virus as a fragment which also contains the SV40 viral origin of replication (Fiers et al., Nature 273:113, 1978). Smaller or larger SV40 fragments may also be used, provided the approximately 250 bp sequence extending from the Hind III site toward the BglI site located in the viral origin of replication is included. Further, viral genomic promoter, control and/or signal sequences may be utilized, provided such control sequences are compatible with the host cell chosen.
Exemplary vectors can be constructed as disclosed by Okayama and Berg (Mol.
Cell. Biol.
3:280, 1983).
A useful system for stable high level expression of mammalian receptor cDNAs in C127 murine mammary epithelial cells can be constructed substantially as described by Cosman et al. (Mol. Immunol. 23:935, 1986). A preferred eukaryotic vector for expression of RANK DNA is referred to as pDC406 (McMahan et al., EMBO J.
10:2821, 1991), and includes regulatory sequences derived from SV40, human immunodeficiency virus (HIV), and Epstein-Barr virus (EBV). Other preferred vectors include pDC409 and pDC410, which are derived from pDC406. pDC410 was derived from pDC406 by substituting the EBV origin of replication with sequences encoding the SV40 large T
antigen. pDC409 differs from pDC406 in that a Bgl II restriction site outside of the multiple cloning site has been deleted, making the Bgl II site within the multiple cloning site unique.
A useful cell line that allows for episomal replication of expression vectors, such as pDC406 and pDC409, which contain the EBV origin of replication, is CV-1/EBNA
(ATCC
CRL 10478). The CV-1/EBNA cell line was derived by transfection of the CV-1 cell line with a gene encoding Epstein-Barr virus nuclear antigen-1 (EBNA-1) and constitutively express EBNA-1 driven from human CMV immediate-early enhancer/promoter.

Host Cells Transformed host cells are cells which have been transformed or transfected with expression vectors constructed using recombinant DNA techniques and which contain sequences encoding the proteins of the present invention. Transformed host cells may express the desired protein (RANK, or homologs or analogs thereof), but host cells transformed for purposes of cloning or amplifying the inventive DNA do not need to express the protein. Expressed proteins will preferably be secreted into the culture supernatant, depending on the DNA selected, but may be deposited in the cell membrane.
Suitable host cells for expression of proteins include prokaryotes, yeast or higher eukaryotic cells under the control of appropriate promoters. Prokaryotes include gram negative or gram positive organisms, for example E. coli or Bacillus spp.
Higher eukaryotic cells include established cell lines of mammalian origin as described below.
Cell-free translation systems could also be employed to produce proteins using RNAs derived from the DNA constructs disclosed herein. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, and mammalian cellular hosts are described by Pouwels et al. (Cloning Vectors: A Laboratory Manual, Elsevier, New York, 1985).
Prokaryotic expression hosts may be used for expression of RANK, or homologs or analogs thereof that do not require extensive proteolytic and disulfide processing.
Prokaryotic expression vectors generally comprise one or more phenotypic selectable markers, for example a gene encoding proteins conferring antibiotic resistance or supplying an autotrophic requirement, and an origin of replication recognized by the host to ensure amplification within the host. Suitable prokaryotic hosts for transformation include E. coli, Bacillus subtilis, Salmonella typhimurium, and various species within the genera Pseudomonas, Streptomyces, and Staphylococcus, although others may also be employed as a matter of choice.
Recombinant RANK may also be expressed in yeast hosts, preferably from the Saccharomyces species, such as S. cerevisiae. Yeast of other genera, such as Pichia or Kluyveromyces may also be employed. Yeast vectors will generally contain an origin of replication from the 2g yeast plasmid or an autonomously replicating sequence (ARS), promoter, DNA encoding the protein, sequences for polyadenylation and transcription termination and a selection gene. Preferably, yeast vectors will include an origin of replication and selectable marker permitting transformation of both yeast and E. coli, e.g., the ampicillin resistance gene of E. coli and S. cerevisiae trpl gene, which provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, and a promoter derived from a highly expressed yeast gene to induce transcription of a structural sequence downstream. The presence of the trpl lesion in the yeast host cell genome then provides an effective environment for detecting transformation by growth in the absence of tryptophan.
Suitable yeast transformation protocols are known to those of skill in the art; an exemplary technique is described by Hinnen et al., Proc. Natl. Acad. Sci. USA
75:1929, 1978, selecting for Trp+ transformants in a selective medium consisting of 0.67% yeast nitrogen base, 0.5% casamino acids, 2% glucose, 10 g/ml adenine and 20.tg/ml uracil.
Host strains transformed by vectors comprising the ADH2 promoter may be grown for expression in a rich medium consisting of I % yeast extract, 2% peptone, and I% glucose supplemented with 80 jig/nil adenine and 80 .tg/ml uracil. Derepression of the promoter occurs upon exhaustion of medium glucose. Crude yeast supernatants are fiarvested by filtration and held at 4 C prior to further purification.
Various mammalian or insect cell culture systems can be employed to express recombinant protein. Baculovirus systems for production of heterologous proteins in insect cells are reviewed by Luckow and Summers, Biolfechnology 6:47 (1988). Examples of suitable mammalian host cell lines include the COS-7 lines of monkey kidney cells, described by Gluzman (Cell 23:175, 1981), and other cell lines capable of expressing an appropriate vector including, for example, CV-I/EBNA (ATCC CRL 10478), L
cells, C127, 3T3, Chinese hamster ovary (CHO), HeLa and BHK cell lines. Mammalian expression vectors may comprise nontranscribed elements such as an origin of replication, a suitable promoter and enhancer linked to the gene to be expressed, and other 5' or 3' flanking nontranscribed sequences, and 5' or 3' nontranslated sequences, such as necessary ribosome binding sites, a polyadenylation site, splice donor and acceptor sites, and transcriptional termination sequences.

Purification of Recombinant RANK
Purified RANK, and homologs or analogs thereof are prepared by culturing suitable host/vector systems to express the recombinant translation products of the DNAs of the present invention, which are then purified from culture media or cell extracts. For example, supernatants from systems which secrete recombinant protein into culture media can be first concentrated using a commercially available protein. concentration filter, for example, an AmiconTM or Millipore PelliconTM ultrafiltration unit.
Following the concentration step, the concentrate can be applied to a suitable purification matrix. For example, a suitable affinity matrix can comprise a counter structure protein or lectin or antibody molecule bound to a suitable support.
Alternatively, an anion exchange resin can be employed, for example, a matrix or substrate having pendant diethylaminoethyl (DEAE) groups. The matrices can be acrylamide, agarose, dextran, cellulose or other types commonly employed in protein purification.
Alternatively, a cation exchange step can be employed. Suitable cation exchangers include various insoluble matrices comprising sulfopropyl or carboxymethyl groups. Sulfopropyl groups are preferred. Gel filtration chromatography also provides a means of purifying the inventive proteins.
Affinity chromatography is a particularly preferred method of purifying RANK
and homologs thereof. For example, a RANK expressed as a fusion protein comprising an immunoglobulin Fc region can be purified using Protein A or Protein G affinity chromatography. Moreover, a RANK protein comprising an oligomerizing zipper domain may be purified on a resin comprising an antibody specific to the oligomerizing zipper domain. Monoclonal antibodies against the RANK protein may also be useful in affinity chromatography purification, by utilizing methods that are well-known in the art. A ligand may also be used to prepare an affinity matrix for affinity purification of RANK.
Finally, one or more reversed-phase high performance liquid chromatography (RP-HPLC) steps employing hydrophobic RP-HPLC media, e.g., silica gel having pendant methyl or other aliphatic groups, can be employed to further purify a RANK
composition.
Some or all of the foregoing purification steps, in various combinations, can also be employed to provide a homogeneous recombinant protein.
Recombinant protein produced in bacterial culture is usually isolated by initial extraction from cell pellets, followed by one or more concentration, salting-out, aqueous ion exchange or size exclusion chromatography steps. Finally, high performance liquid chromatography (HPLC) can be employed for final purification steps. Microbial cells employed in expression of recombinant protein can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents.
Fermentation of yeast which express the inventive protein as a secreted protein greatly simplifies purification. Secreted recombinant protein resulting from a large-scale fermentation can be purified by methods analogous to those disclosed by Urdal et al. (J.
Chromatog. 296:171, 1984). This reference describes two sequential, reversed-phase HPLC steps for purification of recombinant human GM-CSF on a preparative HPLC
column.
Protein synthesized in recombinant culture is characterized by the presence of cell components, including proteins, in amounts and of a character which depend upon the purification steps taken to recover the inventive protein from the culture.
These components ordinarily will be of yeast, prokaryotic or non-human higher eukaryotic origin and preferably are present in innocuous contaminant quantities, on the order of less than about 1 percent by weight. Further, recombinant cell culture enables the production of the inventive proteins free of other proteins which may be normally associated with the proteins as they are found in nature in the species of origin.

Uses and Administration of RANK Compositions The present invention provides methods of using therapeutic compositions comprising an effective amount of a protein and a suitable diluent and carrier, and methods for regulating an immune or inflammatory response. The use of RANK in conjunction with soluble cytokine receptors or cytokines, or other immunoregulatory molecules is also contemplated.
For therapeutic use, purified protein is administered to a patient, preferably a human, for treatment in a manner appropriate to the indication. Thus, for example, RANK
protein compositions administered to regulate immune function can be given by bolus injection, continuous infusion, sustained release from implants, or other suitable technique.
Typically, a therapeutic agent will be administered in the form of a composition comprising purified RANK, in conjunction with physiologically acceptable carriers, excipients or diluents. Such carriers will be nontoxic to recipients at the dosages and concentrations employed.
Ordinarily, the preparation of such protein compositions entails combining the inventive protein with buffers, antioxidants such as ascorbic acid, low molecular weight (less than about 10 residues) polypeptides, proteins, amino acids, carbohydrates including glucose, sucrose or dextrins, chelating agents such as EDTA, glutathione and other stabilizers and excipients. Neutral buffered saline or saline mixed with conspecific serum albumin are exemplary appropriate diluents. Preferably, product is formulated as a lyophilizate using appropriate excipient solutions (e.g., sucrose) as diluents. Appropriate dosages can be determined in trials. The amount and frequency of administration will depend, of course, on such factors as the nature and severity of the indication being treated, the desired response, the condition of the patient, and so forth.
Soluble forms of RANK and other RANK antagonists such as antagonistic monoclonal antibodies can be administered for the purpose of inhibiting RANK-induced induction of NF-KB activity. NF-KB is a transcription factor that is utilized extensively by cells of the immune system, and plays a role in the inflammatory response.
Thus, inhibitors of RANK signalling will be useful in treating conditions in which signalling through RANK has given rise to negative consequences, for example, toxic or septic shock, or graft-versus-host reactions. They may also be useful in interfering with the role of NF-KB in cellular transformation. Tumor cells are more responsive to radiation when their NF-KB is blocked; thus, soluble RANK (or other antagonists of RANK
signalling) will be useful as an adjunct therapy for disease characterized by neoplastic cells that express RANK.

The following examples are offered by way of illustration, and not by way of limitation. Those skilled in the art will recognize that variations of the invention embodied in the examples can be made, especially in light of the teachings of the various references cited herein.
EXAMPLE]
The example describes the identification and isolation of a DNA encoding a novel member of the TNF receptor superfamily. A partial cDNA insert with a predicted open reading frame having some similarity to CD40 (a cell-surface antigen present on the surface of both normal and neoplastic human B cells that has been shown to play an important role in B-cell proliferation and differentiation; Stamenkovic et a]., EMBO J.
8:1403, 1989), was identified in a database containing sequence information from cDNAs generated from human bone marrow-derived dendritic cells (DC). The insert was excised from the vector by restriction endonuclease digestion, gel purified. labeled with 32P, and used to hybridize to colony blots generated from a DC cDNA library containing larger cDNA
inserts using high stringency hybridization and washing techniques (hybridization in 5xSSC, 50%
formamide at 42 C overnight, washing in 0.5xSSC at 63 C); other suitable high stringency conditions are disclosed in Sambrook et al. in Molecular Cloning: A Laboratory Manual, 2nd ed. (Cold Spring Harbor Laboratory, Cold Spring Harbor, NY; 1989), 9.52-9.55.
Initial experiments yielded a clone referred to as 9D-8A (SEQ ID NO: 1);
subsequent analysis indicated that this clone contained all but the extreme 5' end of a novel cDNA, with predicted intron sequence at the extreme 5' end (nucleotides 1-92 of SEQ ID
NO: 1).
Additional colony hybridizations were performed, and a second clone was isolated. The second clone, referred to as 9D-15C (SEQ ID NO:3), contained the 5' end without intron interruption but not the full 3'end. SEQ ID NO:5 shows the nucleotide and amino acid sequence of a predicted full-length protein based on alignment of the overlapping sequences of SEQ ID NOs: I and 3.
The encoded protein was designated RANK, for receptor activator of NF-icB. The cDNA encodes a predicted Type I transmembrane protein having 616 amino acid residues, with a predicted 24 amino acid signal sequence (the computer predicted cleavage site is after Leu24), a 188 amino acid extracellular domain, a 21 amino acid transmembrane domain, and a 383 amino acid cytoplasmic tail. The extracellular region of RANK
displayed significant amino acid homology (38.5% identity, 52.3% similarity) to CD40. A
cloning vector (pB]uescriptSKTM-) containing human RANK sequence, designated pBluescript:huRANK (in E. coli DHIOB), was deposited with the American Type Culture Collection, Manassas, VA (ATCC) on December 20, 1996, under terms of the Budapest Treaty, and given accession number 98285.

This example describes construction of a RANK DNA construct to express a RANK/Fc fusion protein. A soluble form of RANK fused to the Fc region of human IgG, was constructed in the mammalian expression vector pDC409 (USSN 08/571,579).
This expression vector encodes the leader sequence of the Cytomegalovirus (CMV) open reading frame R27080 (SEQ ID NO:9), followed by amino acids 33-213 of RANK, followed by a mutated form of the constant domain of human IgG, that exhibits reduced affinity for Fc receptors (SEQ ID NO:8; for the fusion protein, the Fc portion of the construct consisted of Arg3 through Lys232). An alternative expression vector encompassing amino acids 1-213 of RANK (using the native leader sequence) followed by the IgG, mutein was also -prepared. - Both expression vectors were found to induce high levels of expression of the RANK/Fc fusion protein in transfected cells.
To obtain RANK/Fc protein, a RANK/Fc expression plasmid is transfected into CV-1/EBNA cells, and supernatants are collected for about one week. The RANK/Fc fusion protein is purified by means well-known in the art for purification of Fc fusion proteins, for example, by protein A sepharoseTM column chromatography according to manufacturer's recommendations (i.e., Pharmacia, Uppsala, Sweden). SDS-polyacrylamide gel electrophoresis analysis indicted that the purified RANK/Fc protein migrated with a molecular weight of -55kDa in the presence of a reducing agent, and at a molecular weight of -I I OkDa in the absence of a reducing agent.
N-terminal amino acid sequencing of the purified protein made using the CMV
R27080 leader showed 60% cleavage after Ala2O, 20% cleavage after Pro22 and 20%
cleavage after Arg28 (which is the Furin cleavage site; amino acid residues are relative to SEQ ID NO:9); N-terminal amino acid analysis of the fusion protein expressed with the native leader showed cleavage predominantly after G1n25 (80% after G1n25 and 20% after Arg23; amino acid residues are relative to SEQ ID NO:6, full-length RANK).
Both fusion proteins were able to bind a ligand for RANK is a specific manner (i.e., they bound to the surface of various cell lines such as a murine thymoma cell line, EL4), indicating that the presence of additional amino acids at the N-terminus of RANK does not interfere with its ability to bind RANKL. Moreover, the construct comprising the CMV leader encoded RANK beginning at amino acid 33; thus, a RANK peptide having an N-terminus at an amino acid between Arg23 and Pro33, inclusive, is expected to be able to bind a ligand for RANK in a specific manner.
Other members of the TNF receptor superfamily have a region of amino acids between the transmembrane domain and the ligand binding domain that is referred to as a 'spacer' region, which is not necessary for ligand binding. In RANK, the amino acids between 196 and 213 are predicted to form such a spacer region. Accordingly, a soluble form of RANK that terminates with an amino acid in this region is expected to retain the ability to bind a ligand for RANK in a specific manner. Preferred C-terminal amino acids for soluble RANK peptides are selected from the group consisting of amino acids 213 and 196 of SEQ ID NO:6, although other amino acids in the spacer region may be utilized as a C-terminus.

