US20050244411A1 - BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders - Google Patents

BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders Download PDF

Info

Publication number
US20050244411A1
US20050244411A1 US11/065,669 US6566905A US2005244411A1 US 20050244411 A1 US20050244411 A1 US 20050244411A1 US 6566905 A US6566905 A US 6566905A US 2005244411 A1 US2005244411 A1 US 2005244411A1
Authority
US
United States
Prior art keywords
baff
cells
mice
cell
antibody
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/065,669
Inventor
Fabienne MacKay
Susan Kalled
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.)
Biogen MA Inc
Original Assignee
Biogen Idec MA Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US2000/001788 external-priority patent/WO2000043032A2/en
Application filed by Biogen Idec MA Inc filed Critical Biogen Idec MA Inc
Priority to US11/065,669 priority Critical patent/US20050244411A1/en
Publication of US20050244411A1 publication Critical patent/US20050244411A1/en
Priority to US12/061,398 priority patent/US9545086B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New breeds of animals
    • A01K67/027New breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0278Humanized animals, e.g. knockin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70575NGF/TNF-superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2875Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF/TNF superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2878Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the NGF-receptor/TNF-receptor superfamily, e.g. CD27, CD30, CD40, CD95
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2207/00Modified animals
    • A01K2207/15Humanized animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0306Animal model for genetic diseases
    • A01K2267/0325Animal model for autoimmune diseases
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/0331Animal model for proliferative diseases
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0368Animal model for inflammation
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0375Animal model for cardiovascular diseases
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • A01K2267/035Animal model for multifactorial diseases
    • A01K2267/0381Animal model for diseases of the hematopoietic system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to the use of a ligand, BAFF, a ⁇ -cell activating factor belonging to the Tumor Necrosis Family and its blocking agents to either stimulate or inhibit the expression of B-cells and immunoglobulins.
  • This protein and its receptor may have anti-cancer and/or immunoregulatory applications as well as uses for the treatment of immunosuppressive disorders such as HIV.
  • the ligand and its blocking agents may play a role in the development of hypertension and its related disorders.
  • cells transfected with the gene for this ligand may be used in gene therapy to treat tumors, autoimmune diseases or inherited genetic disorders involving B-cells.
  • Blocking agents such as recombinant variants or antibodies specific to the ligand or its receptor, may have immunoregulatory applications as well.
  • Use of BAFF as a B-cell stimulator for immune suppressed diseases including for example uses for patients undergoing organ transplantation (i.e. bone marrow transplant) as well as recovering from cancer treatments to stimulate production of B-cells are contemplated.
  • Use of BAFF as an adjuvant and or costimulator to boast and or restore B cells levels to approximate normal levels are also contemplated.
  • TNF tumor-necrosis factor
  • TNF-related cytokines are mediators of host defense and immune regulation.
  • Members of this family exist in membrane-anchored forms, acting locally through cell-to-cell contact, or as secreted proteins capable of diffusing to more distant targets.
  • a parallel family of receptors signals the presence of these molecules leading to the initiation of cell death or cellular proliferation and differentiation in the target tissue.
  • the TNF family of ligands and receptors has at least 11 recognized receptor-ligand pairs, including: TNF:TNF-R; LT- ⁇ :TNF-R; LT- ⁇ / ⁇ :LT- ⁇ -R; FasL:Fas; CD40L:CD40; CD30L:CD30; CD27L:CD27; OX40L:OX40 and 4-1BBL:4-1BB.
  • the DNA sequences encoding these ligands have only about 25% to about 30% identity in even the most related cases, although the amino acid relatedness is about 50%.
  • This family of cytokine receptors The defining feature of this family of cytokine receptors is found in the cysteine rich extracellular domain initially revealed by the molecular cloning of two distinct TNF receptors.
  • This family of genes encodes glycoproteins characteristic of Type I transmembrane proteins with an extracellular ligand binding domain, a single membrane spanning region and a cytoplasmic region involved in activating cellular functions.
  • the cysteine-rich ligand binding region exhibits a tightly knit disulfide linked core domain, which, depending upon the particular family member, is repeated multiple times. Most receptors have four domains, although there may be as few as three, or as many as six.
  • Proteins in the TNF family of ligands are characterized by a short N-terminal stretch of normally short hydrophilic amino acids, often containing several lysine or arginine residues thought to serve as stop transfer sequences.
  • the C-terminal binding region comprises the bulk of the protein, and often, but not always, contains glycosylation sites.
  • cleavage in the stalk region can occur early during protein processing and the ligand is then found primarily in secreted form. Most ligands, however, exist in a membrane form, mediating localized signaling.
  • TNF and lymphotoxin-I are both structured into a sandwich of two anti-parallel ⁇ -pleated sheets with the “jelly roll” or Greek key topology.
  • the rms deviation between the C ⁇ and ⁇ residues is 0.61 C, suggesting a high degree of similarity in their molecular topography.
  • a structural feature emerging from molecular studies of CD40L, TNF and LT- ⁇ is the propensity to assemble into oligomeric complexes. Intrinsic to the oligomeric structure is the formation of the receptor binding site at the junction between the neighboring subunits creating a multivalent ligand.
  • TNF, CD40L and LT- ⁇ have been shown to exist as trimers by analysis of their crystal structures. Many of the amino acids conserved between the different ligands are in stretches of the scaffold ⁇ -sheet. It is likely that the basic sandwich structure is preserved in all of these molecules, since portions of these scaffold sequences are conserved across the various family members. The quaternary structure may also be maintained since the subunit conformation is likely to remain similar.
  • TNF family members can best be described as master switches in the immune system controlling both cell survival and differentiation.
  • TNF and LT ⁇ are currently recognized as secreted cytokines contrasting with the other predominantly membrane anchored members of the TNF family.
  • a membrane form of TNF has been well-characterized and is likely to have unique biological roles, secreted TNF functions as a general alarm signaling to cells more distant from the site of the triggering event.
  • TNF secretion can amplify an event leading to the well-described changes in the vasculature lining and the inflammatory state of cells.
  • the membrane bound members of the family send signals though the TNF type receptors only to cells in direct contact.
  • T cells provide CD40 mediated “help” only to those B cells brought into direct contact via cognate TCR interactions. Similar cell-cell contact limitations on the ability to induce cell death apply to the well-studied Fas system.
  • TNF TNF
  • Fas ligand and TRAIL can efficiently induce cell death in many lines and their receptors mostly likely have good canonical death domains.
  • DR-3 TRAMP/WSL-1
  • TWEAK CD30 ligand
  • LTa1b2 TWEAK
  • How this group can trigger cell death in the absence of a canonical death domain is an interesting question and suggests that a separate weaker death signaling mechanism exists.
  • members that cannot efficiently deliver a death signal Probably all groups can have antiproliferative effects on some cell types consequent to inducing cell differentiation e.g. CD40. Funakoshi et al. (1994).
  • TNF family has grown dramatically in recent years to encompass at least 11 different signaling pathways involving regulation of the immune system.
  • the widespread expression patterns of TWEAK and TRAIL indicate that there is still more functional variety to be uncovered in this family.
  • This aspect has been especially highlighted recently in the discovery of two receptors that affect the ability of rous sacroma and herpes simplex virus to replicate as well as the historical observations that TNF has anti-viral activity and pox viruses encode for decoy TNF receptors. Brojatsch et al. (1996); Montgomery et al. (1996); Smith et al. (1994), 76 Cell 959-962; Vassalli et al. (1992), 10 Immunol. 411-452.
  • TNF is a mediator of septic shock and cachexia, and is involved in the regulation of hematopoietic cell development. It appears to play a major role as a mediator of inflammation and defense against bacterial, viral and parasitic infections as well as having antitumor activity. TNF is also involved in different autoimmune diseases. TNF may be produced by several types of cells, including macrophages, fibroblasts, T cells and natural killer cells. TNF binds to two different receptors, each acting through specific intracellular signaling molecules, thus resulting in different effects of TNF. TNF can exist either as a membrane bound form or as a soluble secreted cytokine.
  • LT-I shares many activities with TNF, i.e. binding to the TNF receptors, but unlike TNF, appears to be secreted primarily by activated T cells and some ⁇ -lymphoblastoid tumors.
  • the heteromeric complex of LT- ⁇ and LT- ⁇ is a membrane bound complex which binds to the LT- ⁇ receptor.
  • the LT system (LTs and LT-R) appears to be involved in the development of peripheral lymphoid organs since genetic disruption of LT- ⁇ leads to disorganization of T and B cells in the spleen and an absence of lymph nodes.
  • the LT- ⁇ system is also involved in cell death of some adenocarcinoma cell lines.
  • Fas-L another member of the TNF family, is expressed predominantly on activated T cells. It induces the death of cells bearing its receptor, including tumor cells and HIV-infected cells, by a mechanism known as programmed cell death or apoptosis. Furthermore, deficiencies in either Fas or Fas-L may lead to lymphoproliferative disorders, confirming the role of the Fas system in the regulation of immune responses. The Fas system is also involved in liver damage resulting from hepatitis chronic infection and in autoimmunity in HIV-infected patients. The Fas system is also involved in T-cell destruction in HIV patients. TRAIL, another member of this family, also seems to be involved in the death of a wide variety of transformed cell lines of diverse origin.
  • CD40-L another member of the TNF family, is expressed on T cells and induces the regulation of CD40-bearing B cells. Furthermore, alterations in the CD40-L gene result in a disease known as X-linked hyper-IgM syndrome.
  • the CD40 system is also involved in different autoimmune diseases and CD40-L is known to have antiviral properties. Although the CD40 system is involved in the rescue of apoptotic B cells, in non-immune cells it induces apoptosis. Many additional lymphocyte members of the TNF family are also involved in costimulation.
  • the members of the TNF family have fundamental regulatory roles in controlling the immune system and activating acute host defense systems. Given the current progress in manipulating members of the TNF family for therapeutic benefit, it is likely that members of this family may provide unique means to control disease.
  • Some of the ligands of this family can directly induce the apoptotic death of many transformed cells e.g. LT, TNF, Fas ligand and TRAIL. Nagata (1997) 88 Cell 355-365. Fas and possibly TNF and CD30 receptor activation can induce cell death in nontransformed lymphocytes which may play an immunoregulatory function. Amakawa et al. (1996) 84 Cell 551-562; Nagata (1997) 88 Cell 355-365; Sytwu et al.
  • death is triggered following the aggregation of death domains which reside on the cytoplasmic side of the TNF receptors.
  • the death domain orchestrates the assembly of various signal transduction components which result in the activation of the caspase cascade. Nagata (1997) 88 Cell 355-365.
  • Some receptors lack canonical death domains, e.g. LTb receptor and CD30 (Browning et al. (1996); Lee et al. (1996)) yet can induce cell death, albeit more weakly.
  • Sjögren's syndrome is a chronic inflammatory disorder characterized by the destruction of exocrine glands such as salivary and lacrimal glands, leading to symptoms of dry mouth (xerostomia) and eyes (keratoconjuncitivitis sicca).
  • Jonsson et al. (2000) Sjogren's syndrome in Arthritis and allied conditions 1826-1849.
  • SS is regarded as an autoimmune disease characterized by the presence of large mononuclear cell infiltrates in exocrine glands, B cell hyper-reactivity and various serum autoantibodies.
  • SS can develop alone or in association with other autoimmune disorders such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Jonnson et al. (2000); Manoussakis et al. (1998).
  • SLE systemic lupus erythematosus
  • RA rheumatoid arthritis
  • Abnormal B cell activity is a predominant feature of SS, which is manifested by massive polyclonal B cell activation and elevated secretion of autoantibodies such as rheumatoid factors (RF), anti-Ro (SS-A), anti-La (SS-B) and anti- ⁇ -fodrin autoantibodies.
  • RF rheumatoid factors
  • SS-A anti-Ro
  • SS-B anti-La
  • Intense B cell activity such as germinal center reactions occur in exocrine glands of some patients, placing them in a high risk category for the development of lymphomas.
  • Intense B cell activity such as germinal center reactions occur in exocrine glands of some patients, placing them in a high risk category for the development of lymphomas.
  • BAFF B cell activating factor belonging to the TNF family
  • TNF TNF cytokine-family
  • TNF TNF cytokine-family
  • transgenic mice overexpressing BAFF under the control of a liver-specific promoter. These mice have excessive numbers of mature B cells, spontaneous germinal center reactions, secrete autoantibodies, and have high plasma cell numbers in secondary lymphoid organs and Ig deposition in the kidney.
  • the BAFF Tg mice develop as they age a secondary condition to their lupus-like disease, showing interesting similarities with that of SS in humans.
  • the present invention is directed to the use of BAFF-ligands, blocking agents and antibodies for the ligand, to either stimulate or inhibit the growth of B-cells and the secretion of immunoglobulin.
  • the claimed invention may be used for therapeutic applications in numerous diseases and disorders, as discussed in more detail below, as well as to obtain information about, and manipulate, the immune system and its processes. Further, this invention can be used as a method of stimulating or inhibiting the growth of B-cells and the secretion of immunoglobulins.
  • BAFF associated molecules as described by this invention, may also have utility in the treatment of autoimmune diseases, disorders relating to B-cell proliferation and maturation, BAFF ligand regulation and inflammation. The invention may be involved in the regulation or prevention of hypertension and hypertension-related disorders of the renal and cardiovascular tissue.
  • the invention includes a method of effecting B-cell growth and secretion of immunoglobulins through the administration of various BAFF ligands and related molecules.
  • the invention also contemplates stimulating B-cell growth through the use of BAFF ligands or active fragments of the polypeptide.
  • the polypeptide may be use alone or with a CD40 ligand or an anti-murine antibody.
  • the invention relates to methods of stimulation of dendritic cell-induced B-cell growth and maturation through the use of BAFF ligands or active fragments of BAFF.
  • the polypeptide may be used alone or with CD40 ligand or anti- ⁇ antibodies.
  • blocking agents of BAFF and the BAFF receptor have been used to inhibit B-cell growth and immunoglobulin secretion. These agents can be inoperative, recombinant BAFF, BAFF specific antibodies, BAFF-receptor specific antibodies or an anti-BAFF ligand molecule.
  • the invention relates to the use of BAFF, BAFF related molecules and BAFF blocking agents to treat hypertension, hypertension related disorders, immune disorders, autoimmune diseases, inflammation and B-cell lympho-proliferate disorders.
  • the invention relates to the use of BAFF, BAFF-related molecules and BAFF blocking agents to treat Sjogren's syndrome.
  • the invention relates to methods for treating or reducing the advancement, severity or effects of Sjogren's syndrome in a patient by administering a pharmaceutical preparation comprising a therapeutically effective amount of a BAFF blocking agent and a pharmaceutically acceptable carrier.
  • the BAFF blocking agent may be a soluble BAFF receptor molecule, an antibody directed against BAFF-ligand or an antibody directed against a BAFF receptor.
  • the BAFF receptor may be BCMA, TACI, or BAFF R.
  • the invention encompasses the use of BAFF and BAFF-related molecules as either agonists or antagonists in effecting immune responses by effecting the growth and/or maturation of B-cells and secretion of immunoglobulin.
  • the invention relates in other embodiments to soluble constructs comprising BAFF which may be used to directly trigger BAFF mediated pharmacological events. Such events may have useful therapeutic benefits in the treatment of cancer, tumors or the manipulation of the immune system to treat immunologic diseases.
  • the claimed invention relates to antibodies directed against BAFF ligand, which can be used, for example, for the treatment of cancers, and manipulation of the immune system to treat immunologic disease.
  • the invention relates to methods of gene therapy using the genes for BAFF.
  • compositions of the invention may, optionally, include pharmaceutically acceptable carriers, adjuvants, fillers, or other pharmaceutical compositions, and may be administered in any of the numerous forms or routes known in the art.
  • FIG. 1 depicts the predicted amino acid sequence of human [SEQ. ID. NO.: 1] and mouse BAFF [SEQ. ID. NO.:2].
  • the predicted transmembrane domain TMD, dashed line
  • the potential N-linked glycosylation sites stars
  • the natural processing site of human BAFF arrow
  • the double line above hBAFF indicates the sequence obtained by Edman degradation of the processed form of BAFF.
  • B Depicts a comparison of the extracellular protein sequence of BAFF [SEQ. ID. NO.: 3] and some members of the TNF ligand family [SEQ. ID. NO.: 4 (hAPRIL); SEQ. ID. NO.: 5 (hTNF alpha); SEQ. ID.
  • FIG. 2 is a schematic characterization of recombinant BAFF (A) Schematic representation of recombinant BAFF constructs. Soluble recombinant BAFFs starting at Leu 83 and Gln 136 are expressed fused to a N-terminal Flag tag and a 6 amino acid linker. The long form is cleaved between Arg 133 and Ala 134 (arrow) in 293 T cells, to yield a processed form of BAFF. Asn 124 and Asn 242 belong to N-glycosylation consensus sites. N-linked glycan present on Asn 124 is shown as a Y. TMD: transmembrane domain.
  • FIG. 3 depicts expression of BAFF
  • A Northern blots (2 ⁇ g poly A + RNA per lane) of various human tissues were probed with BAFF antisense mRNA.
  • B Reverse transcriptase amplification of BAFF, IL-2 receptor alpha chain and actin from RNA of purified blood T cells at various time points of PHA activation, E-rosetting negative blood cells (B cells and monocytes), in vitro derived immature dendritic cells, 293 cells, and 293 cells sterilely transfected with full length BAFF (293-BAFF). Control amplifications were performed in the absence of added cDNA. IL-2 receptor alpha chain was amplified as a marker of T cell activation.
  • FIG. 4 depicts BAFF binding to mature B cells.
  • A Binding of soluble BAFF to BJAB and Jurkat cell lines, and to purified CD19 + cells of cord blood. Cells were stained with the indicated amount (in ng/50 ⁇ l) of Flag-BAFF and analyzed by flow cytometry.
  • B Binding of soluble BAFF to PBLS. PBLs were stained with anti-CD8-FITC or with anti-CD19-FITC (horizontal axis) and with Flag-BAFF plus M2-biotin and avidin-PE (vertical axis). Flag-BAFF was omitted in controls.
  • FIG. 5 depicts BAFF costimulates B cell proliferation.
  • A Surface expression of BAFF in stably transfected 293 cells. 293-BAFF and 293 wild-type cells were stained with anti-BAFF mAb 43.9 and analyzed by flow cytometry.
  • B Costimulation of PBLs by 293-BAFF cells. PBLs (10 5 /well) were incubated with 15.000 glutaraldehyde-fixed 293 cells (293 wt or 293-BAFF) in the presence or absence of anti-B cell receptor antibody (anti- ⁇ ). Fixed 293 cells alone incorporated 100 cpm.
  • C Dose dependent costimulation of PBL proliferation by soluble BAFF in the presence of anti- ⁇ .
  • Purified CD19 + B cells were activated by coculture with EL-4 T cells and activated T cell supernatants for 5-6 d, then re-isolated and cultured for another 7 days in the presence of medium only ( ⁇ ) or containing 5% activated T cell supernatants (T-SUP) or a blend of cytokines (IL-2, IL-4, IL-10).
  • the columns represent means of Ig concentrations for cultures with or without 1 ⁇ g/ml BAFF. Means ⁇ SD in terms of “fold increase” were 1.23 ⁇ 0.11 for medium only, 2.06 ⁇ 0.18 with T cell supernatants (4 experiments) and 1.45 ⁇ 0.06 with IL-2, IL-4 and IL-10 (2 experiments).
  • FIG. 6 depicts that BAFF acts as a cofactor for B cell proliferation.
  • the proliferation of human PBL was measured alone (500 cpm), with the presence of BAFF ligand alone, with the presence of goat anti-murine (mu) alone, and with both BAFF ligand and anti-mu.
  • the combination of both anti-mu and BAFF significantly raised proliferation of PBL as the concentration of BAFF increased suggesting BAFF's cofactor characteristics.
  • FIG. 7 depicts increased B cell numbers in BAFF Tg mice.
  • the graph compares 12 control littermates (left panel) with 12 BAFF Tg mice (right panel). Lymphocytes counts are shown with circles and granulocytes (including neutrophils, eosinophils, basophils) with diamonds.
  • B Increased proportion of B cells in PBL from BAFF Tg mice.
  • PBL were stained with both anti-B220-FITC and anti-CD4-PE for FACS analysis and gated on live cells using the forward side scatter. Percentages of CD4 and B220 positive cells are indicated.
  • One control mouse (left) and two BAFF Tg mice (right) are shown and the results were representative of 7 animals analysed in each group.
  • (E) Increased Bcl-2 expression in B cells from BAFF Tg mice PBL. Bcl-2 expression was measured by intracytoplasmic staining and cells were analysed by FACS. In both (D) and (E) Live cells were gated on the forward side scatter. Four control littermates (white bars) and 4 BAFF Tg mice are shown and are representative of at least 12 animals analysed for each group. MFI: mean of fluorescence intensity. The difference between control animals and BAFF Tg mice was statistically significant (P ⁇ 0.005).
  • FIG. 8 depicts increased B cell compartments in the spleen but not in the bone marrow of BAFF Tg mice.
  • FIG. 9 depicts increased Ig, RF and CIC levels in BAFF Tg mice
  • FIG. 10 depicts the presence of anti-ssDNA and anti-dsDNA autoantibodies in some BAFF Tg mice.
  • FIG. 11 depicts enlarged Peyer's patches in BAFF Tg mice. Photography of Peyers patches (indicated with an arrow) on the small intestine of a control mouse (left) and a BAFF Tg mouse (right). This pictures is representative of at least 12 mice sacrificed for each group. Magnification 5 ⁇
  • FIG. 12 depicts disrupted T and B cell organization, intense germinal center reactions, decreased number of dendritic cells and increased number of plasma cells in the spleen of BAFF Tg mice.
  • a control mouse is shown in A, C, E and G and a BAFF Tg in B, D, F, and H.
  • B cells are blue and T cells brown (A and B).
  • Germinal centers are shown with an arrow (C and D). Only few residual germinal centers are seen in control mice (C).