This example illustrates the preparation of monoclonal antibodies against RANK.
Preparations of purified recombinant RANK, for example, or transfected cells expressing high levels of RANK, are employed to generate monoclonal antibodies against RANK
using conventional techniques, such as those disclosed in U.S. Patent 4,411,993. DNA
encoding RANK can also be used as an immunogen, for example, as reviewed by Pardoll and Beckerleg in Immunity 3:165, 1995. Such antibodies are likely to be useful in interfering with RANK-induced signaling (antagonistic or blocking antibodies) or in inducing a signal by cross-linking RANK (agonistic antibodies), as components of diagnostic or research assays for RANK or RANK activity, or in affinity purification of RANK.
To immunize rodents, RANK immunogen is emulsified in an adjuvant (such as complete or incomplete Freund's adjuvant, alum, or another adjuvant, such as Ribi adjuvant R700 (Ribi, Hamilton, MT), and injected in amounts ranging from 10-100 Ag subcutaneously into a selected rodent, for example, BALB/c mice or Lewis rats.
DNA may be given intradermally (Raz et al., Proc. Natl. Acad. Sci. USA 91:9519, 1994) or intamuscularly (Wang et al., Proc. Natl. Acad. Sci. USA 90:4156, 1993); saline has been found to be a suitable diluent for DNA-based antigens. Ten days to three weeks days later, the immunized animals are boosted with additional immunogen and periodically boosted thereafter on a weekly, biweekly or every third week immunization schedule.
Serum samples are periodically taken by retro-orbital bleeding or tail-tip excision for testing by dot-blot assay (antibody sandwich), ELISA (enzyme-linked immunosorbent assay), immunoprecipitation, or other suitable assays, including FACS
analysis.
Following detection of an appropriate antibody titer, positive animals are given an intravenous injection of antigen in saline. Three to four days later, the animals are sacrificed, splenocytes harvested, and fused to a murine myeloma cell line (e.g., NS I or preferably Ag 8.653 [ATCC CRL 1580]). Hybridoma cell lines generated by this procedure are plated in multiple microtiter plates in a selective medium (for example, one containing hypoxanthine, aminopterin, and thymidine, or HAT) to inhibit proliferation of non-fused cells, myeloma-myeloma hybrids, and splenocyte-splenocyte hybrids.
Hybridoma clones thus generated can be screened by ELISA for reactivity with RANK, for example, by adaptations of the techniques disclosed by Engvall et al., Immunochem. 8:871 (1971) and in U.S. Patent 4,703,004. A preferred screening technique is the antibody capture technique described by Beckman et al., J.
Immunol.
144:4212 (1990). Positive clones are then injected into the peritoneal cavities of syngeneic rodents to produce ascites containing high concentrations (>I mg/ml) of anti-RANK
monoclonal antibody. The resulting monoclonal antibody can be purified by ammonium sulfate precipitation followed by gel exclusion chromatography. Alternatively, affinity chromatography based upon binding of antibody to protein A or protein G can also be used, as can affinity chromatography based upon binding to RANK protein.
Monoclonal antibodies were generated using RANK/Fc fusion protein as the immunogen. These reagents were screened to confirm reactivity against the RANK
protein. Using the methods described herein to monitor the activity of the mAbs, both blocking (i.e., antibodies that bind RANK and inhibit binding of a ligand to RANK) and non-blocking (i.e., antibodies that bind RANK and do not inhibit ligand binding) were isolated.

This example illustrates the induction of NF-KB activity by RANK in 293/EBNA
cells (cell line was derived by transfection of the 293 cell line with a gene encoding Epstein-Barr virus nuclear antigen-1 (EBNA-1) that constitutively express EBNA-1 driven from human CMV immediate-early enhancer/promoter). Activation of NF-KB activity was measured in 293/EBNA cells essentially as described by Yao et al. (Immunity 3:811, 1995). Nuclear extracts were prepared and analyzed for NF-KB activity by a gel retardation assay using a 25 base pair oligonucleotide spanning the NF-KB binding sites.
Two million cells were seeded into 10 cm dishes two days prior to DNA transfection and cultured in DMEM-F12 media containing 2.5% FBS (fetal bovine serum). DNA transfections were performed as described herein for the IL-8 promoter/reporter assays.
Nuclear extracts were prepared by solubilization of isolated nuclei with 400 mM
NaCl (Yao et al., supra). Oligonucleotides containing an NF-xB binding site were annealed and endlabeled with 32P using T4 DNA polynucleotide kinase. Mobility shift reactions contained 10.tg of nuclear extract, 4 tg of poly(dI-dC) and 15,000 cpm labeled double-stranded oligonucleotide and incubated at room temperature for 20 minutes.
Resulting protein-DNA complexes were resolved on a 6% native polyacrylamide gel in 0.25 X Tris-borate-EDTA buffer.
Overexpression of RANK resulted in induction of NF-KB activity as shown by an appropriate shift in the mobility of the radioactive probe on the gel. Similar results were observed when RANK was triggered by a ligand that binds RANK and transduces a signal to cells expressing the receptor (i.e., by co-transfecting cells with human RANK and murine RANKL DNA; see Example 7 below), and would be expected to occur when triggering is done with agonistic antibodies.

This example describes a gene promoter/reporter system based on the human Interleukin-8 (IL-8) promoter used to analyze the activation of gene transcription in vivo.
The induction of human IL-8 gene transcription by the cytokines Interleukin-1 (IL-1) or tumor necrosis factor-alpha (TNF-a) is known to be dependent upon intact NF-KB
and NF-IL-6 transcription factor binding sites. Fusion of the cytokine-responsive promoter with a cDNA encoding the murine IL-4 receptor (mIL-4R) allows measurement of promoter activation by detection of the heterologous reporter protein (mIL-4R) on the cell surface of transfected cells.
Human kidney epithelial cells (293/EBNA) are transfected (via the DEAE/DEXTRAN method) with plasmids encoding: 1). the reporter/promoter construct (referred to as pIL-8rep), and 2). the cDNA(s) of interest. DNA concentrations are always kept constant by the addition of empty vector DNA. The 293/EBNA cells are plated at a density of 2.5 x 104 cells/ml (3 ml/ well) in a 6 well plate and incubated for two days prior to transfection. Two days after transfection, the mIL-4 receptor is detected by a radioimmunoassay (RIA) described below.
In one such experiment, the 293JEBNA cells were co-transfected with DNA
encoding RANK and with DNA encoding RANKL (see Example 7 below). Co-expression of this receptor and its counterstructure by cells results in activation of the signaling process of RANK. For such co-transfection studies, the DNA concentration/well for the DEAF
transfection were as follows: 40 ng of pIL-8rep [pBluescriptSK- vector (Stratagene)]; 0.4 ng CD40 (DNA encoding CD40, a control receptor; pCDM8 vector); 0.4 ng RANK
(DNA
encoding RANK; pDC409 vector), and either 1-50 ng CD40L (DNA encoding the ligand for CD40, which acts as a positive control when co-transfected with CD40 and as a negative control when co-transfected with RANK; in pDC304) or RANKL (DNA
encoding a ligand for RANK; in pDC406). Similar experiments can be done using soluble RANKL
or agonistic antibodies to RANK to trigger cells transfected with RANK.
For the mIL-4R-specific RIA, a monoclonal antibody reactive with mIL-4R is labeled with 1211 via a Chloramine T conjugation method; the resulting specific activity is typically 1.5 x 1016 cpm/nmol. After 48 hours, transfected cells are washed once with media (DMEM/F12 5% FBS). Non-specific binding sites are blocked by the addition of pre-warmed binding media containing 5% non-fat dry milk and incubation at 37 C/5% CO2 in a tissue culture incubator for one hour. The blocking media is decanted and binding buffer containing 125I anti-mlL-4R (clone M 1; rat IgG 1) is added to the cells and incubated with rocking at room temperature for 1 hour. After incubation of the cells with the radio-labeled antibody, cells are washed extensively with binding buffer (2X) and twice with phosphate-buffered saline (PBS). Cells are lysed in I ml of 0.5M NaOH, and total radioactivity is measured with a gamma counter.
Using this assay, 293/EBNA coo-transfected with DNAs encoding RANK
demonstrated transcriptional activation, as shown by detection of mulL-4R on the cell surface. Overexpression of RANK resulted in transcription of muIL-4R, as did triggering of the RANK by RANKL. Similar results are observed when RANK is triggered by agonistic antibodies.

This example illustrates the association of RANK with TRAF proteins.
Interaction of RANK with cytoplasmic TRAF proteins was demonstrated by co-immunoprecipitation assays essentially as described by Hsu et al. (Cell 84:299; 1996). Briefly, 293/EBNA cells were co-transfected with plasmids that direct the synthesis of RANK and epitope-tagged (FLAG ; SEQ ID NO:7) TRAF2 or TRAF3. Two days after transfection, surface proteins were labeled with biotin-ester, and cells were lysed in a buffer containing 0.5% NP-40TH.
RANK and proteins associated with this receptor were immunoprecipitated with anti-RANK, washed extensively, resolved by electrophoretic separation on a 6-10%
SDS
polyacrylamide gel and electrophoretically transferred to a nitrocellulose membrane for Western blotting. The association of TRAF2 and TRAF3 proteins with RANK was visualized by probing the membrane with an antibody that specifically recognizes the FLAG epitope. TRAFs 2 and 3 did not immunopreciptitate with anti-RANK in the absence of RANK expression.

This example describes isolation of a ligand for RANK, referred to as RANKL, by direct expression cloning. The ligand was cloned essentially as described in US Patent No. 5,961,974, filed May 24, 1994, for CD40L. Briefly, a library was prepared from a clone of a mouse thymoma cell line EL-4 (ATCC TIB 39), called EL-40.5, derived by sorting five times with biotinylated CD40/Fc fusion protein in a FACS (fluorescence activated cell sorter). The cDNA library was made using standard methodology; the plasmid DNA was isolated and transfected into sub-confluent CVI-EBNA cells using a DEAE-dextran method.
Transfectants were screened by slide autoradiography for expression of RANKL
using a two-step binding method with RANK/Fc fusion protein as prepared in Example 2 followed by radioiodinated goat anti-human IgG antibody.
A clone encoding a protein that specifically bound RANK was isolated and sequenced; the clone was referred to as 11H. An expression vector containing murine RANKL sequence, designated pDC406:muRANK-L (in E. coli DH I OB), was deposited with the American Type Culture Collection, Manassas, VA (ATCC) on December 20, 1996, under terms of the Budapest Treaty, and given accession number 98284.
The nucleotide sequence and predicted amino acid sequence of this clone are illustrated in SEQ
ID NO: 10. This clone did not contain an initiator methionine; additional, full-length clones were obtained from a 7B9 library (prepared substantially as described in US
patent 5,599,905, issued February 4, 1997); the 5' region was found to be identical to that of human RANKL as shown in SEQ ID NO: 12, amino acids 1 through 22, except for substitution of a Gly for a Thr at residue 9.
This ligand is useful for assessing the ability of RANK to bind RANKL by a number of different assays. For example, transfected cells expressing RANKL
can be used in a FACS assay (or similar assay) to evaluate the ability of soluble RANK to bind RANKL. Moreover, soluble forms of RANKL can be prepared and used in assays that are known in the art (i.e., ELISA or BIAcoreTM assays essentially as described in US Patent No. 5,961,974, filed May 24, 1994). RANKL is also useful in affinity purification of RANK, and as a reagent in methods to measure the levels of RANK in a sample. Soluble RANKL
is also useful in inducing NF-KB activation and thus protecting cells that express RANK
from apoptosis.

This example describes the isolation of a human RANK ligand (RANKL) using a PCR-based technique. Murine RANK ligand-specific oligonucleotide primers were used in PCR reactions using human cell line-derived first strand cDNAs as templates.
Primers corresponded to nucleotides 478-497 and to the complement of nucleotides 858-878 of murine RANK ligand (SEQ ID NO:10). An amplified band approximately 400 bp in length from one reaction using the human epidermoid cell line KB (ATCC CCL- 17) was gel purified, and its nucleotide sequence determined; the sequence was 859b identical to the corresponding region of murine RANK ligand, confirming that the fragment was from human RANKL.
To obtain full-length human RANKL cDNAs, two human RANKL-specific oligonucleotides derived from the KB PCR product nucleotide sequence were radiolabeled and used as hybridization probes to screen a human PBL cDNA library prepared in lambda gt10 (Stratagene, La Jolla, CA), substantially as described in US patent 5,599,905, issued February 4, 1997 . Several positive hybridizing plaques were identified and purified, their inserts subcloned into pBluescript SK- (Stratagene, La Jolla, CA), and their nucleotide sequence determined. One isolate, PBL3, was found to encode most of the predicted human RANKL, but appeared to be missing approximately 200 bp of 5' coding region. A
second isolate, PBL5 was found to encode much of the predicted human RANKL, including the entire 5' end and an additional 200 bp of 5' untranslated sequence.

The 5' end of PBL5 and the 3' end of PBL3 were ligated together to form a full length cDNA encoding human RANKL. The nucleotide and predicted amino acid sequence of the full-length human RANK ligand is shown in SEQ ID NO: 12. Human RANK
ligand shares 83% nucleotide and 84% amino acid identity with murine RANK ligand. A
plasmid vector containing human RANKL sequence, designated pBluescript:huRANK-L (in E.
coli DH10B), was deposited with the American Type Culture Collection, Manassas, VA
.
(ATCC) on March 11, 1997 under terms of the Budapest Treaty, and given accession number 98354.
Morin and human RANKL are Type 2 transmembrane proteins. Murine RANKL
contains a predicted 48 amino acid intracellular domain, 21 amino acid transmembrane domain and 247 amino acid extracellular domain. Human RANKL contains a predicted 47 amino acid intracellular domain, 21 amino acid transmembrane domain and 249 amino acid extracellular domain.

This example describes the chromosomal mapping of human RANK using PCR-based mapping strategies. Initial human chromosomal assignments were made using RANK and RANKL-specific PCR primers and a BIOS Somatic Cell Hybrid PCRableTM
DNA kit from BIOS Laboratories (New Haven, CT), following the manufacturer's instructions. RANK mapped to human chromosome 18; RANK ligand mapped to human chromosome 13. More detailed mapping was performed using a radiation hybrid mapping panel GenebridgeTM 4 Radiation Hybrid Panel (Research Genetics, Huntsville, AL;
described in Walter, MA et al., Nature Genetics 7:22-28, 1994). RANK mapped to chromosome 18q22. 1, and RANKL mapped to chromosome 13814.

This example illustrates the preparation of monoclonal antibodies against RANKL.
Preparations of purified recombinant RANKL, for example, or transfixed cells expressing high levels of RANKL, are employed to generate monoclonal antibodies against RANKL
using conventional techniques, such as those disclosed in US Patent 4,411,993.
DNA
encoding RANKL can also be used as an immunogen, for example, as reviewed by Pardoll and Beckerleg in Immunity 3:165, 1995. Such antibodies are likely to be useful in interfering with RANKL signaling (antagonistic or blocking antibodies), as components of diagnostic or research assays for RANKL or RANKL activity, or in affinity purification of RANKL.
To immunize rodents, RANKL immunogen is emulsified in an adjuvant (such as complete or incomplete Freund's adjuvant, alum, or another adjuvant, such as Ribi adjuvant R700 (Ribi, Hamilton, MT), and injected in amounts ranging from 10-100 p.g subcutaneously into a selected rodent, for example, BALB/c mice or Lewis rats.
DNA may be given intradermally (Raz et al., Proc. Natl. Acad. Sci. USA 91:9519, 1994) or intamuscularly (Wang et al., Proc. Natl. Acad. Sci. USA 90:4156, 1993); saline has been found to be a suitable diluent for DNA-based antigens. Ten days to three weeks days later, the immunized animals are boosted with additional immunogen and periodically boosted thereafter on a weekly, biweekly or every third week immunization schedule.
Serum samples are periodically taken by retro-orbital bleeding or tail-tip excision for testing by dot-blot assay (antibody sandwich), ELISA (enzyme-linked immunosorbent assay), immunoprecipitation, or other suitable assays, including FACS
analysis.
Following detection of an appropriate antibody titer, positive animals are given an intravenous injection of antigen in saline. Three to four days later, the animals are sacrificed, splenocytes harvested, and fused to a murine myeloma cell line (e.g., NS 1 or preferably Ag 8.653 [ATCC CRL 1580]). Hybridoma cell lines generated by this procedure are plated in multiple microtiter plates in a selective medium (for example, one containing hypoxanthine, aminopterin, and thymidine, or HAT) to inhibit proliferation of non-fused cells, myeloma-myeloma hybrids, and splenocyte-splenocyte hybrids.
Hybridoma clones thus generated can be screened by ELISA for reactivity with RANKL, for example, by adaptations of the techniques disclosed by Engvall et al., Immunochem. 8:871 (1971) and in US Patent 4,703,004. A preferred screening technique is the antibody capture technique described by Beckman et al., J. Immunol.
144:4212 (1990). Positive clones are then injected into the peritoneal cavities of syngeneic rodents to produce ascites containing high concentrations (>I mg/ml) of anti-RANK
monoclonal antibody. The resulting monoclonal antibody can be purified by ammonium sulfate precipitation followed by gel exclusion chromatography. Alternatively, affinity chromatography based upon binding of antibody to protein A or protein G can also be used, as can affinity chromatography based upon binding to RANKL protein.
Using the methods described herein to monitor the activity of the mAbs, both blocking (i.e., antibodies that bind RANKL and inhibit binding to RANK) and non-blocking (i.e., antibodies that bind RANKL and do not inhibit binding) are isolated.