  • CD11c positive dendritic cells are brown and appear in the T cell zone, bridging channels and the marginal zone (E). Very few are present in BAFF Tg mice (F).
  • Syndecan-1-positive plasma cells were only detectable in the red pulp of BAFF Tg mice (H) but not control mice (G).
  • FIG. 13 depicts disrupted T and B cells organization, intense germinal center reactions and large number of plasma cells in the MLN of BAFF Tg mice.
  • the control mouse is shown in A, C, E and G and the BAFF Tg mouse is shown in B, D, F, and H.
  • the immunohistochemistry was performed as described in FIG. 6 .
  • T and B cell staining is shown in A and B, germinal centers in C and D, dendritic cells E and F and plasma cells in G and H.
  • GC germinal center. Magnification 100 ⁇ .
  • FIG. 14 depicts enlarged and inflamed salivary glands BAFF Tg mice.
  • A Mice 15-17 months old, 4 control littermates (white bar) and 4 BAFF Tg mice (grey bar) were sacrificed the same day for organ collection. Both right and left submaxillary glands were collected and weighted. The data shows the mean t standard deviation of weight for both glands. These results are representative of at least 4 separate groups of age-matched animals dissected.
  • B Paraffin sections of submaxillary glands from a control littermate (left panel) and two BAFF Tg mice (middle and right panel) were stained with H&E. Arrows indicate ducts and acinar cells in the left and right panels.
  • FIG. 15 depicts tumor-like clusters of B-lymphoid cells in a submaxillary tumor or infiltrates present in submaxillary glands of BAFF Tg mice.
  • Frozen sections of submaxillary glands (A and C) or a submaxillary tumor (B, D and E) were stained with H&E (A, B and C) or Biotin-labelled anti-mouse B220 (D) or anti-mouse syndecan-1 (E), follwed by Streptavidin-HRP brown staining in D and E).
  • Arrows in A, B, C and D indicate the tumor-like clusters
  • arrow in E shows the presence of syndecan-1 positive plasma cells whereas the stars indicate the location of tumor-like clusters.
  • Panels (A) and (C) are representative of 4 animals in which these clusters were detected as cells infiltrating submaxillary glands.
  • Panels (B), (D) and (E) are representative of 3 submaxillary tumors analysed. Magnification is indicated for each panel.
  • a germinal center (GC) is also indicated in panel (B).
  • FIG. 16 depicts identification of a Mz-like B cell population infiltrating salivary glands of BAFF Tg mice.
  • Submaxillary glands from 13-17 months old BAFF Tg mice and age-matched control littermates were digested with collagenase for purification of infiltrating lymphocytes as described in materials and methods.
  • Using four-color flow cytometry analysis cells were stained with anti-B220, anti-CD5, anti-IgM and anti-CD43.
  • (A) shows the gating on B220 hi and B220 lo/int B cells on control (left) and BAFF Tg mice (right) as indicated.
  • (B) shows four dot plots for expression of IgM and CD5 on B220 hi cells (left) and B220 lo/int (right) from BAFF Tg mice and control mice as indicated.
  • the IgM hi B cell gate on B220 hi cells (left) and the B-1a and B-1b gates on B220 lo/int cells are shown.
  • (C) shows histograms for the expression of CD43 on gated IgM hi /B220 hi cells (left) and gated B-1a and B-1b cells (right) from BAFF Tg mice as indicated.
  • Lymphocytes prepared from submaxillary glands (left panel) and inguinal lymph nodes (right panel) of a BAFF Tg mouse were stained with anti-B220, anti-IgM and anti-L-selectin, using a four-color flow cytometry procedure. Cells from submaxillary glands were gated on B220 hi /IgM hi (left panel) and on IgM+(right panel). A histogram of L-selectin expression is shown in each panel.
  • E Lymphocytes from a BAFF Tg mice (right) and a control littermate (left) were prepared as in A and stained with anti-B220, anti-IgM and either anti-IgD, anti-CD23, anti-CD21, anti-CD1, anti-HSA, using a three color flow cytometry procedure. Cells were gated on B220 hi and IgM hi cells as indicated on the top histograms and expression of IgD, CD23, CD1, CD21 and HSA on these gated cells is shown on histograms for control (left) and Tg mice (right) as indicated.
  • FIG. 17 depicts decreased saliva flow in older BAFF Tg mice. 13 BAFF Tg mice (diamonds) and 14 control littermates (circles) were injected with pilocarpine prior to saliva collection as described in materials and methods. Mice 13-15.5 months old are shown in (A) and mice 8-10 months old are shown in (B). Means of saliva flow are shown with a bar and p values are indicated in each panel.
  • FIG. 18 depicts elevated levels of BAFF in sera and salivary gland tissues from patients suffering from primary SS but lack of correlation of these levels with levels of total IgG, RF and presence of anti-Ro/La autoantibodies.
  • A individual BAFF levels in 39 healthy controls (square), 41 patients with primary SS (diamond), 53 patients with SLE (circle) and 53 patients with RA (triangle) were measured in sera by ELISA and BAFF levels plotted on a log scale. Controls were drawn from normal healthy donors. The horizontal black bars indicates the average for each group: normal 10.4 ⁇ 13 (ng/ml), SS patients 53 ⁇ 67 (ng/ml), SLE patients 12.7 ⁇ 24.4 (ng/ml) and RA patients 23 ⁇ 47 (ng/ml).
  • This invention relates to the use of BAFF and BAFF related molecules to effect the growth and maturation of B-cells and the secretion of immunoglobulin.
  • the invention relates to the use of BAFF and BAFF related molecules to effect responses of the immune system, as necessitated by immune-related disorders. Additionally, this invention encompasses the treatment of cancer and immune disorders through the use of a BAFF, BAFF blocking agents, or BAFF related gene through gene therapy methods.
  • the BAFF ligand and homologs thereof produced by hosts transformed with the sequences of the invention, as well as native BAFF purified by the processes known in the art, or produced from known amino acid sequences, are useful in a variety of methods for anticancer, antitumor and immunoregulatory applications. They are also useful in therapy and methods directed to other diseases.
  • BAFF blocking agents refers to agents that can diminish BAFF ligand binding to BAFF receptors, or can diminish BAFF receptor signalling, or that can influence how the BAFF receptor signal is interpreted within the cell.
  • a BAFF blocking agent that acts by diminishing ligand-receptor binding can inhibit BAFF ligand binding by at least 20%.
  • BAFF blocking agents include soluble BAFF receptor-Fc molecules, anti-BAFF ligand antibodies and anti-BAFF receptor antibodies.
  • BAFF receptors have been identified and characterized and include TACI (see, e.g., U.S. Pat. No. 5,969,102 and WO98/39361, incorporated herein by reference), BCMA (see, e.g., WO01/12812, incorporated herein by reference), and BAFFR (see, e.g., Thompson et al. (2001) Science 293:2108, incorporated herein by reference).
  • antisense therapy refers to administration or in situ generation of oligonucleotides or their derivatives which specifically hybridize under cellular conditions with the cellular mRNA and/or DNA encoding the ligand of interest, so as to inhibit expression of the encoded protein, i.e. by inhibiting transcription and/or translation.
  • the binding may be by conventional base pair complementarity, or, for example, in the case of binding to DNA duplexes, through specific interactions in the major groove of the double helix.
  • “antisense” therapy refers to a range of techniques generally employed in the art, and includes any therapy which relies on specific binding to oligonucleotide sequences.
  • an antisense construct of the present invention can be delivered, for example, as an expression plasmid, which, when transcribed in the cell, produces RNA which is complementary to at least a portion of the cellular mRNA which encodes Kay-ligand.
  • the antisense construct can be an oligonucleotide probe which is generated ex vivo.
  • Such oligonucleotide probes are preferably modified oligonucleotides which are resistant to endogenous nucleases, and are therefor stable in vivo.
  • Exemplary nucleic acids molecules for use as antisense oligonucleotides are phosphoramidates, phosphothioate andmethylphosphonate analogs of DNA (See, e.g., U.S. Pat.
  • the BAFF-ligand of the invention is a member of the TNF family and is described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herein.
  • the protein, fragments or homologs thereof may have wide therapeutic and diagnostic applications.
  • the BAFF-ligand is present primarily in the spleen and in peripheral blood lymphocytes, strongly indicating a regulatory role in the immune system. Comparison of the claimed BAFF-ligand sequences with other members of the human TNF family reveals considerable structural similarity. All the proteins share several regions of sequence conservation in the extracellular domain.
  • the precise three-dimensional structure of the claimed ligand is not known, it is predicted that, as a member of the TNF family, it may share certain structural characteristics with other members of the family.
  • novel polypeptides of the invention specifically interact with a receptor, which has not yet been identified.
  • the peptides and methods disclosed herein enable the identification of receptors which specifically interact with the BAFF-ligand or fragments thereof.
  • the claimed invention in certain embodiments includes methods of using peptides derived from BAFF-ligand which have the ability to bind to their receptors.
  • Fragments of the BAFF-ligands can be produced in several ways, e.g., recombinantly, by PCR, proteolytic digestion or by chemical synthesis. Internal or terminal fragments of a polypeptide can be generated by removing one or more nucleotides from one end or both ends of a nucleic acid which encodes the polypeptide. Expression of the mutagenized DNA produces polypeptide fragments.
  • Polypeptide fragments can also be chemically synthesized using techniques known in the art such as conventional Merrifield solid phase f-moc or t-boc chemistry.
  • peptides and DNA sequences of the present invention may be arbitrarily divided into fragments of desired length with no overlap of the fragment, or divided into overlapping fragments of a desired length. Methods such as these are described in more detail below.
  • Soluble forms of the BAFF-ligand can often signal effectively and hence can be administered as a drug which now mimics the natural membrane form. It is possible that the BAFF-ligand claimed herein are naturally secreted as soluble cytokines, however, if not, one can reengineer the gene to force secretion. To create a soluble secreted form of BAFF-ligand or BAFF receptor, one would remove at the DNA level the N-terminus transmembrane regions, and some portion of the stalk region, and replace them with a type I leader or alternatively a type II leader sequence that will allow efficient proteolytic cleavage in the chosen expression system.
  • a skilled artisan could vary the amount of the stalk region retained in the secretion expression construct to optimize both receptor binding properties and secretion efficiency.
  • the constructs containing all possible stalk lengths i.e. N-terminal truncations, could be prepared such that proteins starting at amino acids 81 to 139 would result. The optimal length stalk sequence would result from this type of analysis.
  • Soluble forms of BAFF receptors can be prepared using techniques well-known to those of ordinary skill in the art. Immunoglobulin Fc portions may be fused to the BAFF receptor protein to increase the half-life of the soluble BAFF receptor. Production of such soluble BAFF receptor proteins is described, for example, in WO 01/12812, WO 01/24811, and PCT/US01/40626, the entire disclosures of which are incorporated herein by reference.
  • the invention also includes antibodies specifically reactive with the claimed BAFF-ligand or its receptors.
  • Anti-protein/anti-peptide antisera or monoclonal antibodies can be made by standard protocols (See, for example, Antibodies: A Laboratory Manual ed. by Harlow and Lane (Cold Spring Harbor Press: 1988)).