This example demonstrates that RANK expression can be up-regulated. Human peripheral blood T cells were purified by flow cytometry sorting or by negative selection using antibody coated beads, and activated with anti-CD3 (OKT3, Dako) coated plates or phytohemagglutinin in the presence or absence of various cytokines, including Interleukin-4 (IL-4),Transforming Growth Factor-B (TGF-B) and other commercially available cytokines ( IL 1-a, IL-2, IL-3, IL-6, IL-7, IL-8, IL-10, IL-12, IL-15, IFN-y, TNF- cc).
Expression of RANK was evaluated by FACS in a time course experiment for day 2 to day 8, using a mouse monoclonal antibody mAb144 (prepared as described in Example 3), as shown in the table below. Results are expressed as `+' to `++++' referring to the relative increase in intensity of staining with anti-RANK. Double labeling experiments using both anti-RANK and anti-CD8 or anti-CD4 antibodies were also performed.

Table 1: Upregulation of RANK by Cytokines Cytokine (concentration) Results:
IL-4 (50 ng/ml) +
TGF-B (5 ng/ml) + to ++
IL-4 (50 ng/ml) +TGF-B (5 ng/ml) ++++
IL 1-a (l Ong/ml) -IL-2 (20ng/ml) -IL-3 (25ng/ml) -IL-7 (20ng/ml) -IL-8 (lOng/ml) -IL-10 (50ng/ml) -IL-12 (1 Ong/ml) -IL-15 (l Ong/ml) -IFN-y (100U/ml) -TNF-a (l Ong/ml) -Of the cytokines tested, IL-4 and TGF-B increased the level of RANK expression on both CD8+ cytotoxic and CD4+ helper T cells from day 4 to day 8. The combination of IL-4 and TGF-B acted synergistically to upregulate expression of this receptor on activated T cells. This particular combination of cytokines is secreted by suppresser T
cells, and is believed to be important in the generation of tolerance (reviewed in Mitchison and Sieper, Z. Rheumatol. 54:141, 1995), implicating the interaction of RANK in regulation of an immune response towards either tolerance or induction of an active immune response.

This example illustrates the influence of RANK.Fc and hRANKL on activated T
cell growth. The addition of TGFB to anti-CD3 activated human peripheral blood T
lymphocytes induces proliferation arrest and ultimately death of most lymphocytes within the first few days of culture. We tested the effect of RANK:RANKL interactions on TGFB-treated T cells by adding RANK.Fc or soluble human RANKL to T cell cultures.
Human peripheral blood T cells (7 x IO 5 PBT) were cultured for six days on anti-CD3 (OKT3, 5 g/ml) and anti-Flag (M1, 5 g/ml) coated 24 well plates in the presence of TGFB (l ng/ml) and IL-4 (l 0ng/ml ), with or without recombinant FLAG-tagged soluble hRANKL (1 g/ml) or RANK.Fc (10 g/ml). Viable T cell recovery was determined by triplicate trypan blue countings.
The addition of RANK.Fc significantly reduced the number of viable T cells recovered after six days, whereas soluble RANKL greatly increased the recovery of viable T cells (Figure 1). Thus, endogenous or exogenous RANKL enhances the number of viable T cells generated in the presence of TGFB. TGFB, along with IL-4, has been implicated in immune response regulation when secreted by the TH3/regulatory T
cell subset. These T cells are believed to mediate bystander suppression of effector T cells.
Accordingly, RANK and its ligand may act in an auto/paracrine fashion to influence T cell tolerance. Moreover, TGFB is known to play a role in the evasion of the immune system effected by certain pathogenic or opportunistic organisms. In addition to playing a role in the development of tolerance, RANK may also play a role in immune system evasion by pathogens.

This example illustrates the influence of the interaction of RANK on CDla+
dendritic cells (DC). Functionally mature dendritic cells (DC) were generated in vitro from CD34+ bone marrow (BM) progenitors. Briefly, human BM cells from normal healthy volunteers were density fractionated using FicollTM medium and CD34+ cells immunoaffinity isolated using an anti-CD34 matrix column (CeprateTM, Cell Pro). The CD34+ BM cells were then cultured in human GM-CSF (20 ng/ml), human IL-4 (20 ng/ml), human TNF-a (20 ng/ml), human CHO-derived FIt3L (FL; 100 ng/ml) in Super McCoy's medium supplemented with 10% fetal calf serum in a fully humidified 37~C incubator (5% CO2) for 14 days. CD1a+, HLA-DR+ DC were then sorted using a FACStar P1usTM, and used for biological evaluation of RANK.

On human CDIa+ DC derived from CD34+ bone marrow cells, only a subset (20-30%) of CDIa+ DC expressed RANK at the cell surface as assessed by flow cytometric analysis. However, addition of CD40L to the DC cultures resulted in RANK
surface expression on the majority of CDIa+ DC. CD40L has been shown to activate DC by enhancing in vitro cluster formation, inducing DC morphological changes and upregulating HLA-DR, CD54, CD58, CD80 and CD86 expression Addition of RANKL to DC cultures significantly increased the degree of DC
aggregation and cluster formation above control cultures, similar to the effects seen with CD40L (Figure 2). Sorted human CDla+ DC were cultured in a cytokine cocktail (GM-CSF, IL-4, TNF-a and FL) (upper left panel), in cocktail plus CD40L (l g/ml) (upper right), in cocktail plus RANKL (1 g/ml) (lower left), or in cocktail plus heat inactivated (AH) RANKL (1 gg/ml) (lower right) in 24-well flat bottomed culture plates in 1 ml culture media for 48-72 hours and then photographed using an inversion microscope. An increase in DC aggregation and cluster formation above control cultures was not evident when heat inactivated RANKL was used, indicating that this effect was dependent on biologically active protein. However, initial phenotypic analysis of adhesion molecule expression indicated that RANKL-induced clustering was not due to increased levels of CD2, CD 11 a, CD54 or CD58.
The addition of RANKL to CDIa+ DC enhanced their allo-stimulatory capacity in a mixed lymphocyte reaction (MLR) by at least 3- to 10-fold, comparable to CD40L-cultured DC (Figure 3). Allogeneic T cells (1x105) were incubated with varying numbers of irradiated (2000 rad) DC cultured as indicated above for Figure 2 in 96-well round bottomed culture plates in 0.2 ml culture medium for four days. The cultures were pulsed with 0.5 mCi [3H]-thymidine for eight hours and the cells harvested onto glass fiber sheets for counting on a gas phase (i counter. The background counts for either T
cells or DC
cultured alone were <100 cpm. Values represent the mean SD of triplicate cultures. Heat inactivated RANKL had no effect. DC allo-stimulatory activity was not further enhanced when RANKL and CD40L were used in combination, possibly due to DC functional capacity having reached a maximal level with either cytokine alone. Neither RANKL nor CD40L enhanced the in vitro growth of DC over the three day culture period.
Unlike CD40L, RANKL did not significantly increase the levels of HLA-DR expression nor the expression of CD80 or CD86.
RANKL can enhance DC cluster formation and functional capacity without modulating known molecules involved in cell adhesion (CD 18, CD54), antigen presentation (HLA-DR) or costimulation (CD86), all of which are regulated by CD40/CD40L signaling. The lack of an effect on the expression of these molecules suggests that RANKL may regulate DC function via an alternate pathway(s) distinct from CD40/CD40L. Given that CD40L regulates RANK surface expression on in vitro-generated DC and that CD40L is upregulated on activated T cells during DC-T
cell interactions, RANK and its ligand may form an important part of the activation cascade that is induced during DC-mediated T cell expansion. Furthermore, culture of DC in RANKL
results in decreased levels of CDlb/c expression, and increased levels of CD83. Both of these molecules are similarly modulated during DC maturation by CD40L (Caux et al. J.
Exp. Med. 180:1263; 1994), indicating that RANKL induces DC maturation.
Dendritic cells are referred to as "professional" antigen presenting cells, and have a high capacity for sensitizing MHC-restricted T cells. There is growing interest in using dendritic cells ex vivo as tumor or infectious disease vaccine adjuvants (see, for example, Romani, et al., J. Exp. Med., 180:83, 1994). Therefore, an agent such as RANKL
that induces DC maturation and enhances the ability of dendritic cells to stimulate an immune response is likely to be useful in immunotherapy of various diseases.

This example describes the isolation of the murine homolog of RANK, referred to as muRANK. MuRANK was isolated by a combination of cross-species PCR and colony hybridization. The conservation of Cys residues in the Cys-rich pseudorepeats of the extracellular domains of TNFR superfamily member proteins was exploited to design human RANK-based PCR primers to be used on murine first strand cDNAs from various sources. Both the sense upstream primer and the antisense downstream primer were designed to have their 3' ends terminate within Cys residues.
The upstream sense primer encoded nucleotides 272-295 of SEQ ID NO:5 (region encoding amino acids 79-86); the downstream antisense primer encoded the complement of nucleotides 409-427 (region encoding amino acids 124-130). Standard PCR
reactions were set up and run, using these primers and first strand cDNAs from various murine cell line or tissue sources. Thirty reaction cycles of 94 C for 30 seconds, 50 C
for 30 seconds, and 72 C for 20 seconds were run. PCR products were anlyzed by electrophoresis, and specific bands were seen in several samples. The band from one sample was gel purified and DNA sequencing revealed that the sequence between the primers was approximately 85% identical to the corresponding human RANK nucleotide sequence.
A plasmid based cDNA library prepared from the murine fetal liver epithelium line FLE18 (one of the cell lines identified as positive in the PCR screen) was screened for full-length RANK cDNAs using murine RANK-specific oligonucleotide probes derived from the murine RANK sequence determined from sequencing the PCR product. Two cDNAs, one encoding the 5' end and one encoding the 3' end of full-length murine RANK
(based on sequence comparison with the full-length human RANK) were recombined to generate a full-length murine RANK cDNA. The nucleotide and amino acid seqeunce of muRANK
are shown in SEQ ID Nos: 14 and 15.

The cDNA encodes a predicted Type 1 transmembrane protein having 625 amino acid residues, with a predicted 30 amino acid signal sequence, a 184 amino acid extracellular domain, a 21 amino acid transmembrane domain, and a 390 amino acid cytoplasmic tail. The extracellular region of muRANK displayed significant amino acid homology (69.7% identity, 80.8% similarity) to huRANK. Those of skill in the art will recognize that the actual cleavage site can be different from that predicted by computer;
accordingly, the N-terminal of RANK may be from amino acid 25 to amino acid 35.
Other members of the TNF receptor superfamily have a region of amino acids between the transmembrane domain and the ligand binding domain that is referred to as a `spacer' region, which is not necessary for ligand binding. In muRANK, the amino acids between 197 and 214 are predicted to form such a spacer region. Accordingly, a soluble form of RANK that terminates with an amino acid in this region is expected to retain the ability to bind a ligand for RANK in a specific manner. Preferred C-terminal amino acids for soluble RANK peptides are selected from the group consisting of amino acids 214, and 197 of SEQ ID NO:14, although other amino acids in the spacer region may be utilized as a C-terminus.

This example illustrates the preparation of several different soluble forms of RANK
and RANKL. Standard techniques of restriction enzyme cutting and ligation, in combination with PCR-based isolation of fragments for which no convenient restriction sites existed, were used. When PCR was utilized, PCR products were sequenced to ascertain whether any mutations had been introduced; no such mutations were found.
In addition to the huRANK/Fc described in Example 2, another RANK/Fc fusion protein was prepared by ligating DNA encoding amino acids 1-213 of SEQ ID
NO:6, to DNA encoding amino acids 3-232 of the Fc mutein described previously (SEQ ID
NO:8).
A similar construct was prepared for murine RANK, ligating DNA encoding amino acids 1-213 of full-length murine RANK (SEQ ID NO: 15) to DNA encoding amino acids 3-232 of the Fc mutein (SEQ ID NO:8).
A soluble, tagged, poly-His version of huRANKL was prepared by ligating DNA
encoding the leader peptide from the immunoglobulin kappa chain (SEQ ID NO:
16) to DNA encoding a short version of the FLAGTM tag (SEQ ID NO:17), followed by codons encoding Gly Ser, then a poly-His tag (SEQ ID NO: 18), followed by codons encoding Gly Thr Ser, and DNA encoding amino acids 138-317 of SEQ ID NO: 13. A soluble, poly-His tagged version of murine RANKL was prepared by ligating DNA encoding the CMV
leader (SEQ ID NO:9) to codons encoding Arg Thr Ser, followed by DNA encoding poly-His (SEQ ID NO: 18) followed by DNA encoding amino acids 119-294 of SEQ ID NO: 11.

A soluble, oligomeric form of huRANKL was prepared by ligating DNA encoding the CMV leader (SEQ ID NO:9) to a codon encoding Asp followed by DNA ending a trimer-former "leucine" zipper (SEQ ID NO:19), then by codons encoding Thr Arg Ser followed by amino acids 138-317 of SEQ ID NO:13.
These and other constructs are prepared by routine experimentation. The various DNAs are then inserted into a suitable expression vector, and expressed.
Particularly preferred expression vectors are those which can be used in mammalian cells.
For example, pDC409 and pDC304, described herein, are useful for transient expression. For stable transfection, the use of CHO cells is preferred; several useful vectors are described in US Patent No. 6,027,915, for example, one of the 2A5-3 A-derived expression vectors discussed therein.

This example demonstrates that RANKL expression can be up-regulated on murine T cells. Cells were obtained from mesenteric lymph nodes of C57BL/6 mice, and activated with anti-CD3 coated plates, Concanavalin A (ConA) or phorbol rnyristate acetate in combination with ionomycin (anti-CD3: 500A2; Immunex Corporation, Seattle WA;
ConA, PMA, ionomycin, Sigma, St. Louis, MO) substantially as described herein, and cultured from about 2 to 5 days. Expression of RANKL was evaluated in a three color analysis by FACS, using antibodies to the T cell markers CD4, CD8 and CD45RB, and RANK/Fc, prepared as described herein.
RANKL was not expressed on unstimulated murine T cells. T cells stimulated with either anti-CD3, ConA, or PMA/ionomycin, showed differential expression of RANKL:
CD4+/CD45RBL0 and CD4'/CD45RBHi cells were positive for RANKL, but CD8+ cells were not. RANKL was not observed on B cells, similar to results observed with human cells.

This example illustrates the effects of murine RANKL on cell proliferation and activation. Various cells or cell lines representative of cells that play a role in an immune response (murine spleen, thymus and lymphnode) were evaluated by culturing them under conditions promoting their viability, in the presence or absence of RANKL.
RANKL did not stimulate any of the tested cells to proliferate. One cell line, a macrophage cell line referred to as RAW 264.7 (ATCC accession number TIB 71) exhibited some signs of activation.
RAW cells constitutively produce small amounts of TNF-a. Incubation with either human or murine RANKL enhanced production of TNF-a by these cells in a dose dependent manner. The results were not due to contamination of RANKL
preparations with endotoxin, since boiling RANKL for 10 minutes abrogated TNF-a production, whereas a similar treatment of purified endotoxin (LPS) did not affect the ability of the LPS
to stimulate TNF-a production. Despite the fact that RANKL activated the macrophage cell line RAW T64.7 for TNF-a production, neither human RANKL nor murine RANKL
stimulated nitric oxide production by these cells.

This example illustrates the effects of murine RANKL on growth and development of the thymus in fetal mice. Pregnant mice were injected with 1 mg of RANK/Fc or vehicle control protein (murine serum albumin; MSA) on days 13, 16 and 19 of gestation. After birth, the neonates continued to be injected with RANK/Fc intraperitoneally (IP) on a daily basis, beginning at a dose of I g, and doubling the dose about every four days, for a final dosage of 4 g. Neonates were taken at days 1, 8 and 15 post birth, their thymuses and spleens harvested and examined for size, cellularity and phenotypic composition.
A slight reduction in thymic size at day 1 was observed in the neonates born to the female injected with RANK/Fc; a similar decrease in size was not observed in the control neonates. At day 8, thymic size and cellularity were reduced by about 50% in the RANK/Fc-treated animals as compared to MSA treated mice. Phenotypic analysis demonstrated that the relative proportions of different T cell populations in the thymus were the same in the RANK/Fc mice as the control mice, indicating that the decreased cellularity was due to a global depression in the number of thymic T cells as opposed to a decrease in a specific population(s). The RANK/Fc-treated neonates were not significantly different from the control neonates at day 15 with respect to either size, cellularity or phenotype of thymic cells. No significant differences were observed in spleen size, cellularity or composition at any of the time points evaluated. The difference in cellularity on day 8 and not on day 15 may suggest that RANK/Fc may assert its effect early in thymic development.