  • a mammal such as a mouse, a hamster or rabbit can be immunized with an immunogenic form of the peptide.
  • Techniques for conferring immunogenicity on a protein or peptide include conjugation to carriers, or other techniques, well known in the art.
  • An immunogenic portion of BAFF-ligand or its receptors can be administered in the presence of an adjuvant.
  • the progress of immunization can be monitored by detection of antibody titers in plasma or serum.
  • Standard ELISA or other immunoassays can be used with the immunogen as antigen to assess the levels of antibodies.
  • the subject antibodies are immunospecific for antigenic determinants of BAFF-ligand or its receptors, (e.g. antigenic determinants of a polypeptide of SEQ. ID. NO.: 2, said sequence as described in PCT application number PCTI/US98/19037 (WO99/12964) and is incorporated in its entirety herewith), or a closely related human or non-human mammalian homolog (e.g. 70, 80 or 90 percent homologous, more preferably at least 95 percent homologous).
  • the anti-BAFF-ligand or anti-BAFF-ligand-receptor antibodies do not substantially cross react (i.e.
  • a protein which is e.g., less than 80 percent homologous to SEQ. ID. NO.: 2 or 6 said sequence as described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herewith; preferably less than 90 percent homologous with SEQ. ID. NO.: 2 said sequence as described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herewith; and, most preferably less than 95 percent homologous with SEQ. ID. NO.: 2 said sequence as described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herewith.
  • the antibody has a binding affinity for a non-homologous protein which is less than 10 percent, more preferably less than 5 percent, and even more preferably less than 1 percent, of the binding affinity for a protein of SEQ. ID. NO.: 2 said sequence as described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herewith.
  • antibody as used herein is intended to include fragments thereof which are also specifically reactive with BAFF-ligand, or its receptors.
  • Antibodies can be fragmented using conventional techniques and the fragments screened for utility in the same manner as described above for whole antibodies. For example, F(ab′) 2 fragments can be generated by treating antibody with pepsin. The resulting F(ab′) 2 fragment can be treated to reduce disulfide bridges to produce Fab′ fragments.
  • the antibodies of the present invention are further intended to include biospecific and chimeric molecules having anti-BAFF-ligand or anti-BAFF-ligand-receptor activity.
  • both monoclonal and polyclonal antibodies (Ab) directed against BAFF-ligand, Tumor-ligand and their receptors, and antibody fragments such as Fab′ and F(ab′) 2 , can be used to block the action of the Ligand and their respective receptor.
  • chimeric antibodies can be constructed in which the antigen binding domain from an animal antibody is linked to a human constant domain (e.g. Cabilly et al., U.S. Pat. No. 4,816,567, incorporated herein by reference). Chimeric antibodies may reduce the observed immunogenic responses elicited by animal antibodies when used in human clinical treatments.
  • Humanized antibodies which recognize BAFF-ligand or its receptors can be synthesized.
  • Humanized antibodies are chimeras comprising mostly human IgG sequences into which the regions responsible for specific antigen-binding have been inserted. Animals are immunized with the desired antigen, the corresponding antibodies are isolated, and the portion of the variable region sequences responsible for specific antigen binding are removed. The animal-derived antigen binding regions are then cloned into the appropriate position of human antibody genes in which the antigen binding regions have been deleted. Humanized antibodies minimize the use of heterologous (i.e. inter species) sequences in human antibodies, and thus are less likely to elicit immune responses in the treated subject.
  • Construction of different classes of recombinant antibodies can also be accomplished by making chimeric or humanized antibodies comprising variable domains and human constant domains (CH1, CH2, CH3) isolated from different classes of immunoglobulins.
  • antibodies with increased antigen binding site valencies can be recombinantly produced by cloning the antigen binding site into vectors carrying the human: chain constant regions.
  • standard recombinant DNA techniques can be used to alter the binding affinities of recombinant antibodies with their antigens by altering amino acid residues in the vicinity of the antigen binding sites.
  • the antigen binding affinity of a humanized antibody can be increased by mutagenesis based on molecular modeling. Queen et al., (1989) Proc. Natl. Acad. Sci. 86: 10029-33 incorporated herein by reference.
  • Analogs of the BAFF-ligand can differ from the naturally occurring BAFF-ligand in amino acid sequence, or in ways that do not involve sequence, or both.
  • Non-sequence modifications include in vivo or in vitro chemical derivatization of the BAFF-ligand.
  • Non-sequence modifications include, but are not limited to, changes in acetylation, methylation, phosphorylation, carboxylation or glycosylation.
  • Preferred analogs include BAFF-ligand biologically active fragments thereof, whose sequences differ from the sequence given in SEQ. ID NO. 2 said sequence as described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herewith, by one or more conservative amino acid substitutions, or by one or more non-conservative amino acid substitutions, deletions or insertions which do not abolish the activity of BAFF-ligand.
  • Conservative substitutions typically include the substitution of one amino acid for another with similar characteristics, e.g.
  • valine glycine
  • glycine alanine
  • valine isoleucine, leucine
  • aspartic acid glutamic acid
  • asparagine glutamine
  • serine threonine
  • lysine arginine
  • phenylalanine tyrosine
  • the anti-Flag M2 monoclonal antibody, biotinylated anti-Flag M2 antibody and the anti-Flag M2 antibody coupled to agarose were purchased from Sigma. Cell culture reagents were obtained from Life Sciences (Basel, Switzerland) and Biowhittaker (Walkersville, Md.). Flag-tagged soluble human APRIL (residues K 110 -L 250 ) was produced in 293 cells as described (10, 11). FITC-labeled anti-CD4, anti-CD8 and anti-CD19 antibodies were purchased from Pharmingen (San Diego, Calif.). Goat F(ab) 2 specific for the Fc 5 , fragment of human IgM were purchased from Jackson ImmunoResearch (West Grove, Pa.). Secondary antibodies were obtained from either Pharmingen or from Jackson ImmunoResearch and used at the recommended dilutions.
  • Human embryonic kidney 293 T (12) cells and fibroblast cell lines were maintained in DMEM containing 10% heat-inactivated fetal calf serum (FCS).
  • Human embryonic kidney 293 cells were maintained in DMEM-nutrient mix F12 (1:1) supplemented with 2% FCS.
  • T cell lines, B cell lines, and macrophage cell lines were grown in RPMI supplemented with 10% FCS.
  • Molt-4 cells were cultiv-atedin Iscove's medium-supplemented with 10% FCS.
  • Epithelial cell lines were grown in MEM-alpha medium containing 10% FCS, 0.5 mM non-essential amino acids, 10 mM Na-Hepes and 1 mM Na pyruvate.
  • HUVECs were maintained in M199 medium supplemented with 20% FCS, 100 ⁇ g/ml of epithelial cell growth factor (Collaborative Research, Inotech, Dottikon, Switzerland) and 100 ⁇ g/ml of heparin sodium salt (Sigma). All media contained penicillin and streptomycin antibiotics.
  • Peripheral blood leukocytes were isolated from heparinized blood of healthy adult volunteers by Ficoll-Paque (Pharmacia, Uppsala, Sweden) gradient centrifugation and cultured in RPMI, 10% FCS.
  • T cells were obtained from non-adherents PBLs by rosetting with neuraminidase-treated sheep red blood cells and separated from non-rosetting cells (mostly B cells and monocytes) by Ficoll-Paque gradient centrifugation. Purified T cells were activated for 24 h with phytohemagglutinin (Sigma) (1 ⁇ g/ml), washed and cultured in RPMI, 10% FCS, 20 U/ml of IL 2.
  • CD14 + monocytes were purified by magnetic cell sorting using anti-CD 14 antibodies, goat anti-mouse-coated microbeads and a MinimacsTM device (Miltenyi Biotech), and cultivated in the presence of GM-CSF (800 U/ml, Leucomax®, Essex Chemie, Luzern, Switzerland) and IL-4 (20 ng/ml, Lucerna Chem, Luzern, Switzerland) for 5 d, then with GM-CSF, IL4 and TNF ⁇ (200 U/ml, Bender, Vienna, Austria) for an additional 3 d to obtain a CD83 + , dentritic cell-like population.
  • Human B cells of >97% purity were isolated from peripheral blood or umbilical cord blood using anti-CD19 magnetic beads (M450, Dynal, Oslo, Norway) as described (13).
  • Northern blot analysis was carried out using Human Multiple Tissue Northern Blots I and II (Clontech #7760-1 and #7759-1). The membranes were incubated in hybridization solution (50% formamide, 2.5 ⁇ Denhardt's, 0.2% SDS, 10 mM EDTA, 2 ⁇ SSC, 50 mM NaH 2 PO 4 , pH 6.5, 200 ⁇ g/ml sonicated salmon sperm DNA) for 2 h at 60° C. Antisense RNA probe containing the nucleotides corresponding to amino acids 136-285 of hBAFF was heat-denatured and added at 2 ⁇ 10 6 cpm/ml in fresh hybridization solution.
  • hybridization solution 50% formamide, 2.5 ⁇ Denhardt's, 0.2% SDS, 10 mM EDTA, 2 ⁇ SSC, 50 mM NaH 2 PO 4 , pH 6.5, 200 ⁇ g/ml sonicated salmon sperm DNA
  • the membrane was hybridized 16 h at 62° C., washed once in 2 ⁇ SSC, 0.05% SDS (30 min at 25° C.), once in 0.1 ⁇ SSC, 0.1% SDS (20 min at 65° C.) and exposed at ⁇ 70° C. to X-ray films.
  • a partial sequence of human BAFF cDNA was contained in several EST clones (e.g. GenBank Accession numbers T87299 and AA166695) derived from fetal liver and spleen and ovarian cancer libraries.
  • the 5′ portion of the cDNA was obtained by 5′-RACE-PCR (Marathon-Ready cDNA, Clonetech, Palo Alto, Calif.) amplification with oligonucleotides AP1 and JT1013 (5′-ACTGTTTCTTCTGGACCCTGAACGGC-3′) [SEQ ID. NO.: 9] using the provided cDNA library from a pool of human leukocytes as template, as recommended by the manufacturer.
  • PCR-0 blunt Invitrogen, Nev. Leek, The Netherlands
  • subcloned as EcoRI/PstI fragment into pT7T3 Pac vector (Pharmacia) containing EST clone T87299.
  • Full-length hBAFF cDNA was therefore obtained by combining 5′ and 3′ fragments using the internal PstI site of BAFF. Sequence has been assigned GenBank accession number AF116456.
  • a partial 617 bp sequence of murine BAFF was contained in two overlapping EST clones (AA422749 and AA254047).
  • a PCR fragment spanning nucleotides 158 to 391 of this sequence was used as a probe to screen a mouse spleen cDNA library (Stratagene, La Jolla, Calif.).
  • hBAFF Full length hBAFF was amplified using oligos JT1069 (5′-GACAAGCTTGCCACCATGGATGACTCCACA-3′) [SEQ. ID. NO.: 10] and JT637 (5′-ACTAGTCACAGCAGTITCAATGC-3) [SEQ. ID. NO.: 11].
  • the PCR product was cloned into PCR-0 blunt and re-subcloned as HindIII/EcoRI fragment into PCR-3 mammalian expression vector.
  • a short version of soluble BAFF (amino acids Q136-L285) was amplified using oligos JT636 (5′-CTGCAGGGTCCAGAAGAAACAG-3) [SEQ. ID. NO.: 12] and JT637.
  • a long version of soluble BAFF (aa L83-L285) was obtained from full length BAFF using internal PstI site. Soluble BAFFs were resubcloned as PstI/EcoRI fragments behind the haemaglutinin signal peptide and Flag sequence of a modified PCR-3 vector, and as PstI/SpeI fragments into a modified pQE16 bacterial expression vector in frame with a N-terminal Flag sequence (14). Constructs were sequenced on both strands. The establishment of stable 293 cell lines expressing the short soluble form or full length BAFF, and the expression and purification of recombinant soluble BAFF from bacteria and mammalian 293 cells was performed as described (14, 15).
  • RNA extracted from T cells, B cells, in vitro derived immature dendritic cells, 293 wt and 293-BAFF (full length) cells was reverse transcribed using the Ready to Go system (Pharmacia) according to the manufacturer's instructions.
  • BAFF and ⁇ -actin cDNAs were detected by PCR amplification with Taq DNA polymerase (steps of 1 min each at 94° C., 55° C. and 72° C. for 30 cycles) using specific oligonucleotides: for BAFF, JT1322 5′-GGAGAAGGCAACTCCAGTCAGAAC-3′ [SEQ. ID.
  • 293T cells were transiently transfected with the short form of soluble BAFF and grown in serum-free Optimem medium for 7 d.
  • Conditionned supernatants were concentrated 20 ⁇ , mixed with internal standards catalase and ovalbumin, and loaded onto a Superdex-200 HR10/30 column. Proteins were eluted in PBS at 0.5 ml/min and fractions (0.25 ml) were precipitated with trichloroacetic acid and analyzed by Western blotting using anti-Flag M2 antibody.
  • the column was calibrated with standard proteins: ferritin (440 kDa), catalase (232 kDa), aldolase (158 kDa), bovine serum albumine (67 kDa), ovalbumine (43 kDa), chymotrypsinogen A (25 kDa) and ribonuclease A (13.7 kDa).
  • Samples were heated in 20 ⁇ l of 0.5% SDS, 1% 2-mercaptoethanol for 3 min at 95° C., then cooled and supplemented with 10% Nonidet P40 (2 ⁇ l), 0.5 M sodium phosphate, pH 7.5 (2 ⁇ l) and Peptide N-glycanase F (125 units/ ⁇ l, 1 ⁇ l, or no enzyme in controls). Samples were incubated for 3 h at 37° C. prior to analysis by Western blotting.
  • 293 T cells were transiently transfected with the long form of soluble BAFF and grown in serum-free Optimem medium for 7 d.
  • Conditioned supernatants were concentrated 20 ⁇ , fractionated by SDS-PAGE and blotted onto polyvinylidene difluoride membrane (BioRad Labs, Hercules, Calif.) as previously described (16), and then sequenced using a gas phase sequencer (ABI 120A, Perkin Elmer, Foster City, Calif.) coupled to an analyzer (ABI 120A, Perkin Elmer) equipped with a phenylthiohydantoin C18 2.1 ⁇ 250 mm column. Data was analyzed using software ABI 610 (Perkin. Elmer).
  • Polyclonal antibodies were generated by immunizing rabbits (Eurogentec, Seraing, Belgium) with recombinant soluble BAFF. Spleen of rats immunized with the same antigen were fused to x63Ag8.653 mouse myeloma cells, and hybridoma were screened for BAFF-specific IgGs.
  • FACS buffer PBS, 10% FCS, 0.02% NaN 3
  • Anti-BAFF mAb 43.9 was used at 40 ⁇ g/ml.
  • peripheral blood lymphocytes were stained with Flag tagged soluble BAFF/long (2 ⁇ g/ml), followed by biotinylated anti-Flag M2 (1/400) and PE-labeled streptavidin (1/100), followed by either FITC-labeled anti-CD4, anti-CD8 or anti-CD19.
  • Peripheral blood leukocytes were incubated in 96-well plates (10 5 cells/well in 100 ⁇ l RPMI supplemented with 10% FCS) for 72 h in the presence or absence of 2 ⁇ g/ml of goat anti-human p chain antibody (Sigma) or control F(ab′) 2 and with the indicated concentration of native or boiled soluble BAFF/long. Cells were pulsed for an additional 6 h with [ 3 H]thymidine (1 ⁇ Ci/well) and harvested. [ 3 H]thymidine incorporation was monitored by liquid scintillation counting.
  • recombinant soluble BAFF was replaced by 293 cells stably transfected with full length BAFF (or 293 wt as control) that had been fixed for 5 min at 25° C. in 1% paraformaldeyde. Assay was performed as described (17). In further experiments, CD19 + cells were isolated form PBL with magnetic beads and the remaining CD19 ⁇ cells were irradiated (3000 rads) prior to renconstitution with CD19+cells. Proliferation assay with sBAFF was then performed as described above.
  • Purified B cells were activated in the EL4 culture system as described (13). Briefly, 10 4 B cells mixed with 5 ⁇ 10 4 irradiated murine ELA thymoma cells (clone B5) were cultured for 5-6 d in 200 ⁇ l medium containing 5% v/v of culture supernatants from human T cells (106/ml) which had been activated for 48 h with PHA (1 ⁇ g/ml) and PMA (1 ng/ml).
  • B cells were then reisolated with anti-CD19 beads and cultured for another 7 d (5 ⁇ 10 4 cells in 200 ⁇ l, duplicate or triplicate culture in flat bottomed 96 well plates) in medium alone or in medium supplemented with 5% T cell supernatants, or with 50 ng/ml IL-2 (a kind gift from the former Glaxo Institute for Molecular Biology, Geneva) and 10 ng/ml each IL4 and IL-10 (Peprotech, London, UK), in the presence or absence of sBAFF.
  • the anti-Flag M2 antibody was added at a concentration of 2 ⁇ g/ml and had no effect by itself.
  • IgM, IgG and IgA in culture supernatants were quantitated by ELISA assays as described (13).
  • BAFF Human BAFF was identified by sequence homology as a possible novel member of the TNF ligand family while we screened public databases using an improved profile search (18).
  • a cDNA encoding the complete protein of 285 amino acids (aa) was obtained by combining EST-clones (covering the 3′ region) with a fragment (5′ region) amplified by PCR.
  • the absence of a signal peptide suggested that BAFF was a type II membrane protein that is typical of the members of the TNF-ligand family.
  • the protein has a predicted cytoplasmic domain of 46 aa, a hydrophobic transmembrane region, and an extracellular domain of 218 aa containing two potential N-glycosylation sites ( FIG. 1A ).
  • the sequence of the extracellular domain of BAFF shows highest homology with APRIL (33% amino acid identities, 48% homology), whereas the identity with other members of the family such as TNF, FasL, LTa, TRAIL or RANKL is below 20% ( FIG. 1B , C).
  • the mouse BAFF cDNA clone isolated from a spleen library encoded a slightly longer protein (309 aa) due to an insertion between the transmembrane region and the first of several ⁇ -strands which constitute the receptor binding domain in all TNF ligand members (19).
  • This ⁇ -strand rich ectodomain is almost identical in mouse and human BAFF (86% identity, 93% homology) suggesting that the BAFF gene has been highly conserved during evolution ( FIG. 1A ).
  • TNF family members are synthesized as membrane-inserted ligands, cleavage in the stalk region between transmembrane and receptor binding domain is frequently observed.
  • TNF or FasL are readily cleaved from the cell surface by metalloproteinases (20, 21).
  • metalloproteinases (20, 21).
  • a recombinant soluble 32 kDa form of BAFF aa 83-285, sBAFF/long
  • a N-terminal Flag-tag in addition to the receptor binding domain
  • R133 lies at the end of a polybasic region which is conserved between human (R-N-K-R) and mouse (R-N-R-R).
  • membrane-bound full length BAFF was expressed in 293T cells ( FIG. 2C ).
  • the 32 kDa complete BAFF and some higher molecular mass species were readily detectable in cellular extracts, but more than 95% of BAFF recovered from the supernatant corresponded to the processed 18 kDa form, indicating that BAFF was also processed when synthesized as a membrane-bound ligand.
  • a soluble BAFF was engineered (Q136-L285, sBAFF/short) whose sequence started 2 aa downstream of the processing site ( FIG. 1 B ). As predicted, the Flag-tag attached to the N-terminus of this recombinant molecule was not removed (data not shown) which allowed its purification by an anti-Flag affinity column. To test its correct folding, the purified sBAFF/short was analyzed by gel filtration where the protein eluted at an apparent molecular mass of 55 kDa ( FIG. 2D ). The sBAFF/short correctly assembles into a homotrimer (3 ⁇ 20 kDa) in agreement with the quaternary structure of other TNF family members (19). Finally, unprocessed sBAFF/long was readily expressed in bacteria, indicating that the cleavage event was specific to eukaryotic cells.
  • a sequence tagged site (STS, SHGC-36171) was found in the database which included the human BAFF sequence. This site maps to human chromosome 13, in a 9 cM interval between the markers D13S286 und D13S1315. On the cytogenetic map, this interval corresponds to 13q32-34. Of the known TNF ligand family members, only RANKL (Trance) has been localized to this chromosome (22) though quite distant to BAFF (13q14).
  • BAFF-R BAFF receptor
  • the two mouse B cell lines tested were negative using the human BAFF as a probe, although weak binding was observed on mouse splenocytes (data not shown).
  • the presence of BAFF-R on B cells was corroborated by analysis of umbilical cord and peripheral blood lymphocytes. While CD8 + and CD4 + T cells lacked BAFF-R ( FIG. 4B and data not shown), abundant staining was observed on CD19 + B cells ( FIGS. 4A and 4B ), indicating that BAFF-R is expressed on all blood B cells, including naive and memory ones.
  • Peripheral blood lymphocytes were stimulated with anti-IgM ( ⁇ ) antibodies together with fixed 293 cells stably expressing surface BAFF ( FIG. 5A ).
  • the levels of [ 3 H]thymidine incorporation induced by anti- ⁇ alone was not altered by the presence of control cells but was increased two-fold in the presence of BAFF-transfected cells ( FIG. 5B ).
  • a dose-dependent proliferation of PBL was also obtained when BAFF-transfected cells were replaced by purified sBAFF ( FIG. 5C ), indicating that BAFF does not require membrane attachment to exert its activity.
  • peripheral or cord blood B cells were preactivated by coculture with EL-4 T cells in the presence of a cytokine mixture from supernatants of PHA/PMA stimulated T cells (23). These B cells were reisolated to 98% purity and yielded a two-fold increase in Ig secretion during a secondary culture in the presence of BAFF and activated T cell cytokines as compared to cytokines alone. A very modest effect occurred in the absence of exogenous cytokines, and an intermediate (1.5-fold) effect was observed in the presence of the recombinant cytokines IL-2, IL4 and IL-10 ( FIG. 5E , F).