This example demonstrates that the C-terminal region of the cytoplasmic domain of RANK is important for binding of several different TRAF proteins. RANK
contains at least two recognizable PXQX(X)T motifs that are likely TRAF docking sites.
Accordingly, the importance of various regions of the cytoplasmic domain of RANK for TRAF
binding was evaluated. A RANK/GST fusion protein was prepared substantially as described in Smith and Johnson, Gene 67:31 (1988), and used in the preparation of various truncations as described below.

Comparison of the nucleotide sequence of murine and human RANK indicated that there were several conserved regions that could be important for TRAF binding.
Accordingly, a PCR-based technique was developed to facilitate preparation of various C-terminal truncations that would retain the conserved regions. PCR primers were designed to introduce a stop codon and restriction enzyme site at selected points, yielding the truncations described in Table 1 below. Sequencing confirmed that no undesired mutations had been introduced in the constructs.
Radio-labeled (35S-Met, Cys) TRAF proteins were prepared by in vitro translation using a commercially available reticulocyte lysate kit according to manufacturer's instructions (Promega). Truncated GST fusion proteins were purified substantially as described in Smith and Johnson (supra). Briefly, E. coli were transfected with an expression vector encoding a fusion protein, and induced to express the protein. The bacteria were lysed, insoluble material removed, and the fusion protein isolated by precipitation with glutathione-coated beads (Sepahrose 4B, Pharmacia, Uppsala Sweden) The beads were washed, and incubated with various radiolabeled TRAF proteins.
After incubation and wash steps, the fusion protein/TRAF complexes were removed from the beads by boiling in 0.1% SDS + 13-mercaptoethanol, and loaded onto 12% SDS
gels (Novex). The gels were subjected to autoradiography, and the presence or absence of radiolabeled material recorded. The results are shown in Table 2 below.
Table 2: Binding of Various TRAF Proteins to the Cytoplasmic Domain of RANK
C terminal Truncations: E206-S339 E206-Y421 E206-M476 E206-G544 Full length TRAF1 - - - - ++
TRAF2 - - - - ++
TRAF3 - - - - ++

TRAF5 - - - - +
TRAF6 - + + + ++

These results indicate that TRAF1, TRAF2, TRAF3, TRAF 5 and TRAF6 bind to the most distal portion of the RANK cytoplasmic domain (between amino-acid G544 and A616). TRAF6 also has a binding site between S339 and Y421. In this experiment, TRAF5 also bound the cytoplasmic domain of RANK.

SEQUENCE LISTING
(1) GENERAL INFORMATION:

(i) APPLICANT: IMMUNEY CORPORATION

(ii) TITLE OF INVENTION: RECEPTOR ACTIVATOR OF NF-KAPPA B, RECEPTOR IS
MEMBER OF TNF RECEPTOR SUPERFAMILY

(iii) NUMBER OF SEQUENCES: 19 (iv) CORRESPONDENCE ADDRESS:

(A) ADDRESSEE: SMART & BIGGAR

(B) STREET: P.O. BOX 2999, STATION D
(C) CITY: OTTAWA

(D) STATE: ONT

(E) COUNTRY: CANADA
(F) ZIP: K1P 5Y6 (v) COMPUTER READABLE FORM:

(A) MEDIUM TYPE: Floppy disk (B) COMPUTER: IBM PC compatible (C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE;. ASCII (text) (vi) CURRENT APPLICATION DATA:

(A) APPLICATION NUMBER: CA 2,274,803 (B) FILING DATE: 22-DEC-1997 (C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA:

(A) APPLICATION NUMBER: US 60/059,978 (B) FILING DATE: 23--DEC-1996 (vii) PRIOR APPLICATION DATA:

(A) APPLICATION NUMBER: US 08/813,509 (B) FILING DATE: 07-MAR-1997 (vii) PRIOR APPLICATION DATA:

(A) APPLICATION NUMBER: US 60/064,671 (B) FILING DATE: 14-OCT-1997 (viii) ATTORNEY/AGENT INFORMATION:

(A) NAME: SMART & BIGGAR

(B) REGISTRATION NUMBER:

(C) REFERENCE/DOCKET NUMBER: 72249-88 (ix) TELECOMMUNICATION INFORMATION:

(A) TELEPHONE: (6131-232-2486 (B) TELEFAX: (613)-232-8440 (2) INFORMATION FOR SEQ ID NO::i:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 3115 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single 37a (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA
(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO

(vi) ORIGINAL SOURCE:
(A) ORGANISM: HOMO SAPIENS
(vii) IMMEDIATE SOURCE:
(A) LIBRARY: BONE-MARROW DERIVED DENDRITIC CELLS
(B) CLONE: 9D-8A

(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 93..1868 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1:

Val Ala Leu Gln Ile Ala Pro Pro Cys Thr Ser Glu Lys His Tyr Glu His Leu Gly Arg Cys Cys Asn Lys Cys Glu Pro Gly Lys Tyr Met Ser Ser Lys Cys Thr Thr Thr Ser Asp Ser Val Cys Leu Pro Cys Gly Pro Asp Glu Tyr Leu Asp Ser Trp Asn Glu Glu Asp Lys Cys Leu Leu His Lys Val Cys Asp Thr Gly Lys Ala Leu Val Ala Val Val Ala Gly Asn Ser Thr Thr Pro Arg Arg Cys Ala Cys Thr Ala Gly Tyr His Trp Ser Gln Asp Cys Glu Cys Cys Arg Arg Asn Thr Glu Cys Ala Pro Gly Leu Gly Ala Gln His Pro Leu Gln Leu Asn Lys Asp Thr Val Cys Lys Pro Cys Leu Ala Gly Tyr Phe Ser Asp Ala Phe Ser Ser Thr Asp Lys Cys Arg Pro Trp Thr Asn Cys Thr Phe Leu Gly Lys Arg Val Glu His His Gly Thr Glu Lys Ser Asp Ala Val Cys Ser Ser Ser Leu Pro Ala Arg Lys Pro Pro Asn Glu Pro His Val Tyr Leu Pro Gly Leu Ile Ile Leu Leu Leu Phe Ala Ser Val Ala Leu Val Ala Ala Ile Ile Phe Gly Val Cys Tyr Arg Lys Lys Gly Lys Ala Leu Thr Ala Asn Leu Trp His Trp Ile Asn Glu Ala Cys Gly Arg Leu Ser Gly Asp Lys Glu Ser Ser Gly Asp Ser Cys Val Ser Thr His Thr Ala Asn Phe Gly Gln Gln Gly Ala Cys Glu Gly Val Leu Leu Leu Thr Leu Glu Glu Lys Thr Phe Pro Glu Asp Met Cys Tyr Pro Asp Gln Gly Gly Val Cys Gln Gly Thr Cys Val Gly Gly Gly Pro Tyr Ala Gln Gly Glu Asp Ala Arg Met Leu Ser Leu Val Ser Lys Thr Glu Ile Glu Glu Asp Ser Phe Arg Gln Met Pro Thr Glu Asp Glu Tyr Met Asp Arg Pro Ser Gln Pro Thr Asp Gin Leu Leu Phe Leu Thr Glu Pro Gly Ser Lys Ser Thr Pro Pro Phe Ser Glu Pro Leu Glu Val Gly Glu Asn Asp Ser Leu Ser Gln Cys Phe Thr Gly Thr Gln Ser Thr Val Gly Ser Glu Ser Cys Asn Cys Thr Glu Pro Leu Cys Arg Thr Asp Trp Thr Pro Met Ser Ser Glu Asn Tyr Leu Gln Lys Glu Val Asp Ser Gly His Cys Pro His Trp Ala Ala Ser Pro Ser Pro Asn Trp Ala Asp Val Cys Thr Gly Cys Arg Asn Pro Pro Gly Glu Asp Cys Glu Pro Leu Val Gly Ser Pro Lys Arg Gly Pro Leu Pro Gln Cys Ala Tyr Gly Met Gly Leu Pro Pro Glu Glu Glu Ala Ser Arg Thr Glu Ala Arg Asp Gln Pro Glu Asp Gly Ala Asp Gly Arg Leu Pro Ser Ser Ala Arg Ala Gly Ala Gly Ser Gly Ser Ser Pro Gly Gly Gln Ser Pro Ala Ser Gly Asn Val Thr Gly Asn Ser Asn Ser Thr Phe Ile Ser Ser Gly Gln Val Met Asn Phe Lys Gly Asp Ile Ile Val Val Tyr Val Ser Gln Thr Ser Gln Glu Gly Ala Ala Ala Ala Ala Glu Pro Met Gly Arg Pro Val Gln Glu Glu Thr Leu Ala Arg Arg Asp Ser Phe Ala Gly Asn Gly Pro Arg Phe Pro Asp Pro Cys Gly Gly Pro Glu Gly Leu Arg Glu Pro Glu Lys Ala Ser Arg Pro Val Gln Glu Gln Gly Gly Ala Lys Ala (2) INFORMATION FOR SEQ ID NO:2:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 591 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:

Val Ala Leu Gln Ile Ala Pro Pro Cys Thr Ser Glu Lys His Tyr Glu His Leu Gly Arg Cys Cys Asn Lys Cys Glu Pro Gly Lys Tyr Met Ser Ser Lys Cys Thr Thr Thr Ser Asp Ser Val Cys Leu Pro Cys Gly Pro Asp Glu Tyr Leu Asp Ser Trp Asn Glu Glu Asp Lys Cys Leu Leu His Lys Val Cys Asp Thr Gly Lys Ala Leu Val Ala Val Val Ala Gly Asn Ser Thr Thr Pro Arg Arg Cys Ala Cys Thr Ala Gly Tyr His Trp Ser Gln Asp Cys Glu Cys Cys Arg Arg Asn Thr Glu Cys Ala Pro Gly Leu Gly Ala Gln His Pro Leu Gln Leu Asn Lys Asp Thr Val Cys Lys Pro Cys Leu Ala Gly Tyr Phe Ser Asp Ala Phe Ser Ser Thr Asp Lys Cys Arg Pro Trp Thr Asn Cys Thr Phe Leu Gly Lys Arg Val Glu His His Gly Thr Glu Lys Ser Asp Ala Val Cys Ser Ser Ser Leu Pro Ala Arg Lys Pro Pro Asn Glu Pro His Val Tyr Leu Pro Gly Leu Ile Ile Leu Leu Leu Phe Ala Ser Val Ala Leu Val Ala Ala Ile Ile Phe Gly Val Cys Tyr Arg Lys Lys Gly Lys Ala Leu Thr Ala Asn Leu Trp His Trp Ile Asn Glu Ala Cys Gly Arg Leu Ser Gly Asp Lys Glu Ser Ser Gly Asp Ser Cys Val Ser Thr His Thr Ala Asn Phe Gly Gln Gln Gly Ala Cys Glu Gly Val Leu Leu Leu Thr Leu Glu Glu Lys Thr Phe Pro Glu Asp Met Cys Tyr Pro Asp Gln Gly Gly Val Cys Gln Gly Thr Cys Val Gly Gly Gly Pro Tyr Ala Gln Gly Glu Asp Ala Arg Met Leu Ser Leu Val Ser Lys Thr Glu Ile Glu Glu Asp Ser Phe Arg Gln Met Pro Thr Glu Asp Glu Tyr Met Asp Arg Pro Ser Gln Pro Thr Asp Gln Leu Leu Phe Leu Thr Glu Pro Gly Ser Lys Ser Thr Pro Pro Phe Ser Glu Pro Leu Glu Val Gly Glu Asn Asp Ser Leu Ser Gln Cys Phe Thr Gly Thr Gln Ser Thr Val Gly Ser Glu Ser Cys Asn Cys Thr Glu Pro Leu Cys Arg Thr Asp Trp Thr Pro Met Ser Ser Glu Asn Tyr Leu Gln Lys Glu Val Asp Ser Gly His Cys Pro His Trp Ala Ala Ser Pro Ser Pro Asn Trp Ala Asp Val Cys Thr Gly Cys Arg Asn Pro Pro Gly Glu Asp Cys Glu Pro Leu Val Gly Ser Pro Lys Arg Gly Pro Leu Pro Gln Cys Ala Tyr Gly Met Gly Leu Pro Pro Glu Glu Glu Ala Ser Arg Thr Glu Ala Arg Asp Gln Pro Glu Asp Gly Ala Asp Gly Arg Leu Pro Ser Ser Ala Arg Ala Gly Ala Gly Ser Gly Ser Ser Pro Gly Gly Gln Ser Pro Ala Ser Gly Asn Val Thr Gly Asn Ser Asn Ser Thr Phe Ile Ser Ser Gly Gln Val Met Asn Phe Lys Gly Asp Ile Ile Val Val Tyr Val Ser Gln Thr Ser Gln Glu Gly Ala Ala Ala Ala Ala Glu Pro Met Gly Arg Pro Val Gln Glu Glu Thr Leu Ala Arg Arg Asp Ser Phe Ala Gly Asn Gly Pro Arg Phe Pro Asp Pro Cys Gly Gly Pro Glu Gly Leu Arg Glu Pro Glu Lys Ala Ser Arg Pro Val Gln Glu Gln Gly Gly Ala Lys Ala (2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1391 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA

(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: HOMO SAPIENS
(vii) IMMEDIATE SOURCE:
(A) LIBRARY: BONE-MARROW DERIVED DENDRITIC CELLS
(B) CLONE: 9D-15C

(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 39..1391 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:

Met Ala Pro Arg Ala Arg Arg Arg Arg Pro Leu Phe Ala Leu Leu Leu Leu Cys Ala Leu Leu Ala Arg Leu Gln Val Ala Leu Gln Ile Ala Pro Pro Cys Thr Ser Glu Lys His Tyr Glu His Leu Gly Arg Cys Cys Asn Lys Cys Glu Pro Gly Lys Tyr Met Ser Ser Lys Cys Thr Thr Thr Ser Asp Ser Val Cys Leu Pro Cys Gly Pro Asp Glu Tyr Leu Asp Ser Trp Asn Glu Glu Asp Lys Cys Leu Leu His Lys Val Cys Asp Thr Gly Lys Ala Leu Val Ala Val Val Ala Gly Asn Ser Thr Thr Pro Arg Arg Cys Ala Cys Thr Ala Gly Tyr His Trp Ser Gln Asp Cys Glu Cys Cys Arg Arg Asn Thr Glu Cys Ala Pro Gly Leu Gly Ala Gln His Pro Leu Gln Leu Asn Lys Asp Thr Val Cys Lys Pro Cys Leu Ala Gly Tyr Phe Ser Asp Ala Phe Ser Ser Thr Asp Lys Cys Arg Pro Trp Thr Asn Cys Thr Phe Leu Gly Lys Arg Val Glu His His Gly Thr Glu Lys Ser Asp Ala Val Cys Ser Ser Ser Leu Pro Ala Arg Lys Pro Pro Asn Glu Pro His Val Tyr Leu Pro Gly Leu Ile Ile Leu Leu Leu Phe Ala Ser Val Ala Leu Val Ala Ala Ile Ile Phe Gly Val Cys Tyr Arg Lys Lys Gly Lys Ala Leu Thr Ala Asn Leu Trp His Trp Ile Asn Glu Ala Cys Gly Arg Leu Ser Gly Asp Lys Glu Ser Ser Gly Asp Ser Cys Val Ser Thr His Thr Ala Asn Phe Gly Gln Gln Gly Ala Cys Glu Gly Val Leu Leu Leu Thr Leu Glu Glu Lys Thr Phe Pro Glu Asp Met Cys Tyr Pro Asp Gln Gly Gly Val Cys Gln Gly Thr Cys Val Gly Gly Gly Pro Tyr Ala Gln Gly Glu Asp Ala Arg Met Leu Ser Leu Val Ser Lys Thr Glu Ile Glu Glu Asp Ser Phe Arg Gln Met Pro Thr Glu Asp Glu Tyr Met Asp Arg Pro Ser Gln Pro Thr Asp Gin Leu Leu Phe Leu Thr Glu Pro Gly Ser Lys Ser Thr Pro Pro Phe Ser Glu Pro Leu Glu Val Gly Glu Asn Asp Ser Leu Ser Gln Cys Phe Thr Gly Thr Gln Ser Thr Val Gly Ser Glu Ser Cys Asn Cys Thr Glu Pro Leu Cys Arg Thr Asp Trp Thr Pro Met Ser Ser Glu Asn Tyr Leu Gin Lys Glu Val Asp Ser Gly His Cys Pro His Trp Ala Ala Ser *rB