  • BAFF biochemical analysis of BAFF is also consistent with the typical homotrimeric structure of TNF family members.
  • BAFF exhibits the highest level of sequence similarity with APRIL which we have recently characterized as a ligand stimulating growth of various tumor cells (11).
  • APRIL Unlike TNF and LTO which are two family members with equally high homology (33% identity) and whose genes are linked on chromosome 6, APRIL and BAFF are not clustered on the same chromosome.
  • APRIL is located on chromosome 17 (J. L. B., unpublished data) whereas BAFF maps to the distal arm of human chromosome 13 (13q34).
  • Burkitt lymphomas Abnormalities in this locus were characterized in Burkitt lymphomas as the second most frequent defect (24) besides the translocation involving the myc gene into the Ig locus (25). Considering the high expression levels of BAFF-R on all Burkitt lymphoma cell lines analyzed (see Table 1), this raises the intriguing possibility that some Burkitt lymphomas may have deregulated BAFF expression, thus stimulating growth in an autocrine manner.
  • antigen-specific B lymphocytes during the different stages of the immune response is highly dependent on signals and contacts from helper T cells and antigen-presenting cells such as dendritic cells (20).
  • B lymphocytes first receive these signals early on during the immune response when they interact with T cells at the edge of the B cell follicles in lymphoid tissues, leading to their proliferation and differentiation into low affinity antibody forming cells (18).
  • some antigen-specific B cells also migrate to the B cell follicle and contribute to the formation of germinal centers, another site of B cell proliferation but also affinity maturation and generation of memory B cells and high affinity plasma cells (19).
  • CD40UCD40 interaction does not account for all contact-dependent T-cell help for B cells.
  • CD40L-deficient T cells isolated from either knock-out mice or patients with X-linked hyper IgM syndrome have been shown to sucessfully induce proliferation of B cells and their differentiation into plasma cells.
  • Studies using blocking antibodies against CD40L showed that a subset of surface IgD positive B cells isolated from human tonsils proliferate and differentiate in response to activated T cells in a CD40-independent manner.
  • TNF TNF- and surface Ig-independent stimulation of B cells.
  • CD40-deficient B cells can be stimulated to proliferate and differentiate into plasma cells by helper T cells as long as the surface Ig receptors are triggered at the same time.
  • BAFF as well as CD30L and CD40L is expressed by T cells but its originality resides in its expression by dendritic cells as well as the highly specific location of its receptor on B cells which is in contrast to CD40, CD30 and the TNF receptor which expression has been descrided on many different cell. This observation suggests independent and specific BAFF-induced functions on B cells.
  • BAFF costimulates proliferation of blood-derived B cells concomitantly with cross-linking of the B cell receptors, and, thus, independently of CD40 signalling.
  • CD19 positive B cells differentiated in vitro into a pre-plasma cell/GC-like B cell (14) we observed a costimulatory effect of BAFF on Ig secretion by these B cells in the presence of supernatant from activated T cells or a blend of IL-2, IL-4 and IL-10.
  • BAFF can signal in both naive B cells as well as GC-conunited B cells in vitro. Whether this observation will translate or not during a normal immune response will have to be addressed by proper in vivo experiments.
  • B cell growth was efficiently costimulated with recombinant soluble BAFF lacking the transmembrane domain.
  • This activity is in contrast to several TNF family members which are active only as membrane-bound ligand such as TRAIL, FasL and CD40L. Soluble forms of these ligands have poor biological activity which can be enhanced by their cross-linking, thereby mimicking the membrane-bound ligand (15).
  • cross-linking Flag-tagged sBAFF with anti-Flag antibodies or the use of membrane-bound BAFF expressed on the surface of epithelial cells did not further enhance the mitogenic activity of BAFF, suggesting that it can act systemically as a secreted cytokine, like TNF does.
  • the PCR reaction contained 1 ⁇ Pfu buffer (Stratagene, La Jola, Calif.), 0.2 mM dNTPs, 10% DMSO, 12.5 pM primers, 5 units Pfu enzyme (Stratagene) and the following primers with Not1 restriction sites 5′-TAAGAATGCGGCCGCGGAATGGATGAGTCTGCAAA-3′ [SEQ. ID. NO.: 19] and 5′-TAAGAATGCGGCCGCGGGATCACGCACTCCAGCAA-3′ [SEQ. ID. NO.: 20].
  • the template was amplified for 30 cycles at 94° C. for 1 min, 54° C. for 2 min and 72° C. for 3 min followed by a 10 min extension at 72° C.
  • This sequence corresponds to nucleotides 214 to 1171 of the GenBank file AF119383.
  • the PCR fragment was digested with Not1 and then cloned into a modified pCEP4 vector (Invitrogen, Carlsbad, Calif.).
  • the fragment containing murine BAFF was removed with Xba1 in order to include the SV40 polyA addition site sequence.
  • This fragment was cloned into a pUC based vector where the promoter sequence was added.
  • the promoter, a 1 Kb blunt Bgl2-Not1 fragment containing the human ApoE enhancer and AAT (alpha anti-trypsin) promoter was purified from the plasmid clone 540B (a kind gift from Dr.
  • RNA samples were subject to reduced SDS-PAGE analysis using a linear 12.5% gel.
  • Total RNA from mouse liver was prepared and processed for Northern Blot analysis using an isolation kit from Promega (Madison, Wis.) according to the manufacturer's guidelines.
  • BAFF transgene-specific mRNA was detected using a probe spanning the SV40 poly A tail of the transgene construct and obtained by digestion of the modified pCEP4 vector with Xba1 and BamH1. The probe recognizes a 1.8-2 Kd band corresponding to mRNA from the BAFF transgene.
  • PCR analysis of tail DNA from BAFF Tg mice was carried using 12.5 pM of the following primers 5′-GCAGTTTCACAGCGATGTCCT-3′ [SEQ. ID.
  • mice urine The presence of proteins in mouse urine was measured using Multistix 10 SG reagent strips for urinalysis (Bayer Corporation, Diagnostics Division, Elkhart, Ind.).
  • mice coli as well as mammalian cell-derived human and mouse Flag-tagged BAFF were previously described (Schneider et al., 1999). All antibodies were used according to the manufacturer's specifications.
  • PBL were purified from mouse blood as follows: mouse blood was collected in microtubes containing EDTA and was diluted 1 ⁇ 2 with PBS. Five hundred ⁇ l of diluted blood was applied on top of 1 ml of ficoll (Celardane, Hornby, Ontario, Canada) in a 4 ml glass tube, the gradient was performed at 2000 rpm for 30 min at room temperature and the interface containing the lymphocytes was collected and washed twice in PBS prior to FACS staining.
  • Spleen, bone marrow and mesenteric lymph nodes were ground into a single cell suspension in RPMI medium (Life Technologies, Inc., Grand Island, N.Y.) and washed in FACS buffer (PBS supplemented with 2% fetal calf serum (JRH Biosciences, Lenexa, Kans.).
  • FACS buffer PBS supplemented with 2% fetal calf serum (JRH Biosciences, Lenexa, Kans.).
  • Cells were first suspended in FACS buffer supplemented with the following blocking reagents: 10 ⁇ g/ml human Ig (Sandoz, Basel, Switzerland) and 10 ⁇ g/ml anti-mouse Fc blocking antibody (Pharmingen) and incubated 30 min on ice prior to staining with fluorochrome-labeled antibodies. All antibodies were diluted in FACS buffer with the blocking reagent mentioned above. Samples were analyzed using a FACScan cytofluorometer (Becton Dick
  • ELISA plates (Corning glass works, Corning, N.Y.) were coated overnight at 4° C. with a solution of 10 ⁇ g/ml of goat anti-total mouse Ig (Southern Biotechnology Associates, Inc. Birmingham, Ala.) in 50 mM sodium bicarbonate buffer pH 9.6. Plates were washed 3 times with PBS/0.1% Tween and blocked overnight with 1% gelatin in PBS. One hundred ⁇ l/well of serum serial dilutions or standard dilutions was added to the plates for 30 min at 37° C.
  • Mouse Ig were detected using 100 ⁇ l/well of a 1 ⁇ g/ml solution of an Alkaline Phosphatase (AP)-labeled goat anti-total mouse Ig (Southern Biotechnology Associates) for 30 min at 37° C. After a last wash, 3 times with PBS/0.1% Tween, the enzymatic reaction was developed using a solution of 10 ⁇ g/ml of p-nitrophenyl phosphate (Boehringer Mannheim, Indianapolis, Ind.) in 10% diethanolamine. The reaction was stopped by adding 100 ⁇ l of 3N NaOH/well.
  • AP Alkaline Phosphatase
  • optical density was measured at 405 nm using a spectrophotometer from Molecular Devices (Sunnyvale, Calif.). Standard curves were obtained using purified mouse Ig purchased from Southern Biotechnology Associates. In the case of detection of rheumatoid factors (RF), the plates were coated with normal goat Ig (Jackson ImmunoResearch laboratories, Inc., West Grove, Pa.) instead of goat anti-mouse Ig and detection of mouse Ig was performed as described above.
  • mice isotypes in the RF assay was done using AP-labeled goat anti-mouse IgA, IgM, IgG2a, IgG2b and IgG3, as well as purified mouse IgA, IgM, IgG2a, IgG2b and IgG3 for standard curves (Southern Biotechnology Associates Inc.). All statistical comparisons were performed by analysis of variance.
  • ELISA plates (Corning glass works) were coated overnight at 4° C. with 5 ⁇ g/ml of human C1q (Quidel, San Diego, Calif.) in 50 mM sodium bicarbonate buffer pH 9.6. The plates were washed 3 times with PBS/0.1% Tween. Fifty ⁇ l/well of 0.3 M EDTA was added to the plates plus 50 ⁇ l/well of serum serial dilutions or solutions of known concentrations of a standard immune complex (peroxidase-mouse anti-peroxidase (PAP) from DAKO (Carpinteria, Calif.).
  • PAP peroxidase-mouse anti-peroxidase
  • mice Ig in the immune complexes were detected using 100 ⁇ l/well of a 1 ⁇ g/ml solution of an AP-labeled goat anti-mouse Ig (Southern Biotechnology Associates, Inc.) as described above for the ELISA assays. Cryoglobulins were detected by incubating overnight at 4° C. mouse serum diluted 1/15 in water and precipitates were scored visually.
  • Anti-ssDNA were performed using NUNC-immuno Plate MaxiSorp plates (NUNC A/S, Denmark). Plates were coated overnight at 4° C. first with 100 ⁇ g/ml methylated BSA (Calbochem Corp., La Jolla, Calif.), then with 50 ⁇ g/ml grade I calf thymus DNA (Sigma, St. Louis, Mo.). The calf thymus DNA was sheared by sonication and then digested with S1 nuclease before use. For the anti-ssDNA assay, the DNA was boiled for 10 min and chilled on ice before use. After blocking, serial dilutions of the serum samples were added and incubated at room temperature for 2 h.
  • Spleen and lymph nodes were frozen in O.C.T. embedding medium (Miles, Elkhart, Ind.) and mounted for cryostat sectioning. Sections 7-10 ⁇ m thick were dried and fixed in acetone. All Ab incubations (10 ⁇ g/ml) were done for 1 hr at room temperature in a humidified box after dilution in Tris-buffered saline A (TBS-A, 0.05M Tris, 0.15M NaCl, 0.05% Tween-20 (v/v), 0.25% BSA), rinsed in TBS-B (0.05M Tris, 0.15M NaCl, 0.05% Tween-20) and fixed 1 min in methanol before initiating the enzymatic reaction.
  • TBS-A Tris-buffered saline A
  • TBS-B 0.05M Tris, 0.15M NaCl, 0.05% Tween-20
  • HRP horseradish peroxidase
  • AP alkaline phosphatase
  • DAB diaminobenzidine
  • BCIP/NBT 5-bromo-4-chloro-3-indolyl phosphate/nitro blue tetrazolium
  • Stained tissue sections were finally fixed 5 min in methanol and counter stained with Giemsa (Fluka, Buchs, Switzerland).
  • Biotin-labeled antibodies rat anti-B220, anti-CD11c, anti-syndecan-1 as well as unlabeled rat anti-CD4, anti-CD8 ⁇ and anti-CD8 ⁇ were purchased from Pharmingen.
  • Biotin-labeled peanut agglutinin (PNA) was obtained from Vector laboratories (Burlingame, Calif.).
  • HRP horse anti-rat Ig
  • HRP streptavidin
  • AP-labeled streptavidin from Southern Biotechnology Associates, Inc.
  • paraffin section were used, dewaxed and blocked using diluted horse serum from Vector (Burlingame, Calif.), followed by staining with HRP-goat anti-mouse Ig from Jackson Immunoresearch. Detection was performed as described above.
  • BAFF Transgenic (BAFF Tg) Founder Mice have an Abnormal Phenotype.
  • Hematoxylin and eosin (H&E)-stained kidney tissue sections of founder 816 showed that the morphology of glomeruli in that mouse was abnormal, whereas the rest of the kidney tissue seemed normal (data not shown).
  • Many BAFF transgenic founder mice had proteinuria (Table 2). Immunohistochemistry on spleen frozen tissue sections from mouse 816, revealed an abnormal and extensive B cell staining and reduced staining for T cells and this observation was confirmed in the progeny (see below, FIG. 12 ).
  • the transgenic mice population was found to have more lymphocytes in the blood when compared to control negative littermates, reaching values as high as 13000 lymphocytes/ ⁇ l of blood ( FIG. 7A ). In contrast, the number of granulocytes per ⁇ l of blood in both BAFF Tg mice and control mice remained within normal limits ( FIG. 7A ). Since FACS analysis, using anti-CD4 and anti-B220 antibodies, of peripheral blood cells (PBL) from 18 BAFF Tg mice issued from six different founder mice showed increased B/T ratios ( FIGS. 7B and 7C ), the elevated lymphocyte levels resulted from an expanded B cell subset.
  • PBL peripheral blood cells
  • Expanded B Cell Compartments are Composed of Mature Cells.
  • BAFF Tg mice The increased B cell compartment in BAFF Tg mice suggested that the level of total Ig in the blood of these animals might also be increased.
  • SDS-PAGE, analysis of serum from BAFF Tg mice and control littermates showed that the heavy and light chains IgG bands were at least 10 fold more intense in 3 out of 4 BAFF Tg mice compared to the control sera ( FIG. 9A ).
  • an ELISA determination on the sera from BAFF Tg mice show significantly higher total Ig levels when compared to that of the control mice ( FIG. 9B ).
  • Presence of RF can be associated with the presence of high levels of circulating immune complexes (CIC) and cryoglobulin in the blood (Jefferis, 1995).
  • CIC circulating immune complexes
  • CIC circulating immune complexes
  • CIC circulating immune complexes
  • cryoglobulin cryoglobulin in the blood
  • BAFF Tg mice also secreted anti-dsDNA, however, the levels of secretion did not always correlate with that of anti-ssDNA antibodies, as serum from BAFF Tg mouse 697-5 which did not contain detectable levels of anti-ssDNA antibodies, was clearly positive for the presence of anti-dsDNA ( FIG. 10B ). Therefore, BAFF Tg mice showing the most severe hyperglobulinemia secrete pathological levels of anti-DNA autoantibodies. Additionally, and also reminiscent of a lupus-like problem in these mice we detected immunoglobulin deposition in the kidney of six BAFF Tg mice analyzed ( FIG. 10C ), three of these mice did not secrete detectable levels anti-DNA antibodies (data not shown).
  • FIG. 12E was considerably reduced in BAFF Tg mice ( FIG. 12F ).
  • Syndecan-1-positive plasma cells were almost undetectable in the spleen from control littermates ( FIG. 12G ), yet the red pulp of BAFF Tg mice was strongly positive for syndecan-1 ( FIG. 12H ).
  • Very similar observations were made for the MLN ( FIG. 13 ).
  • the B cell areas were dramatically expanded ( FIG. 13B ) in contrast to the normal node where B cell follicles were easily recognizable at the periphery of the node under the capsule with a typical paracortical T cell zone ( FIG. 13A ).
  • BAFF Full length murine BAFF was expressed in transgenic mice using the liver-specific alpha 1 antitrypsin promoter with the Apo E enhancer as previously described.
  • C57BL/6 mice were purchased from ARC (Perth, Australia).
  • BAFF transgenic mice are maintained as heterozygotes for the transgene by backcrossing onto C57BL/6 mice.
  • BAFF Tg mice are screened for the presence of the transgene, both by PCR and southern blot analysis using genomic DNA isolated from 2-3 mm long tail snips. (MacKay et al.). We used animals from two separate lines of BAFF Tg mice issued after 10-12 backcrossings to C57BL/6.
  • Anti-human BAFF antibodies Buffy-2 (rat IgM), Buffy-5 (Rat IgG1) and A21G3.3 (mouse IgG1, K) were obtained after immunization of rats or mice with recombinant soluble human BAFF as previously detailed. (Schneider et al.). These antibodies recognize the TNF homology domain of soluble BAFF.
  • the antibody A21G3.3 was purified as follows: 500 ml of medium from hybridoma cultures were diluted 1 to 1 with 0.1 M Sodium Phosphate (pH 7.2) buffer containing 150 mM of NaCl.
  • the diluted media was loaded onto a protein-L column (Clontech, Palo Alto, Calif.) at 1 ml/minute and eluted with 0.1 M Glycine (pH 2.8). The eluted solution was neutralized with 1 M Sodium Phosphate buffer (pH 7.2). The peak fractions were confirmed by SDS-PAGE. Centricon Plus-20 (Millipore, Bedford, Mass.) was used to exchange the buffer to PBS and to concentrate the purified antibody. The purified A21G3.3 antibody was labelled with 10 ⁇ mole of EZ link sulfo-NHS-LC Biotin (Pierce, Rockford, Ill.) and incubated at room temperature for 30 minutes. The biotinylation reaction was stopped with 150 mM glycine. The sample was further purified with a desalting column to remove the free biotin (Amersham Pharmacia, Uppsala, Sweden). Flow cytometry.
  • mice were sacrificed and spleen and submaxillary glands were collected. Spleens and lymph nodes were dissociated by grinding between frosted glass slides (Menzel-Glaser, Braunschweig, Germany). Cells were filtered through a 70 ⁇ m nylon Cell Strainer (Falcon, Becton Dickinson, Franklin Lakes, N.J.) and erythrocytes (only for spleen) removed by osmotic lysis with red blood cell lysis solution (8.34 mg/ml ammonium chloride, 0.84 mg/ml sodium bicarbonate, 1 mM EDTA, pH 8.0).
  • red blood cell lysis solution 8.34 mg/ml ammonium chloride, 0.84 mg/ml sodium bicarbonate, 1 mM EDTA, pH 8.0.
  • Submaxillary glands were cut into pieces of 2-3 millimeters and incubated in a sterile collagenase solution (Roche Diagnostics, Mannheim, Germany), 1 mg/ml in PBS (Ca 2+ and Mg 2+ free), for 1 h at 37° C., until leukocytes were released from the tissue. After digestion, the cell suspension was filtered through a 70 ⁇ m nylon cell strainer (Falcon) and filtered cells were washed twice with PBS. Leukocytes obtained from spleens or submaxillary glands were resuspended in FACS buffer (1% BSA, 0.05% sodium azide in PBS) at a concentration of 5 ⁇ 10 6 cells/ml.
  • FACS buffer 1% BSA, 0.05% sodium azide in PBS
  • Cy5-conjugated anti-IgM antibody was purchased from Jackson ImmunoResearch laboratories Inc. (West Grove, Pa.). FITC-labelled antibodies were used diluted 1/100 whereas other fluorochrome-labelled antibodies were used at 1/200 final dilution. For flow cytometry we acquired 30,000-100,000 events per sample. Data was collected on a FACSCalibur flow cytometer and analysed using CELLQuesTM software (Becton Dickinson).
  • Spleen (removed comma) and submaxillary glands were collected from both control and BAFF Tg mice.
  • Tissues were either frozen in OCT compound (Tissue-Tek, Sakura Finetek, Torrance, Calif., USA) or fixed in 10% buffered formalin and embedded in paraffin.
  • Biopsies of human parotid glands were processed into paraffin blocks by pathologists at the Flinders Medical center in Sydney. Paraffin sections, 5 ⁇ m thick, were re-hydrated in successive baths of xylene, 100% ethanol and H 2 O. Slides were cooked under pressure in citrate buffer (8.2 mM trisodium citrate, 1.7 mM citrate acid, pH 6.0).
  • mice were anaesthetized and injected ip with 50 ⁇ g of sterile pilocarpine in PBS (Sigma, St Louis, Mo.) per 100 g body weight. After 4 min, saliva was collected for 5 min on a cotton swab. The weight of the cotton swab was measured before and after saliva collection. The amount of saliva collected was normalized to 0 g of saliva per g of body weight. Patients with primary SS and sera.
  • Sera from patients were diluted 1/10 and pre-cleared from human Ig on protein-A-Sepharose beads (10% beads (pelleted beads) v/v, Amersham Pharmacia) overnight at 4° C.
  • ELISA plates (NUNC Nalge International, Rochester, N.Y.) were coated with 2 ⁇ g/ml rat anti-human BAFF antibody (Buffy-5), overnight at 4° C. Following blocking, serial dilutions of the pre-cleared sera were added, followed by the detection antibody, biotin-conjugated mouse anti-human BAFF (0.5 ⁇ g/ml, clone A21G3.3).
  • FIG. 15A , B and C Unusual aggregates of cells organised in diffused foci (small lymphocytes in sinusoids) ( FIG. 15A , B and C) were detected in these tumors ( FIG. 15B ), as well as in the large leukocyte infiltrates found in salivary glands of 4 independent BAFF Tg mice (two examples are shown in FIGS. 15A and C).
  • Cells in these infiltrates were B-lymphoid cells expressing high levels of B220 ( FIG. 15D ), however they were not plasma cells since they were not positive for the plasma cell marker, syndecan-1 ( FIG. 15E ).