Pro Ser Pro Asn Trp Ala Asp Val Cys Thr Gly Cys Arg Asn (2) INFORMATION FOR SEQ ID NO:4:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 451 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:

Met Ala Pro Arg Ala Arg Arg Arg Arg Pro Leu Phe Ala Leu Leu Leu Leu Cys Ala Leu Leu Ala Arg Leu Gln Val Ala Leu Gln Ile Ala Pro Pro Cys Thr Ser Glu Lys His Tyr Glu His Leu Gly Arg Cys Cys Asn Lys Cys Glu Pro Gly Lys Tyr Met Ser Ser Lys Cys Thr Thr Thr Ser Asp Ser Val Cys Leu Pro Cys Gly Pro Asp Glu Tyr Leu Asp Ser Trp Asn Glu Glu Asp Lys Cys Leu Leu His Lys Val Cys Asp Thr Gly Lys Ala Leu Val Ala Val Val Ala Gly Asn Ser Thr Thr Pro Arg Arg Cys Ala Cys Thr Ala Gly Tyr His Trp Ser Gln Asp Cys Glu Cys Cys Arg Arg Asn Thr Glu Cys Ala Pro Gly Leu Gly Ala Gln His Pro Leu Gln Leu Asn Lys Asp Thr Val Cys Lys Pro Cys Leu Ala Gly Tyr Phe Ser Asp Ala Phe Ser Ser Thr Asp Lys Cys Arg Pro Trp Thr Asn Cys Thr Phe Leu Gly Lys Arg Val Glu His His Gly Thr Glu Lys Ser Asp Ala Val Cys Ser Ser Ser Leu Pro Ala Arg Lys Pro Pro Asn Glu Pro His Val Tyr Leu Pro Gly Leu Ile Ile Leu Leu Leu Phe Ala Ser Val Ala Leu Val Ala Ala Ile Ile Phe Gly Val Cys Tyr Arg Lys Lys Gly Lys Ala Leu Thr Ala Asn Leu Trp His Trp Ile Asn Glu Ala Cys Gly Arg Leu Ser Gly Asp Lys Glu Ser Ser Gly Asp Ser Cys Val Ser Thr His Thr Ala Asn Phe Gly Gln Gln Gly Ala Cys Glu Gly Val Leu Leu Leu Thr Leu Glu Glu Lys Thr Phe Pro Glu Asp Met Cys Tyr Pro Asp Gln Gly Gly Val Cys Gln Gly Thr Cys Val Gly Gly Gly Pro Tyr Ala Gln Gly Glu Asp Ala Arg Met Leu Ser Leu Val Ser Lys Thr Glu Ile Glu Glu Asp Ser Phe Arg Gln Met Pro Thr Glu Asp Glu Tyr Met Asp Arg Pro Ser Gln Pro Thr Asp Gln Leu Leu Phe Leu Thr Glu Pro Gly Ser Lys Ser Thr Pro Pro Phe Ser Glu Pro Leu Glu Val Gly Glu Asn Asp Ser Leu Ser Gln Cys Phe Thr Gly Thr Gln Ser Thr Val Gly Ser Glu Ser Cys Asn Cys Thr Glu Pro Leu Cys Arg Thr Asp Trp Thr Pro Met Ser Ser Glu Asn Tyr Leu Gln Lys Glu Val Asp Ser Gly His Cys Pro His Trp Ala Ala Ser Pro Ser Pro Asn Trp Ala Asp Val Cys Thr Gly Cys Arg Asn (2) INFORMATION FOR SEQ ID NO:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 3136 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA

(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: HOMO SAPIENS
(vii) IMMEDIATE SOURCE:

(A) LIBRARY: BONE-MARROW DERIVED DENDRITIC CELLS
(B) CLONE: FULL LENGTH RANK
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 39..1886 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:5:

Met Ala Pro Arg Ala Arg Arg Arg Arg Pro Leu Phe Ala Leu Leu Leu Leu Cys Ala Leu Leu Ala Arg Leu Gin Val Ala Leu Gln Ile Ala Pro Pro Cys Thr Ser Glu Lys His Tyr Glu His Leu Gly Arg Cys Cys Asn Lys Cys Glu Pro Gly Lys Tyr Met Ser Ser Lys Cys Thr Thr Thr Ser Asp Ser Val Cys Leu Pro Cys Gly Pro Asp Glu Tyr Leu Asp Ser Trp Asn Glu Glu Asp Lys Cys Leu Leu His Lys Val Cys Asp Thr Gly Lys Ala Leu Val Ala Val Val Ala Gly Asn Ser Thr Thr Pro Arg Arg Cys Ala Cys Thr Ala Gly Tyr His Trp Ser Gln Asp Cys Glu Cys Cys Arg Arg Asn Thr Glu Cys Ala Pro Gly Leu Gly Ala Gln His Pro Leu Gln Leu Asn Lys Asp Thr Val Cys Lys Pro Cys Leu Ala Gly Tyr Phe Ser Asp Ala Phe Ser Ser Thr Asp Lys Cys Arg Pro Trp Thr Asn Cys Thr Phe Leu Gly Lys Arg Val Glu His His Gly Thr Glu Lys Ser Asp Ala Val Cys Ser Ser Ser Leu Pro Ala Arg Lys Pro Pro Asn Glu Pro His Val Tyr Leu Pro Gly 200 205 ' 210 Leu Ile Ile Leu Leu Leu Phe Ala Ser Val Ala Leu Val Ala Ala Ile Ile Phe Gly Val Cys Tyr Arg Lys Lys Gly Lys Ala Leu Thr Ala Asn Leu Trp His Trp Ile Asn Glu Ala Cys Gly Arg Leu Ser Gly Asp Lys Glu Ser Ser Gly Asp Ser Cys Val Ser Thr His Thr Ala Asn Phe Gly Gln Gln Gly Ala Cys Glu Gly Val Leu Leu Leu Thr Leu Glu Glu Lys Thr Phe Pro Glu Asp Met Cys Tyr Pro Asp Gln Gly Gly Val Cys Gln Gly Thr Cys Val Gly Gly Gly Pro Tyr Ala Gln Gly Glu Asp Ala Arg Met Leu Ser Leu Val Ser Lys Thr Glu Ile Glu Glu Asp Ser Phe Arg Gln Met Pro Thr Glu Asp Glu Tyr Met Asp Arg Pro Ser Gln Pro Thr Asp Gln Leu Leu Phe Leu Thr Glu Pro Gly Ser Lys Ser Thr Pro Pro Phe Ser Glu Pro Leu Glu Val Gly Glu Asn Asp Ser Leu Ser Gln Cys Phe Thr Gly Thr Gln Ser Thr Val Gly Ser Glu Ser Cys Asn Cys Thr Glu Pro Leu Cys Arg Thr Asp Trp Thr Pro Met Ser Ser Glu Asn Tyr Leu Gln Lys Glu Val Asp Ser Gly His Cys Pro His Trp Ala Ala Ser Pro Ser Pro Asn Trp Ala Asp Val Cys Thr Gly Cys Arg Asn Pro Pro Gly Glu Asp Cys Glu Pro Leu Val Gly Ser Pro Lys Arg Gly Pro Leu Pro Gln Cys Ala Tyr Gly Met Gly Leu Pro Pro Glu Glu Glu Ala Ser Arg Thr Glu Ala Arg Asp Gln Pro Glu Asp Gly Ala Asp Gly Arg Leu Pro Ser Ser Ala Arg Ala Gly Ala Gly Ser Gly Ser Ser Pro Gly Gly Gln Ser Pro Ala Ser Gly Asn Val Thr Gly Asn Ser Asn Ser Thr Phe Ile Ser Ser Gly Gln Val Met Asn Phe Lys Gly Asp Ile Ile Val Val Tyr Val Ser Gln Thr Ser Gln Glu Gly Ala Ala Ala Ala Ala Glu Pro Met Gly Arg Pro Val Gln Glu Glu Thr Leu Ala Arg Arg Asp Ser Phe Ala Gly Asn Gly Pro Arg Phe Pro Asp Pro Cys Gly Gly Pro Glu Gly Leu Arg Glu Pro Glu Lys Ala Ser Arg Pro Val Gln Glu Gln Gly Gly Ala Lys Ala (2) INFORMATION FOR SEQ ID NO:6:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 616 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:6:

Met Ala Pro Arg Ala Arg Arg Arg Arg Pro Leu Phe Ala Leu Leu Leu Leu Cys Ala Leu Leu Ala Arg Leu Gln Val Ala Leu Gln Ile Ala Pro Pro Cys Thr Ser Glu Lys His Tyr Glu His Leu Gly Arg Cys Cys An Lys Cys Glu Pro Gly Lys Tyr Met Ser Ser Lys Cys Thr Thr Thr Ser Asp Ser Val Cys Leu Pro Cys Gly Pro Asp Glu Tyr Leu Asp Ser Trp Asn Glu Glu Asp Lys Cys Leu Leu His Lys Val Cys Asp Thr Gly Lys Ala Leu Val Ala Val Val Ala Gly Asn Ser Thr Thr Pro Arg Arg Cys Ala Cys Thr Ala Gly Tyr His Trp Ser Gln Asp Cys Glu Cys Cys Arg Arg Asn Thr Glu Cys Ala Pro Gly Leu Gly Ala Gln His Pro Leu Gln Leu Asn Lys Asp Thr Val Cys Lys Pro Cys Leu Ala Gly Tyr Phe Ser Asp Ala Phe Ser Ser Thr Asp Lys Cys Arg Pro Trp Thr Asn Cys Thr Phe Leu Gly Lys Arg Val Glu His His Gly Thr Glu Lys Ser Asp Ala Val Cys Ser Ser Ser Leu Pro Ala Arg Lys Pro Pro Asn Glu Pro His Val Tyr Leu Pro Gly Leu Ile Ile Leu Leu Leu Phe Ala Ser Val Ala Leu Val Ala Ala Ile Ile Phe Gly Val Cys Tyr Arg Lys Lys Gly Lys Ala Leu Thr Ala Asn Leu Trp His Trp Ile Asn Glu Ala Cys Gly Arg Leu Ser Gly Asp Lys Glu Ser Ser Gly Asp Ser Cys Val Ser Thr His Thr Ala Asn Phe Gly Gln Gln Gly Ala Cys Glu Gly Val Leu Leu Leu Thr Leu Glu Glu Lys Thr Phe Pro Glu Asp Met Cys Tyr Pro Asp Gln Gly Gly Val Cys Gln Gly Thr Cys Val Gly Gly Gly Pro Tyr Ala Gln Gly Glu Asp Ala Arg Met Leu Ser Leu Val Ser Lys Thr Glu Ile Glu Glu Asp Ser Phe Arg Gln Met Pro Thr Glu Asp Glu Tyr Met Asp Arg Pro Ser Gln Pro Thr Asp Gln Leu Leu Phe Leu Thr Glu Pro Gly Ser Lys Ser Thr Pro Pro Phe Ser Glu Pro Leu Glu Val Gly Glu Asn Asp Ser Leu Ser Gln Cys Phe Thr Gly Thr Gln Ser Thr Val Gly Ser Glu Ser Cys Asn Cys Thr Glu Pro Leu Cys Arg Thr Asp Trp Thr Pro Met Ser Ser Glu Asn Tyr Leu Gln Lys Glu Val Asp Ser Gly His Cys Pro His Trp Ala Ala Ser Pro Ser Pro Asn Trp Ala Asp Val Cys Thr Gly Cys Arg Asn Pro Pro Gly Glu Asp Cys Glu Pro Leu Val Gly Ser Pro Lys Arg Gly Pro Leu Pro Gln Cys Ala Tyr Gly Met Gly Leu Pro Pro Glu Glu Glu Ala Ser Arg Thr Glu Ala Arg Asp Gln Pro Glu Asp Gly Ala Asp Gly Arg Leu Pro Ser Ser Ala Arg Ala Gly Ala Gly Ser Gly Ser Ser Pro Gly Gly Gln Ser Pro Ala Ser Gly Asn Val Thr Gly Asn Ser Asn Ser Thr Phe Ile Ser Ser Gly Gln Val Met Asn Phe Lys Gly Asp Ile Ile Val Val Tyr Val Ser Gln Thr Ser Gln Glu Gly Ala Ala Ala Ala Ala Glu Pro Met Gly Arg Pro Val Gln Glu Glu Thr Leu Ala Arg Arg Asp Ser Phe Ala Gly Asn Gly Pro Arg Phe Pro Asp Pro Cys Gly Gly Pro Glu Gly Leu Arg Glu Pro Glu Lys Ala Ser Arg Pro Val Gln Glu Gln Gly Gly Ala Lys Ala (2) INFORMATION FOR SEQ ID NO:7:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 8 amino acids (B) TYPE: amino acid (C) STRANDEDNESS: not relevant (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (vii) IMMEDIATE SOURCE:
(B) CLONE: FLAG peptide (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7:
Asp Tyr Lys Asp Asp Asp Asp Lys (2) INFORMATION FOR SEQ ID NO:8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 232 amino acids (B) TYPE: amino acid *rB

(C) STRANDEDNESS: not relevant (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (vi) ORIGINAL SOURCE:
(A) ORGANISM: Human (vii) IMMEDIATE SOURCE:
(B) CLONE: IgOl Fc mutein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:8:

Glu Pro Arg Ser Cys Asp Lys Thr His Thr Cys Pro Pro Cys Pro Ala Pro Glu Ala Glu Gly Ala Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val Lys Phe Asn Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Tyr Asn Ser Thr Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn Gly Lys Asp Tyr Lys Cys Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Met Gln Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln Val Tyr Thr Leu Pro Pro Ser Arg Asp Glu Leu Thr Lys Asn Gln Val Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Arg His Ile Ala Val Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp Gln Gln Gly Asn Val Phe Ser Cys Ser Val Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu Ser Pro Gly Lys (2) INFORMATION FOR SEQ ID NO:9:

(1) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 31 amino acids (B) TYPE: amino acid (C) STRANDEDNESS: not relevant (D) TOPOLOGY: linear (ii) MOLECULE TYPE: peptide (iii) HYPOTHETICAL: NO

(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: CMV (R2780 Leader) (ix) FEATURE:
(D) OTHER INFORMATION: Metl-Arg28 is the actual leader peptide; Arg29 strengthens the furin cleavage site;
nucleotides encoding Thr30 and Ser3l add a Spel site.

(xi) SEQUENCE DESCRIPTION: SEQ ID NO:9:

Met Ala Arg Arg Leu Trp Ile Leu Ser Leu Leu Ala Val Thr Leu Thr Val Ala Leu Ala Ala Pro Ser Gln Lys Ser Lys Arg Arg Thr Ser (2) INFORMATION FOR SEQ ID NO:10:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1630 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA

(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Mus musculus (vii) IMMEDIATE SOURCE:
(A) LIBRARY:
(B) CLONE: RANKL
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 3..884 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:10:

Gly Val Pro His Glu Gly Pro Leu His Pro Ala Pro Ser Ala Pro Ala Pro Ala Pro Pro Pro Ala Ala Ser Arg Ser Met Phe Leu Ala Leu Leu Gly Leu Gly Leu Gly Gln Val Val Cys Ser Ile Ala Leu Phe Leu Tyr Phe Arg Ala Gln Met Asp Pro Asn Arg Ile Ser Glu Asp Ser Thr His Cys Phe Tyr Arg Ile Leu Arg Leu His Glu Asn Ala Asp Leu Gln Asp Ser Thr Leu Glu Ser Glu Asp Thr Leu Pro Asp Ser Cys Arg Arg Met Lys Gln Ala Phe Gln Gly Ala Val Gln Lys Glu Leu Gln His Ile Val Gly Pro Gln Arg Phe Ser Gly Ala Pro Ala Met Met Glu Gly Ser Trp Leu Asp Val Ala Gln Arg Gly Lys Pro Glu Ala Gln Pro Phe Ala His Leu Thr Ile Asn Ala Ala Ser Ile Pro Ser Gly Ser His Lys Val Thr Leu Ser Ser Trp Tyr His Asp Arg Gly Trp Ala Lys Ile Ser Asn Met Thr Leu Ser Asn Gly Lys Leu Arg Val Asn Gln Asp Gly Phe Tyr Tyr Leu Tyr Ala Asn Ile Cys Phe Arg His His Glu Thr Ser Gly Ser Val Pro Thr Asp Tyr Leu Gln Leu Met Val Tyr Val Val Lys Thr Ser Ile Lys Ile Pro Ser Ser His Asn Leu Met Lys Gly Gly Ser Thr Lys Asn Trp Ser Gly Asn Ser Glu Phe His Phe Tyr Ser Ile Asn Val Gly Gly Phe Phe Lys Leu Arg Ala Gly Glu Glu Ile Ser Ile Gln Val Ser Asn Pro Ser Leu Leu Asp Pro Asp Gln Asp Ala Thr Tyr Phe Gly Ala Phe Lys Val Gln Asp Ile Asp (2) INFORMATION FOR SEQ ID NO:11:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 294 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:11:

Gly Val Pro His Glu Gly Pro Leu His Pro Ala Pro Ser Ala Pro Ala Pro Ala Pro Pro Pro Ala Ala Ser Arg Ser Met Phe Leu Ala Leu Leu Gly Leu Gly Leu Gly Gln Val Val Cys Ser Ile Ala Leu Phe Leu Tyr Phe Arg Ala Gln Met Asp Pro Asn Arg Ile Ser Glu Asp Ser Thr His Cys Phe Tyr Arg Ile Leu Arg Leu His Glu Asn Ala Asp Leu Gln Asp Ser Thr Leu Glu Ser Glu Asp Thr Leu Pro Asp Ser Cys Arg Arg Met Lys Gln Ala Phe Gln Gly Ala Val Gln Lys Glu Leu Gln His Ile Val Gly Pro Gln Arg Phe Ser Gly Ala Pro Ala Met Met Glu Gly Ser Trp Leu Asp Val Ala Gln Arg Gly Lys Pro Glu Ala Gln Pro Phe Ala His Leu Thr Ile Asn Ala Ala Ser Ile Pro Ser Gly Ser His Lys Val Thr Leu Ser Ser Trp Tyr His Asp Arg Gly Trp Ala Lys Ile Ser Asn Met Thr Leu Ser Asn Gly Lys Leu Arg Val Asn Gln Asp Gly Phe Tyr Tyr Leu Tyr Ala Asn Ile Cys Phe Arg His His Glu Thr Ser Gly Ser Val Pro Thr Asp Tyr Leu Gln Leu Met Val Tyr Val Val Lys Thr Ser Ile Lys Ile Pro Ser Ser His Asn Leu Met Lys Gly Gly Ser Thr Lys Asn Trp Ser Gly Asn Ser Glu Phe His Phe Tyr Ser Ile Asn Val Gly Gly Phe Phe Lys Leu Arg Ala Gly Glu Glu Ile Ser Ile Gln Val Ser Asn Pro Ser Leu Leu Asp Pro Asp Gln Asp Ala Thr Tyr Phe Gly Ala Phe Lys Val Gln Asp Ile Asp (2) INFORMATION FOR SEQ ID NO:12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 954 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA

(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:

(A) ORGANISM: Homo sapiens (vii) IMMEDIATE SOURCE:
(A) LIBRARY:
(B) CLONE: huRANKL (full length) (ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..951 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12:

Met Arg Arg Ala Ser Arg Asp Tyr Thr Lys Tyr Leu Arg Gly Ser Glu Glu Met Gly Gly Gly Pro Gly Ala Pro His Glu Gly Pro Leu His Ala Pro Pro Pro Pro Ala Pro His Gln Pro Pro Ala Ala Ser Arg Ser Met Phe Val Ala Leu Leu Gly Leu Gly Leu Gly Gln Val Val Cys Ser Val Ala Leu Phe Phe Tyr Phe Arg Ala Gln Met Asp Pro Asn Arg Ile Ser Glu Asp Gly Thr His Cys Ile Tyr Arg Ile Leu Arg Leu His Glu Asn Ala Asp Phe Gln Asp Thr Thr Leu Glu Ser Gln Asp Thr Lys Leu Ile Pro Asp Ser Cys Arg Arg Ile Lys Gln Ala Phe Gln Gly Ala Val Gln Lys Glu Leu Gln His Ile Val Gly Ser Gln His Ile Arg Ala Glu Lys Ala Met Val Asp Gly Ser Trp Leu Asp Leu Ala Lys Arg Ser Lys Leu Glu Ala Gln Pro Phe Ala His Leu Thr Ile Asn Ala Thr Asp Ile Pro Ser Gly Ser His Lys Val Ser Leu Ser Ser Trp Tyr His Asp Arg Gly Trp Ala Lys Ile Ser Asn Met Thr Phe Ser Asn Gly Lys Leu Ile Val Asn Gln Asp Gly Phe Tyr Tyr Leu Tyr Ala Asn Ile Cys Phe Arg His His Glu Thr Ser Gly Asp Leu Ala Thr Glu Tyr Leu Gln Leu Met Val Tyr Val Thr Lys Thr Ser Ile Lys Ile Pro Ser Ser His Thr Leu Met Lys Gly Gly Ser Thr Lys Tyr Trp Ser Gly Asn Ser Glu Phe His Phe Tyr Ser Ile Asn Val Gly Gly Phe Phe Lys Leu Arg Ser Gly Glu Glu Ile Ser Ile Glu Val Ser Asn Pro Ser Leu Leu Asp Pro Asp Gln Asp Ala Thr Tyr Phe Gly Ala Phe Lys Val Arg Asp Ile Asp (2) INFORMATION FOR SEQ ID NO:13:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 317 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:13:

Met Arg Arg Ala Ser Arg Asp Tyr Thr Lys Tyr Leu Arg Gly Ser Glu Glu Met Gly Gly Gly Pro Gly Ala Pro His Glu Gly Pro Leu His Ala Pro Pro Pro Pro Ala Pro His Gln Pro Pro Ala Ala Ser Arg Ser Met Phe Val Ala Leu Leu Gly Leu Gly Leu Gly Gln Val Val Cys Ser Val Ala Leu Phe Phe Tyr Phe Arg Ala Gln Met Asp Pro Asn Arg Ile Ser Glu Asp Gly Thr His Cys Ile Tyr Arg Ile Leu Arg Leu His Glu Asn Ala Asp Phe Gln Asp Thr Thr Leu Glu Ser Gln Asp Thr Lys Leu Ile Pro Asp Ser Cys Arg Arg Ile Lys Gln Ala Phe Gln Gly Ala Val Gln Lys Glu Leu Gln His Ile Val Gly Ser Gln His Ile Arg Ala Glu Lys Ala Met Val Asp Gly Ser Trp Leu Asp Leu Ala Lys Arg Ser Lys Leu Glu Ala Gln Pro Phe Ala His Leu Thr Ile Asn Ala Thr Asp Ile Pro Ser Gly Ser His Lys Val Ser Leu Ser Ser Trp Tyr His Asp Arg Gly Trp Ala Lys Ile Ser Asn Met Thr Phe Ser Asn Gly Lys Leu Ile Val Asn Gln Asp Gly Phe Tyr Tyr Leu Tyr Ala Asn Ile Cys Phe Arg His His Glu Thr Ser Gly Asp Leu Ala Thr Glu Tyr Leu Gln Leu Met Val Tyr Val Thr Lys Thr Ser Ile Lys Ile Pro Ser Ser His Thr Leu Met Lys Gly Gly Ser Thr Lys Tyr Trp Ser Gly Asn Ser Glu Phe His Phe Tyr Ser Ile Asn Val Gly Gly Phe Phe Lys Leu Arg Ser Gly Glu Glu Ile Ser Ile Glu Val Ser Asn Pro Ser Leu Leu Asp Pro Asp Gln Asp Ala Thr Tyr Phe Gly Ala Phe Lys Val Arg Asp Ile Asp (2) INFORMATION FOR SEQ ID NO:14:

(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1878 base pairs (B) TYPE: nucleic acid (C) STRANDEDNESS: single (D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA

(iii) HYPOTHETICAL: NO
(iv) ANTI-SENSE: NO
(vi) ORIGINAL SOURCE:
(A) ORGANISM: Murine (vii) IMMEDIATE SOURCE:
(A) LIBRARY: Murine Fetal Liver Epithelium (B) CLONE: muRANK

(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..1875 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:14:

Net Ala Pro Arg Ala Arg Arg Arg Arg Gin Leu Pro Ala Pro Leu Leu Ala Leu Cys Val Leu Leu Val Pro Leu Gin Val Thr Leu Gin Val Thr Pro Pro Cys Thr Gin Glu Arg His Tyr Glu His Leu Gly Arg Cys Cys Ser Arg Cys Glu Pro Gly Lys Tyr Leu Ser Ser Lys Cys Thr Pro Thr Ser Asp Ser Val Cys Leu Pro Cys Gly Pro Asp Glu Tyr Leu Asp Thr Trp Asn Glu Glu Asp Lys Cys Leu Leu His Lys Val Cys Asp Ala Gly Lys Ala Leu Val Ala Val Asp Pro Gly Asn His Thr Ala Pro Arg Arg Cys Ala Cys Thr Ala Gly Tyr His Trp Asn Ser Asp Cys Glu Cys Cys Arg Arg Asn Thr Glu Cys Ala Pro Gly Phe Gly Ala Gin His Pro Leu Gin Leu Asn Lys Asp Thr Val Cys Thr Pro Cys Leu Leu Gly Phe Phe Ser Asp Val Phe Ser Ser Thr Asp Lys Cys Lys Pro Trp Thr Asn Cys Thr Leu Leu Gly Lys Leu Glu Ala His Gin Gly Thr Thr Glu Ser Asp Val Val Cys Ser Ser Ser Met Thr Leu Arg Arg Pro Pro Lys Glu Ala Gln Ala Tyr Leu Pro Ser Leu Ile Val Leu Leu Leu Phe Ile Ser Val Val Val Val Ala Ala Ile Ile Phe Gly Val Tyr Tyr Arg Lys Gly Gly Lys Ala Leu Thr Ala Asn Leu Trp Asn Trp Val Asn Asp Ala Cys Ser Ser Leu Ser Gly Asn Lys Glu Ser Ser Gly Asp Arg Cys Ala Gly Ser His Ser Ala Thr Ser Ser Gln Gln Glu Val Cys Glu Gly Ile Leu Leu Met Thr Arg Glu Glu Lys Met Val Pro Glu Asp Gly Ala Gly Val Cys Gly Pro Val Cys Ala Ala Gly Gly Pro Trp Ala Glu Val Arg Asp Ser Arg Thr Phe Thr Leu Val Ser Glu Val Glu Thr Gln Gly Asp Leu Ser Arg Lys Ile Pro Thr Glu Asp Glu Tyr Thr Asp Arg Pro Ser Gln Pro Ser Thr Gly Ser Leu Leu Leu Ile Gln Gln Gly Ser Lys Ser Ile Pro Pro Phe Gln Glu Pro Leu Glu Val Gly Glu Asn Asp Ser Leu Ser Gln Cys Phe Thr Gly Thr Glu Ser Thr Val Asp Ser Glu Gly Cys Asp Phe Thr Glu Pro Pro Ser Arg Thr Asp Ser Met Pro Val Ser Pro Glu Lys His Leu Thr Lys Glu Ile Glu Gly Asp Ser Cys Leu Pro Trp Val Val Ser Ser Asn Ser Thr Asp Gly Tyr Thr Gly Ser Gly Asn Thr Pro Gly Glu Asp His Glu Pro Phe Pro Gly Ser Leu Lys Cys Gly Pro Leu Pro Gln Cys Ala Tyr Ser Met Gly Phe Pro Ser Glu Ala Ala Ala Ser Met Ala Glu Ala Gly Val Arg Pro Gln Asp Arg Ala Asp Glu Arg Gly Ala Ser Gly Ser Gly Ser Ser Pro Ser Asp Gln Pro Pro Ala Ser Gly Asn Val Thr Gly Asn Ser Asn Ser Thr Phe Ile Ser Ser Gly Gln Val Met AAC TTC AAG GGT GAC ATC ATC GTG GTG TAT GTC AGC CAG ACC TCG, CAG 1632 Asn Phe Lys Gly Asp Ile Ile Val Val Tyr Val Ser Gln Thr Ser Gln Glu Gly Pro Gly Ser Ala Glu Pro Glu Ser Glu Pro Val Gly Arg Pro Val Gln Glu Glu Thr Leu Ala His Arg Asp Ser Phe Ala Gly Thr Ala Pro Arg Phe Pro Asp Val Cys Ala Thr Gly Ala Gly Leu Gln Glu Gln Gly Ala Pro Arg Gln Lys Asp Gly Thr Ser Arg Pro Val Gln Glu Gln Gly Gly Ala Gln Thr Ser Leu His Thr Gln Gly Ser Gly Gln Cys Ala Glu (2) INFORMATION FOR SEQ ID NO:15:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 625 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:15:

Met Ala Pro Arg Ala Arg Arg Arg Arg Gln Leu Pro Ala Pro Leu Leu Ala Leu Cys Val Leu Leu Val Pro Leu Gln Val Thr Leu Gln Val Thr Pro Pro Cys Thr Gln Glu Arg His Tyr Glu His Leu Gly Arg Cys Cys Ser Arg Cys Glu Pro Gly Lys Tyr Leu Ser Ser Lys Cys Thr Pro Thr Ser Asp Ser Val Cys Leu Pro Cys Gly Pro Asp Glu Tyr Leu Asp Thr Trp Asn Glu Glu Asp Lys Cys Leu Leu His Lys Val Cys Asp Ala Gly Lys Ala Leu Val Ala Val Asp Pro Gly Asn His Thr Ala Pro Arg Arg Cys Ala Cys Thr Ala Gly Tyr His Trp Asn Ser Asp Cys Glu Cys Cys Arg Arg Asn Thr Glu Cys Ala Pro Gly Phe Gly Ala Gln His Pro Leu Gln Leu Asn Lys Asp Thr Val Cys Thr Pro Cys Leu Leu Gly Phe Phe Ser Asp Val Phe Ser Ser Thr Asp Lys Cys Lys Pro Trp Thr Asn Cys Thr Leu Leu Gly Lys Leu Glu Ala His Gln Gly Thr Thr Glu Ser Asp Val Val Cys Ser Ser Ser Met Thr Leu Arg Arg Pro Pro Lys Glu Ala Gln Ala Tyr Leu Pro Ser Leu Ile Val Leu Leu Leu Phe Ile Ser Val Val Val Val Ala Ala Ile Ile Phe Gly Val Tyr Tyr Arg Lys Gly Gly Lys Ala Leu Thr Ala Asn Leu Trp Asn Trp Val Asn Asp Ala Cys Ser Ser Leu Ser Gly Asn Lys Glu Ser Ser Gly Asp Arg Cys Ala Gly Ser His Ser Ala Thr Ser Ser Gln Gln Glu Val Cys Glu Gly Ile Leu Leu Met Thr Arg Glu Glu Lys Met Val Pro Glu Asp Gly Ala Gly Val Cys Gly Pro Val Cys Ala Ala Gly Gly Pro Trp Ala Glu Val Arg Asp Ser Arg Thr Phe Thr Leu Val Ser Glu Val Glu Thr Gin Gly Asp Leu Ser Arg Lys Ile Pro Thr Glu Asp Glu Tyr Thr Asp Arg Pro Ser Gln Pro Ser Thr Gly Ser Leu Leu Leu Ile Gln Gln Gly Ser Lys Ser Ile Pro Pro Phe Gln Glu Pro Leu Glu Val Gly Glu Asn Asp Ser Leu Ser Gln Cys Phe Thr Gly Thr Glu Ser Thr Val Asp Ser Glu Gly Cys Asp Phe Thr Glu Pro Pro Ser Arg Thr Asp Ser Met Pro Val Ser Pro Glu Lys His Leu Thr Lys Glu Ile Glu Gly Asp Ser Cys Leu Pro Trp Val Val Ser Ser Asn Ser Thr Asp Gly Tyr Thr Gly Ser Gly Asn Thr Pro Gly Glu Asp His Glu Pro Phe Pro Gly Ser Leu Lys Cys Gly Pro Leu Pro Gln Cys Ala Tyr Ser Met Gly Phe Pro Ser Glu Ala Ala Ala Ser Met Ala Glu Ala Gly Val Arg Pro Gln Asp Arg Ala Asp Glu Arg Gly Ala Ser Gly Ser Gly Ser Ser Pro Ser Asp Gln Pro Pro Ala Ser Gly Asn Val Thr Gly Asn Ser Asn Ser Thr Phe Ile Ser Ser Gly Gln Val Met Asn Phe Lys Gly Asp Ile Ile Val Val Tyr Val Ser Gin Thr Ser Gln Glu Gly Pro Gly Ser Ala Glu Pro Glu Ser Glu Pro Val Gly Arg Pro Val Gln Glu Glu Thr Leu Ala His Arg Asp Ser Phe Ala Gly Thr Ala Pro Arg Phe Pro Asp Val Cys Ala Thr Gly Ala Gly Leu Gln Glu Gln Gly Ala Pro Arg Gln Lys Asp Gly Thr Ser Arg Pro Val Gln Glu Gln Gly Gly Ala Gln Thr Ser Leu His Thr Gln Gly Ser Gly Gln Cys Ala Glu (2) INFORMATION FOR SEQ ID NO:16:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 20 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:16:

Met Glu Thr Asp Thr Leu Leu Leu Trp Val Leu Leu Leu Trp Val Pro Gly Ser Thr Gly (2) INFORMATION FOR SEQ ID NO:17:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 5 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:17:
Asp Tyr Lys Asp Glu (2) INFORMATION FOR SEQ ID NO:18:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 6 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:18:
His His His His His His (2) INFORMATION FOR SEQ ID NO:19:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 33 amino acids (B) TYPE: amino acid (D) TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:19:

Arg Met Lys Gln Ile Glu Asp Lys Ile Glu Glu Ile Leu Ser Lys Ile Tyr His Ile Glu Asn Glu Ile Ala Arg Ile Lys Lys Leu Ile Gly Glu Arg

Claims (112)

CLAIMS:
1. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide capable of binding receptor activator of NF-.kappa.B
ligand (RANKL) or binding a Tumor Necrosis Factor receptor-associated factor (TRAF) selected from the group consisting of (a) a polypeptide comprising the amino acid sequence as set forth in SEQ ID NO:6; and (b) a polypeptide comprising the amino acid sequence x to y of SEQ ID NO:6, wherein x is any one of amino acids 1-33 and y is any one of amino acids 196-616;

wherein RANKL consists of the amino acid sequence of SEQ ID NO:13.
2. The isolated polypeptide of claim 1 comprising the amino acid sequence as set forth in SEQ ID NO:6.
3. The isolated polypeptide of claim 1 comprising the amino acid sequence x to y of SEQ ID NO:6, wherein x is any one of amino acids 1-33 and y is any one of amino acids 196-616.
4. The isolated polypeptide of claim 1 consisting of the amino acid sequence as set forth in SEQ ID NO:6.
5. The isolated polypeptide of claim 1 consisting of the amino acid sequence x to y of SEQ ID NO:6, wherein x is any one of amino acids 1-33 and y is any one of amino acids 196-616.
6. The isolated polypeptide of claim 1 or claim 3, wherein x is any one of amino acids 24-33 of SEQ ID NO:6.
7. The isolated polypeptide of claim 1 or claim 3, wherein x is amino acid 1 of SEQ ID NO:6.
8. The isolated polypeptide of any one of claims 1, 3 and 5 to 7, wherein y is any one of amino acids 196-213 of SEQ ID NO:6.
9. The isolated polypeptide of any one of claims 1, 3 and 5 to 7, wherein y is any one of amino acids 213-616 of SEQ ID NO:6.
10. The isolated polypeptide of any one of claims 1, 3 and 5 to 7, wherein y is amino acid 616 of SEQ ID NO:6.
11. The isolated polypeptide of any one of claims 1 and claims 3 to 7, wherein y is amino acid 213 of SEQ ID
NO:6.
12. The isolated polypeptide of claim 1 or claim 3 comprising the amino acid sequence of amino acids 33 through 196 of SEQ ID NO:6.
13. The isolated polypeptide of claim 1 or claim 3 comprising amino acids 33-213 of SEQ ID NO:6.
14. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide encoded by a DNA capable of hybridizing to the complement of nucleotides 33-213 of SEQ ID NO:5 under the conditions of hybridizing at 6XSSC at 63°C and washing in 3XSSC at 55°C, wherein the RANK polypeptide is capable of binding receptor activator of NF-.kappa.B ligand (RANKL) that comprises the amino acid sequence of SEQ ID NO:13 or binding a Tumor Necrosis Factor receptor-associated factor (TRAF).
15. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide comprising amino acids 339-421, inclusive, of SEQ ID NO:6, wherein said polypeptide is capable of binding TRAF6.
16. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide comprising amino acids 544-616, inclusive, of SEQ ID NO:6, wherein said polypeptide is capable of binding a TRAF selected from the group consisting of TRAF1, TRAF2, TRAF3, TRAFS and TRAF6.
17. A fragment of the polypeptide of claim 15 or claim 16, wherein the fragment is capable of binding TRAF6.
18. A fragment of the polypeptide of claim 15 or claim 16, wherein the fragment is capable of binding a TRAF
selected from the group consisting of TRAF1, TRAF2, TRAF3, TRAF5 and TRAF6.
19. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide consisting of the amino acid sequence according to SEQ ID NO:6 or a fragment thereof that is capable of binding a receptor activator of NF-.kappa.B ligand (RANKL) protein that consists of the amino acid sequence shown in SEQ ID
NO:13, wherein said polypeptide or fragment is capable of inducing an antibody that binds specifically with a polypeptide consisting of the amino acid sequence set forth in SEQ ID NO:6.
20. A receptor activator of NF-.kappa.B (RANK) fusion polypeptide capable of binding a receptor activator of NF-.kappa.B
ligand (RANKL) polypeptide consisting of the amino acid sequence of SEQ ID NO:13, said fusion polypeptide comprising the RANK polypeptide according to any one of claims 1 to 19 fused to an Fc region of a human immunoglobulin protein or a GST polypeptide.
21. The fusion polypeptide of claim 20 that comprises the RANK polypeptide of any one of claims 1 to 19 fused to an Fc region of an IgG immunoglobulin protein.
22. The fusion polypeptide according to claim 21, wherein the Fc region consists of amino acids 3-232 of SEQ
ID NO:8.
23. The fusion polypeptide according to claim 21 or claim 22, wherein the IgG immunoglobulin protein is a human IgG1 immunoglobulin protein.
24. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide capable of binding receptor activator of NF-.kappa.B
ligand (RANKL) or binding a Tumor Necrosis Factor receptor-associated factor (TRAF) selected from the group consisting of:

(a) a polypeptide comprising the amino acid sequence as set forth in SEQ ID NO:15; and (b) a polypeptide comprising the amino acid sequence x to y of SEQ ID NO: 15, wherein x is amino acid 1 or any one of amino acids 25-35 and y is any one of amino acids 197-214, wherein RANKL consists of the amino acid sequence of SEQ ID NO:11 or 13.
25. The isolated polypeptide of claim 24 comprising the amino acid sequence as set forth in SEQ ID NO:15.
26. The isolated polypeptide of claim 24 wherein x is amino acid 1 or any one of amino acids 25-35 of SEQ ID NO:15 and y is any one of amino acids 197-214 or amino acid 625 of SEQ ID NO:15.
27. The isolated polypeptide of claim 24 consisting of the amino acid sequence x to y of SEQ ID NO:15, wherein x is amino acid 1 or any one of amino acids 25-35 of SEQ ID NO:15 and y is any one of amino acids 197-214 or amino acid 625 of SEQ ID NO:15.
28. The isolated polypeptide of claim 24 comprising the amino acid sequence of amino acids 35 through 214.
29. A receptor activator of NF-.kappa.B (RANK) fusion polypeptide comprising amino acids x to 213 of SEQ ID NO:15, wherein x is amino acid 1 or 35 of SEQ ID NO:15, and amino acids 3-232 of SEQ ID NO:8, wherein the RANK fusion polypeptide is capable of binding a receptor activator of NF-.kappa.B ligand (RANKL) polypeptide consisting of the amino acid sequence of SEQ ID NO:13 or a TRAF protein.
30. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide encoded by a DNA capable of hybridizing to the complement of the nucleotide sequence set forth in SEQ ID
NO:5 under stringent conditions of hybridizing at 6XSSC at 63°C and washing in 3XSSC at 55°C, and wherein the polypeptide is capable of binding a TRAF selected from the group consisting of TRAF1, TRAF2, TRAF3, TRAF5 and TRAF6.
31. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide produced by a process comprising culturing a host cell transformed or transfected with a recombinant expression vector containing the nucleotide sequence set forth in SEQ ID NO:5 or containing a nucleotide sequence capable of hybridizing to the complement of the nucleotide sequence set forth in SEQ ID NO:5 under the stringent conditions of hybridizing in 6XSSC at 63°C and washing in 3XSSC at 55°C, and isolating the polypeptide resulting from the expression of said nucleotide sequence, said polypeptide being capable of binding a receptor activator of NF-.kappa.B
ligand (RANKL) protein that consists of the amino acid sequence shown in SEQ ID NO:13.
32. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide which is at least 80% identical in sequence to amino acids 544-616 of SEQ ID NO:6, wherein said polypeptide is capable of binding a TRAF selected from the group consisting of TRAF1, TRAF2, TRAF3, TRAF5 and TRAF6.
33. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide that is at least 70% identical in amino acid sequence to a polypeptide with the amino acid sequence x to y of SEQ ID NO:6, wherein x is any one of amino acids 1-33 and y is any one of amino acids 213-616, wherein said isolated polypeptide is capable of binding a receptor activator of NF-.kappa.B ligand (RANKL) polypeptide that consists of the amino acid sequence shown in SEQ ID NO:13.
34. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide comprising an amino acid sequence with at least 80% sequence identity to SEQ ID NO:6, wherein said isolated polypeptide is capable of binding a receptor activator of NF-.kappa.B ligand (RANKL) polypeptide that consists of the amino acid sequence shown in SEQ ID NO:13.
35. An isolated receptor activator of NF-.kappa.B (RANK) polypeptide comprising an amino acid sequence with at least 80% sequence identity to SEQ ID NO:15, wherein said isolated polypeptide is capable of binding to a receptor activator of NF-.kappa.B ligand (RANKL) polypeptide consisting of SEQ ID NO:11.
36. A soluble receptor activator of NF-.kappa.B (RANK) polypeptide comprising the amino acid sequence x to y of SEQ
ID NO:6, wherein x is any one of amino acids 1-33 and y is any one of amino acids 196-213 or fragments thereof, wherein the soluble RANK is capable of binding to a receptor activator of NF-.kappa.B ligand (RANKL) protein that consists of the amino acid sequence of SEQ ID NO:13.
37. The soluble polypeptide of claim 36 that is selected from the group consisting of:

(a) a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, of SEQ ID NO:6, and amino acids 3-232 of SEQ ID NO:8;

(b) a polypeptide comprising amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any of amino acids 24-33 inclusive, or fragments thereof; and (c) a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO:6, wherein x is any one of amino acids 1-33 of SEQ ID NO:6 and an Fc from a human immunoglobulin.
38. A fusion polypeptide comprising a first polypeptide covalently fused with a second polypeptide wherein the first polypeptide is the soluble polypeptide of claim 36 or 37 and the second polypeptide is a peptide selected from the group consisting of an immunoglobulin Fc domain, an immunoglobulin Fc mutein, a FLAG.TM. tag, a peptide comprising at least 6 His residues, a leucine zipper, and combinations thereof, wherein the fusion polypeptide is capable of binding receptor activator of NF-.kappa.B ligand (RANKL) that comprises the amino acid sequence of SEQ ID
NO:13 or binding a Tumor Necrosis Factor receptor-associated factor (TRAF).
39. An isolated DNA molecule encoding the polypeptide of any one of claims 1 to 38.
40. The isolated DNA molecule of claim 39 that is capable of hybridizing to the complement of the sequence encoding SEQ ID NO:6 or SEQ ID NO:15, under stringent conditions of hybridizing at 6XSSC at 63°C and washing in 3XSSC at 55°C.
41. The isolated DNA molecule of claim 39 comprising the nucleotides of SEQ ID NO:5.
42. A primer comprising at least 17 contiguous nucleotides of SEQ ID NO:5 and which binds specifically to the complement of the nucleotide sequence set forth as SEQ
ID NO:5.
43. A primer which comprises at least 17 contiguous nucleotides of SEQ ID NO:5 and which binds specifically to the complement of the nucleotide sequence set forth as SEQ
ID NO:5.
44. A primer which comprises at least 17 contiguous nucleotides of SEQ ID NO:14 and which binds specifically to the complement of the nucleotide sequence set forth as SEQ
ID NO:5.
45. The primer of claim 43, comprising at least 25 contiguous nucleotides of SEQ ID NO:5.
46. The primer of claim 44, comprising at least 25 contiguous nucleotides of SEQ ID NO:14.
47. The primer of claim 43, comprising at least 30 contiguous nucleotides of SEQ ID NO:5.
48. The primer of claim 44, comprising at least 30 contiguous nucleotides of SEQ ID NO:14.
49. A DNA molecule capable of hybridization to the complement of the nucleotide sequence set forth in SEQ ID
NO:5 under stringent conditions of hybridizing at 6XSSC at 63°C and washing in 3XSSC at 55°C, and which encodes receptor activator of NF-.kappa.B (RANK) that is capable of binding to receptor activator of NF-.kappa.B ligand (RANKL) consisting of the amino acid sequence of SEQ ID NO:13.
50. A DNA molecule capable of hybridizing to the complement of nucleotides 33-213 of SEQ ID NO:5 under stringent conditions of hybridizing at 6XSSC at 63°C and washing in 3XSSC at 55°C, and which encodes receptor activator of NF-.kappa.B (RANK) that is capable of binding to receptor activator of NF-.kappa.B ligand (RANKL) consisting of the amino acid sequence of SEQ ID NO:13.
51. A DNA molecule encoding fragments of proteins encoded by the DNA of any one of claims 39 to 41, wherein the fragments are capable of binding to receptor activator of NF-.kappa.B ligand (RANKL) consisting of the amino acid sequence of SEQ ID NO:13.
52. A recombinant expression vector comprising the DNA
molecule of any one of claims 49 to 51.
53. A host cell transformed or transfected with the expression vector of claim 52.
54. A process for preparing a RANK polypeptide, comprising culturing a host cell according to claim 53 under conditions promoting expression, and recovering the RANK
polypeptide.
55. A purified antibody that binds specifically with the polypeptide of any one of claims 1 to 19.
56. The purified antibody of claim 55 that binds specifically with the polypeptide of claim 5.
57. The purified antibody of claim 55 that binds specifically with the polypeptide of claim 27.
58. A purified antibody that binds specifically with the polypeptide of claim 12 or claim 28.
59. An isolated antibody that specifically binds a RANK polypeptide of amino acids 1-616 of SEQ ID NO:6; or amino acids 33-213 of SEQ ID NO:6.
60. The antibody of any one of claims 55 to 59, which is a monoclonal antibody.
61. A method for preparing an antibody, comprising immunizing a non-human mammal with the polypeptide of any one of claims 1 to 38 in an amount effective to produce antibodies which bind specifically to the polypeptide, and harvesting the antibodies.
62. A method for preparing an antibody, comprising immunizing a non-human mammal with the polypeptide of claim 12 or claim 28 in an amount effective to produce antibodies which bind specifically to the polypeptide and harvesting the antibodies.
63. A method for preparing an antibody, comprising immunizing a non-human mammal with a polypeptide comprising amino acids 544-616 of SEQ ID NO:6 in an amount effective to produce antibodies which bind specifically to the polypeptide and harvesting the antibodies.
64. A method for preparing an antibody, comprising immunizing a non-human mammal with a polypeptide comprising amino acids 339-421 of SEQ ID NO:6 in an amount effective to produce antibodies which bind specifically to the polypeptide and harvesting the antibodies.
65. A method for preparing an antibody, comprising immunizing a non-human mammal with a polypeptide comprising amino acids 33-213 of SEQ ID NO:6 in an amount effective to produce antibodies which bind specifically to the polypeptide and harvesting the antibodies.
66. A method for preparing the antibody according to claim 59, wherein the antibody is elicited by immunizing with:

(a) a polypeptide capable of binding receptor activator of NF-.kappa.B ligand (RANKL) and comprising an amino acid sequence that is at least 80% identical to SEQ ID NO:6, wherein RANKL is a polypeptide consisting of amino acids 1-317 of SEQ ID NO:13; or (b) a polypeptide capable of binding RANKL, wherein RANKL consists of amino acids 1-317 of SEQ ID NO:13, and wherein said polypeptide is encoded by a nucleic acid molecule that is capable of hybridizing to the complement of the nucleotide sequence shown in SEQ ID NO:5 under stringent conditions of hybridizing in 6XSSC at 63°C and washing in 3XSSC at 55°C.
67. An antibody produced by the method of any one of claims 61 to 66.
68. Use of the receptor activator of NF-.kappa.B (RANK) polypeptide of any one of claims 1 to 38 for the preparation of an antibody that binds specifically to the polypeptide.
69. A composition comprising the receptor activator of NF-.kappa.B (RANK) polypeptide of any one of claims 1 to 38 and a physiologically acceptable carrier, excipient or diluent.
70. A composition comprising the antibody of any one of claims 55 to 60 and a physiologically acceptable carrier, excipient or diluent.
71. A composition comprising an effective amount of a soluble polypeptide of claim 36 or 37 and a pharmaceutically acceptable carrier, excipient or diluent.
72. A method of inhibiting activation ofRANK signal transduction, comprising contacting an ex vivo cell that expresses membrane-associated receptor activator of NF-.kappa.B
(RANK) with a soluble RANK polypeptide of claim 36 or 37 and allowing the soluble polypeptide to bind receptor activator of NF-.kappa.B ligand (RANKL), thereby inhibiting binding of RANKL
to the membrane associated RANK, wherein RANK comprises SEQ ID NO:6 or 15, and wherein RANKL comprises SEQ ID NO:11 or 13.
73. Use of a soluble RANK polypeptide of claim 36 or 37, on a cell that expresses membrane-associated receptor activator of NF-.kappa.B (RANK), wherein the soluble RANK
polypeptide binds receptor activator of NF-.kappa.B ligand (RANKL), thereby inhibiting activation of RANK for treating a subject who has a tumor or neoplastic disease, wherein RANK comprises SEQ ID NO:6, and wherein RANKL comprises SEQ
ID NO:13.
74. Use of a composition comprising a receptor activator of NF-.kappa.B (RANK) antagonist in an amount sufficient to inhibit RANK signal transduction in a subject and a physiologically acceptable carrier, excipient or diluent, for therapeutically inhibiting RANK activity in said subject, wherein said subject has a tumor or neoplastic disease, and wherein said RANK antagonist is selected from the group consisting of an antibody that specifically binds a RANK protein consisting of the amino acid sequence shown in SEQ ID NO:6, and the soluble RANK polypeptide of claim 36 or 37.
75. Use of a receptor activator of NF-.kappa.B (RANK) antagonist in the preparation of a medicament for therapeutically inhibiting RANK activity in a subject having a tumor or neoplastic disease, wherein said RANK antagonist is selected from the group consisting of an antibody that specifically binds a RANK protein consisting of the amino acid sequence shown in SEQ ID NO:6, the antibody of any one of claims 55 to 60 and the soluble RANK polypeptide of claim 36 or 37.
76. The use of claim 75, wherein said RANK antagonist is the soluble RANK polypeptide of claim 36 or 37.
77. The use of claim 75 wherein said RANK antagonist is the antibody of claim 59.
78. The use of any one of claims 74 to 77, wherein the subject is undergoing radiation therapy.
79. Use of a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, of SEQ ID NO:6, and an Fc from a human IgG1 immunoglobulin for inhibiting RANK
activity in a subject having a tumor or neoplastic disease, wherein RANK activity is the ability of RANK to bind RANK
ligand, andwherein said RANK:FC polypeptide is in an amount sufficient to inhibit RANK signal transduction in said subject, wherein RANK comprises SEQ ID NO:6.
80. The use of claim 74, wherein said RANK antagonist is a soluble RANK of claim 36 or 37, and said soluble RANK
further comprises an Fc from a human immunoglobulin.
81. Use of a fusion protein comprising a receptor activator of NF-.kappa.B (RANK) polypeptide linked to a human immunoglobulin Fc region, wherein said RANK polypeptide has at least 90% amino acid sequence identity with amino acids 33 to 213 of SEQ ID NO:6 and is capable of binding a RANKL protein consisting of the amino acid sequence shown in SEQ ID NO:13, and further wherein said RANK polypeptide is in an amount sufficient to inhibit RANK signal transduction in a subject having a tumor or neoplastic disease.
82. Use of the receptor activator of NF-.kappa.B (RANK) polypeptide or RANK fusion polypeptide of any one of claims 1 to 23, 30 to 34 and 36 to 38, for treating a subject having a tumor or neoplastic disease.
83. Use of a composition that comprises a receptor activator of NF-.kappa.B (RANK) polypeptide and a physiologically acceptable carrier, excipient or diluent for treating a subject having a tumor or neoplastic disease, wherein the RANK polypeptide has an at least 90% amino acid sequence identity with amino acids 33-213 of SEQ ID NO:6;

is capable of binding a receptor activator of NF-.kappa.B ligand (RANKL) polypeptide that consists of the amino acid sequence shown in SEQ ID NO:13;

is in an amount sufficient to inhibit RANK signal transduction in said subject.
84. Use of the receptor activator of NF-.kappa.B (RANK) polypeptide or RANK fusion polypeptide of any one of claims 1 to 38 to screen for an inhibitor of NF-.kappa.B
activation.
85. Use of the composition of claim 70 for inhibiting RANK activity in a subject who has a tumor or neoplastic disease, wherein RANK activity is the ability of RANK to bind RANK ligand.
86. Use of a composition comprising an antagonist of a receptor activator of NF-.kappa.B (RANK) polypeptide of any one of claims 1 to 38 and a physiologically acceptable carrier, excipient or diluent to inhibit RANK activity in a subject in need thereof wherein said RANK antagonist is selected from the group consisting of an antibody that specifically binds a RANK protein consisting of the amino acid sequence shown in SEQ ID NO:6, an antagonistic antibody against a receptor activator of NF-.kappa.B (RANK) polypeptide of any one of claims 1 to 19 and 30 to 34, and the soluble RANK
polypeptide of claim 36 or 37, wherein the subject has a tumor or neoplastic disease, and wherein RANK activity is the ability of RANK to bind RANK ligand.
87. The use according to claim 86, wherein the RANK
antagonist is selected from the group consisting of an antagonistic antibody against a receptor activator of NF-.kappa.B
(RANK) polypeptide of any one of claims 1 to 19, and a soluble RANK polypeptide of claim 36 or 37.
88. The use according to claim 87, wherein the RANK
antagonist is a soluble RANK polypeptide that comprises amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, or fragments thereof.
89. The use according to any one of claims 85 to 88, wherein said composition is in a dosage format adapted for administration by bolus injection, continuous infusion or sustained release from an implant.
90. The use according to any one of claims 85 to 89, wherein the subject is undergoing radiation therapy.
91. A kit comprising:

(a) a soluble polypeptide of claim 36 or 37; and (b) instructions for using said polypeptide to regulate, treat or prevent a tumor or neoplastic disease.
92. A kit comprising:

(a) the isolated receptor activator of NF-.kappa.B
(RANK) polypeptide of any one of claims 1 to 23 and 30 to 34, the soluble RANK polypeptide of claim 36 or 37, or the fusion polypeptide of claim 38; and (b) instructions for using said polypeptide to regulate, treat or prevent a tumor or neoplastic disease.
93. A kit comprising:

(a) the antibody of any one of claims 55 to 60;
and (b) instructions for using said antibody to regulate, treat or prevent a tumor or neoplastic disease.
94. A kit for detecting a receptor activator of NF-.kappa.B
(RANK) or a receptor activator of NF-.kappa.B ligand (RANKL) polypeptide, or for detecting RANK-related activity, the kit comprising:

(a) instructions for the detecting; and (b) the polypeptide of any one of claims 1 to 38 or the antibody of any one of claims 55 to 60.
95. A kit comprising a receptor activator of NF-.kappa.B
(RANK) antagonist in an amount sufficient to inhibit RANK
signal transduction in a subject and a physiologically acceptable carrier, excipient or diluent, wherein said RANK
antagonist is selected from the group consisting of an antibody that specifically binds a RANK protein consisting of the amino acid sequence shown in SEQ ID NO:6, and a soluble RANK polypeptide of claim 36 or 37, together with instructions for therapeutically inhibiting RANK activity in a subject having a tumor or neoplastic disease.
96. The kit of claim 95, wherein the subject is undergoing radiation therapy.
97. A kit comprising a RANK:Fc polypeptide comprising amino acids x to 213 of SEQ ID NO:6, wherein x is amino acid 1, or any one of amino acids 24-33, inclusive, of SEQ ID NO:6, and an Fc from a human immunoglobulin; together with instructions for inhibiting RANK activity in a subject having a tumor or neoplastic disease, wherein said RANK:Fc polypeptide is in an amount sufficient to inhibit RANK

signal transduction in said subject.
98. The kit of claim 95, wherein said RANK antagonist is a soluble RANK of claim 36 or 37, and said soluble RANK
further comprises an Fc from a human immunoglobulin.
99. A kit comprising a fusion protein comprising a RANK polypeptide linked to a human immunoglobulin Fc region, wherein said RANK polypeptide has at least 90% amino acid sequence identity with amino acids 33 to 213 of SEQ ID NO:6 and is capable of binding a RANKL protein consisting of the amino acid sequence shown in SEQ ID NO:13, and further wherein said RANK polypeptide is in an amount sufficient to inhibit RANK signal transduction in said subject, together with instructions for inhibiting RANK activity in a subject having a tumor or neoplastic disease.
100. A kit comprising a composition that comprises a receptor activator of NF-.kappa.B (RANK) polypeptide capable of binding a RANKL polypeptide that consists of the amino acid sequence shown in SEQ ID NO:13, wherein the RANK polypeptide has an at least 90% amino acid sequence identity with amino acids 33-213 of SEQ ID NO:6 and a physiologically acceptable carrier, excipient or diluent together with instructions for treating a subject having a tumor or neoplastic disease.
101. A method of screening a molecule for its capacity to antagonize or agonize receptor activator of NF-.kappa.B (RANK), which comprises SEQ ID NO:6, said method comprising:

a) contacting a RANK polypeptide or RANK fusion polypeptide of any one of claims 1 to 38 with a receptor activator of NF-.kappa.B ligand (RANKL) polypeptide, which comprises SEQ ID NO: 13, or a fragment thereof that binds said RANK polypeptide, in the presence of the molecule under conditions that permit binding of the RANK and RANKL
polypeptides;

(b) detecting the binding of the RANK and RANKL
polypeptides;

(c) determining that the molecule is a RANK
antagonist if the amount of RANK-RANKL binding detected is decreased when the molecule is present; and (d) determining that the molecule is a RANK
agonist if the amount of RANK-RANKL binding detected is increased when the molecule is present.
102. The method according to claim 101, wherein the molecule is an antibody that binds a RANK polypeptide that consists of the amino acid sequence of SEQ ID NO:6 or a RANKL polypeptide that consists of the amino acid sequence of SEQ ID NO:13.
103. The method according to claim 101 or 102, wherein the detecting step is selected from the group consisting of enzyme-linked immunosorbent assay (ELISA), dot blot employing an antibody that binds native RANK, fluorescence activated cell sorter assay (FACS assay) and a solid phase binding assay.
104. A method of screening a molecule for its capacity to antagonize or agonize the binding of a NF-.kappa.B (RANK) polypeptide to a TRAF polypeptide comprising the steps of:

(a) incubating a mixture comprising the molecule, a TRAF polypeptide and a RANK polypeptide of any one of claims 15-18, wherein said mixture is incubated under conditions that allow the RANK polypeptide and the TRAF
polypeptide to form RANK/TRAF complexes;

(b) detecting the RANK/TRAF complexes;

(c) determining that the molecule antagonizes binding of the RANK polypeptide to the TRAF polypeptide if the amount of the RANK/TRAF complexes detected is decreased when the molecule is present; and (d) determining that the molecule agonizes binding of the RANK polypeptide to the TRAF polypeptide if the amount of the RANK/TRAF complexes detected is increased when the molecule is present.
105. The method according to claim 104, wherein the TRAF polypeptide is selected from the group consisting of TRAF1, TRAF2, TRAF3, TRAF5 and TRAF6.
106. The method according to claim 101 or 104, wherein the molecule being screened is selected from the group consisting of a soluble RANK polypeptide as defined in claim 36 or 37, a RANKL polypeptide comprising SEQ ID NO:13 or a RANK-binding fragment thereof, an antisense oligonucleotide capable of binding to an mRNA or DNA that encodes RANK which comprises SEQ ID NO:6, an antibody against RANK which consists of SEQ ID NO:6, an antibody against RANKL which consists of SEQ ID NO:13, and a peptide.
107. The method according to claim 106, wherein the molecule being screened is a soluble RANK polypeptide, and further wherein the soluble RANK polypeptide is selected from the group consisting of:

(a) a RANK polypeptide comprising amino acids x to 213 of SEQ ID NO:15, wherein x is amino acid 1 or an amino acid between 25 and 35 of SEQ ID NO:15;

(b) a RANK polypeptide comprising amino acids x to 213 of SEQ ID NO:6, wherein x is an amino acid between 1 and 33 of SEQ ID NO:6; and (c) a polypeptide which is capable of binding receptor activator of NF-.kappa.B ligand (RANKL) comprising SEQ ID
NO: 13 and is encoded by a nucleic acid molecule that is capable of hybridizing with the complement of the nucleotide sequence shown in SEQ ID NO:5 or SEQ ID NO:14 under stringent conditions of hybridizing in 6XSSC at 63°C
overnight and washing in 3XSSC at 55°C.
108. The method according to claim 107, wherein the soluble RANK polypeptide further comprises an immunoglobulin Fc domain.
109. The method according to claim 106, wherein the molecule being screened is a RANKL polypeptide, and further wherein the RANKL polypeptide is selected from the group consisting of:

(a) a polypeptide comprising amino acids 69-317 of SEQ ID NO:13;

(b) a polypeptide comprising amino acids 138-317 of SEQ ID NO:13;

(c) a polypeptide comprising amino acids 119-294 of SEQ ID NO:11; and (d) a polypeptide which is capable of binding receptor activator of NF-.kappa.B (RANK) comprising SEQ ID NO: 6 and is encoded by a nucleic acid molecule that is capable of hybridizing to the complement of the nucleotide sequence shown in SEQ ID NO:10 or SEQ ID NO:12 under stringent conditions of hybridizing in 6XSSC at 63°C and washing in 3XSSC at 55°C.
110. A method of screening a molecule for its capacity to agonize or antagonize RANK, said method comprising the steps of:

(a) incubating the molecule with a cell that expresses a RANK polypeptide on its surface, wherein said RANK polypeptide has an amino acid sequence selected from the group consisting of:

(i) amino acids x to y of SEQ ID NO:6, wherein x is any one of amino acids 1-33 and y is amino acid 616; and (ii) a polypeptide that is at least 80% identical to amino acids 33-616 of SEQ ID NO:6 and that is capable of binding a RANKL polypeptide consisting of amino acids 1-317 of SEQ ID NO:13;

(b) measuring a biological activity associated with the RANK polypeptide, wherein said biological activity is selected from the group consisting of:

(i) binding a RANKL polypeptide, which comprises SEQ ID NO: 13, or a RANK-binding fragment thereof; and (ii) ability of the RANK polypeptide to bind TRAF1, TRAF2, TRAF3, TRAF5 or TRAF6; and (c) determining that the molecule is a RANK
agonist if the biological activity is increased in the presence of the molecule or that the molecule is a RANK
antagonist if the biological activity is decreased in the presence of the molecule.
111. The method according to claim 110, wherein during the incubation step the cells are contacted with a soluble RANKL polypeptide which is a soluble fragment of SEQ ID

NO:13, a membrane-bound RANKL polypeptide which is a membrane-bound fragment of SEQ ID NO:13, or an agonistic antibody against RANK which comprises SEQ ID NO:6.
112. The method according to claim 111, wherein the cells are selected from the group consisting of epithelial cells, B-cells, activated T cells, CD1a+ dendritic cells, macrophages, and mammalian cells that are expressing exogenous recombinant RANK.
CA2274803A 1996-12-23 1997-12-22 Receptor activator of nf-kappa b, receptor is member of tnf receptor superfamily Expired - Fee Related CA2274803C (en)

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US5997896P 1996-12-23 1996-12-23
US60/059,978 1996-12-23
US81350997A 1997-03-07 1997-03-07
US08/813,509 1997-03-07
US6467197P 1997-10-14 1997-10-14
US60/064,671 1997-10-14
PCT/US1997/023866 WO1998028424A2 (en) 1996-12-23 1997-12-22 Receptor activator of nf-kappa b, receptor is member of tnf receptor superfamily

Publications (2)

Publication Number Publication Date
CA2274803A1 CA2274803A1 (en) 1998-07-02
CA2274803C true CA2274803C (en) 2012-05-22

Family

ID=43981413

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2274803A Expired - Fee Related CA2274803C (en) 1996-12-23 1997-12-22 Receptor activator of nf-kappa b, receptor is member of tnf receptor superfamily

Country Status (4)

Country Link
JP (1) JP2013005802A (en)
CA (1) CA2274803C (en)
IL (3) IL130491A (en)
PT (1) PT946725E (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4138013B2 (en) * 1996-12-23 2008-08-20 イミュネックス・コーポレーション Ligand for receptor activator of NF-KAPPA B, a member of TNF superfamily

Also Published As

Publication number Publication date
IL130491A (en) 2011-10-31
CA2274803A1 (en) 1998-07-02
IL215020A0 (en) 2011-10-31
JP2013005802A (en) 2013-01-10
IL215021A0 (en) 2011-10-31
IL130491A0 (en) 2000-06-01
PT946725E (en) 2011-04-20

Similar Documents

Publication Publication Date Title
CA2274987C (en) Ligand for receptor activator of nf-kappa b, ligand is member of tnf superfamily
WO1998028426A9 (en) Ligand for receptor activator of nf-kappa b, ligand is member of tnf superfamily
CA2274803C (en) Receptor activator of nf-kappa b, receptor is member of tnf receptor superfamily
AU741504B2 (en) Receptor activator of NF-kappa B, receptor is member of TNF receptor superfamily

Legal Events

Date Code Title Description
EEER Examination request
MKLA Lapsed

Effective date: 20151222