  • a consulting pathologist indicated that because no lympho-epithelial aggregates were seen, the infiltrating B cells could not be definitively classified as malignant.
  • Studies to examine the clonality of these unusual B cells are in progress to determine whether they are simply tumor-like clusters of lymphoid cells that do not meet criteria for malignancy or cells with real neoplastic potential.
  • mice Seven BAFF Tg mice that had significantly larger submaxillary glands compared to age-matched control mice were selected for a detailed assessment of infiltrating cells.
  • One gland from each mouse was digested with collagenase, after which mononuclear cells were purified and analysed by flow cytometry. Absolute counts of various cell types showed a consistent and significant increase in the number of B-lymphocytes infiltrating submaxillary glands of aging BAFF Tg mice (Table 1). Numbers of T cells, NK cells, macrophages and granulocytes also increased, however a large variation was seen between animals (Table 1).
  • Tissues were digested and leukocytes purified as described in materials and methods. The Total number of leukocytes per gland was counted using an hemocytometer. Cells were stained with anti-B220 (B cells), anti-CD4 and anti-CD8 (T cells), anti-NK1.1 (NK cells), anti-Mac1 (macrophages) and anti-GR1 (granulocytes) fluorochrome-labelled antibodies. Flow cytometry analysis provided percentage values for each subpopulation # of cells and absolute numbers were calculated relative to the total number of cells collected per gland. ND: not detected, —: not done. Only significant p values are indicated.
  • a flow cytometric profiling analysis of B cells infiltrating the submaxillary glands of BAFF Tg mice revealed two B cell subsets based on their level of expression of B220 ( FIG. 16A ).
  • Analysis of the B-1 B cell population in the B220 low/intermediate (B220 lo/int ) gate revealed a 3-fold increase of B-1b (CD5 ⁇ ) B cells and presence of B-1a (CD5 + ) B cells not detected in control mice ( FIG. 16B ).
  • B220 high (B220 hi ) gate cells were further analysed based on their level of IgM expression.
  • the IgM dull (IgM dull ) subset of B cells is present in both control and BAFF Tg mice, however this population is larger in BAFF Tg mice compared to control animals ( FIG. 16B ).
  • the IgM dull subset in both control and BAFF Tg mice contains B-2-like cells, which are CD21 lo/int , IgD + and CD23 + (data not shown).
  • the B220 hi /IgM hi B cell subset is greatly increased in BAFF Tg mice compared to control mice ( FIG. 16B ).
  • the IgM hi B cells found in BAFF Tg mice are phenotypically different than IgM hi B cells from control mice.
  • the small IgM hi B cell subset detected in control mice had characteristics of B-2 B cells (CD21 lo/int , IgD + and CD23+, add HSA and L-selectin ( FIG. 16D, 16E ), whereas the large IgM hi B cell subset of BAFF Tg mice had no counterpart in control mice and resembled MZ B cells in many respects. These cells were IgD lo , CD21 hi , CD23 lol ⁇ , CD1 hi , HSA ++ ( FIG. 16E ) and L-selectin lo ( FIG. 16D ). These cells also express lower levels of CD43 when compared to B-1 B cells ( FIG. 16C ).
  • BAFF levels in sera of patients suffering from this disease were measured using a human BAFF-specific ELISA assay. This assay showed that at least 15 patients out of 41 (36%) clearly had higher levels of serum BAFF when compared to healthy individuals ( FIG. 18A ). Two patients had very high BAFF levels (over 200 ng/ml, FIG. 18A ). BAFF levels in patients' sera did not correlate with the levels of total IgG or RF ( FIG. 18B and C, respectively). These BAFF levels also did not correlate with the presence of precipitins such as anti-Ro and/or anti-La ( FIG. 18D ). We confirmed human BAFF levels in patients' sera using BAFF-specific immunoprecipitation procedures followed by western blotting techniques (data not shown).

Abstract

The invention provides methods for treating or preventing disorders associated with expression of BAFF comprising BAFF and fragments thereof, antibodies, agonists and antagonists.

Description

    RELATED APPLICATIONS
  • This application is a continuation-in-part of U.S. application Ser. No. 09/911,777, filed Jul. 24, 2001, which claims priority to International Application No. PCT/US00/01788 filed Jan. 25, 2000, which claims priority to U.S. Ser. No. 60/117,169 filed on Jan. 25, 1999 and U.S. Ser. No. 60/143,228 filed Jul. 9, 2001. The entire disclosures of the aforesaid patent applications are incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The present invention relates to the use of a ligand, BAFF, a β-cell activating factor belonging to the Tumor Necrosis Family and its blocking agents to either stimulate or inhibit the expression of B-cells and immunoglobulins. This protein and its receptor may have anti-cancer and/or immunoregulatory applications as well as uses for the treatment of immunosuppressive disorders such as HIV. Specifically, the ligand and its blocking agents may play a role in the development of hypertension and its related disorders. Furthermore, cells transfected with the gene for this ligand may be used in gene therapy to treat tumors, autoimmune diseases or inherited genetic disorders involving B-cells. Blocking agents, such as recombinant variants or antibodies specific to the ligand or its receptor, may have immunoregulatory applications as well. Use of BAFF as a B-cell stimulator for immune suppressed diseases including for example uses for patients undergoing organ transplantation (i.e. bone marrow transplant) as well as recovering from cancer treatments to stimulate production of B-cells are contemplated. Use of BAFF as an adjuvant and or costimulator to boast and or restore B cells levels to approximate normal levels are also contemplated.
  • BACKGROUND OF THE INVENTION
  • The tumor-necrosis factor (TNF)-related cytokines are mediators of host defense and immune regulation. Members of this family exist in membrane-anchored forms, acting locally through cell-to-cell contact, or as secreted proteins capable of diffusing to more distant targets. A parallel family of receptors signals the presence of these molecules leading to the initiation of cell death or cellular proliferation and differentiation in the target tissue. Presently, the TNF family of ligands and receptors has at least 11 recognized receptor-ligand pairs, including: TNF:TNF-R; LT-α:TNF-R; LT-α/β:LT-β-R; FasL:Fas; CD40L:CD40; CD30L:CD30; CD27L:CD27; OX40L:OX40 and 4-1BBL:4-1BB. The DNA sequences encoding these ligands have only about 25% to about 30% identity in even the most related cases, although the amino acid relatedness is about 50%.
  • The defining feature of this family of cytokine receptors is found in the cysteine rich extracellular domain initially revealed by the molecular cloning of two distinct TNF receptors. This family of genes encodes glycoproteins characteristic of Type I transmembrane proteins with an extracellular ligand binding domain, a single membrane spanning region and a cytoplasmic region involved in activating cellular functions. The cysteine-rich ligand binding region exhibits a tightly knit disulfide linked core domain, which, depending upon the particular family member, is repeated multiple times. Most receptors have four domains, although there may be as few as three, or as many as six.
  • Proteins in the TNF family of ligands are characterized by a short N-terminal stretch of normally short hydrophilic amino acids, often containing several lysine or arginine residues thought to serve as stop transfer sequences. Next follows a transmembrane region and an extracellular region of variable length, that separates the C-terminal receptor binding domain from the membrane. This region is sometimes referred to as the “stalk”. The C-terminal binding region comprises the bulk of the protein, and often, but not always, contains glycosylation sites. These genes lack the classic signal sequences characteristic of type I membrane proteins, type I membrane proteins with the C terminus lying outside the cell, and a short N-terminal domain residing in the cytoplasm. In some cases, e.g., TNF and LT-α, cleavage in the stalk region can occur early during protein processing and the ligand is then found primarily in secreted form. Most ligands, however, exist in a membrane form, mediating localized signaling.
  • The structure of these ligands has been well-defined by crystallographic analyses of TNF, LT-α, and CD40L. TNF and lymphotoxin-I (LT-1) are both structured into a sandwich of two anti-parallel β-pleated sheets with the “jelly roll” or Greek key topology. The rms deviation between the Cα and β residues is 0.61 C, suggesting a high degree of similarity in their molecular topography. A structural feature emerging from molecular studies of CD40L, TNF and LT-α is the propensity to assemble into oligomeric complexes. Intrinsic to the oligomeric structure is the formation of the receptor binding site at the junction between the neighboring subunits creating a multivalent ligand. The quaternary structures of TNF, CD40L and LT-α have been shown to exist as trimers by analysis of their crystal structures. Many of the amino acids conserved between the different ligands are in stretches of the scaffold β-sheet. It is likely that the basic sandwich structure is preserved in all of these molecules, since portions of these scaffold sequences are conserved across the various family members. The quaternary structure may also be maintained since the subunit conformation is likely to remain similar.
  • TNF family members can best be described as master switches in the immune system controlling both cell survival and differentiation. Only TNF and LTα are currently recognized as secreted cytokines contrasting with the other predominantly membrane anchored members of the TNF family. While a membrane form of TNF has been well-characterized and is likely to have unique biological roles, secreted TNF functions as a general alarm signaling to cells more distant from the site of the triggering event. Thus TNF secretion can amplify an event leading to the well-described changes in the vasculature lining and the inflammatory state of cells. In contrast, the membrane bound members of the family send signals though the TNF type receptors only to cells in direct contact. For example T cells provide CD40 mediated “help” only to those B cells brought into direct contact via cognate TCR interactions. Similar cell-cell contact limitations on the ability to induce cell death apply to the well-studied Fas system.
  • It appears that one can segregate the TNF ligands into three groups based on their ability to induce cell death. First, TNF, Fas ligand and TRAIL can efficiently induce cell death in many lines and their receptors mostly likely have good canonical death domains. Presumably the ligand to DR-3 (TRAMP/WSL-1) would also all into this category. Next there are those ligands which trigger a weaker death signal limited to few cell types and TWEAK, CD30 ligand and LTa1b2 are examples of this class. How this group can trigger cell death in the absence of a canonical death domain is an interesting question and suggests that a separate weaker death signaling mechanism exists. Lastly, there are those members that cannot efficiently deliver a death signal. Probably all groups can have antiproliferative effects on some cell types consequent to inducing cell differentiation e.g. CD40. Funakoshi et al. (1994).
  • The TNF family has grown dramatically in recent years to encompass at least 11 different signaling pathways involving regulation of the immune system. The widespread expression patterns of TWEAK and TRAIL indicate that there is still more functional variety to be uncovered in this family. This aspect has been especially highlighted recently in the discovery of two receptors that affect the ability of rous sacroma and herpes simplex virus to replicate as well as the historical observations that TNF has anti-viral activity and pox viruses encode for decoy TNF receptors. Brojatsch et al. (1996); Montgomery et al. (1996); Smith et al. (1994), 76 Cell 959-962; Vassalli et al. (1992), 10 Immunol. 411-452.
  • TNF is a mediator of septic shock and cachexia, and is involved in the regulation of hematopoietic cell development. It appears to play a major role as a mediator of inflammation and defense against bacterial, viral and parasitic infections as well as having antitumor activity. TNF is also involved in different autoimmune diseases. TNF may be produced by several types of cells, including macrophages, fibroblasts, T cells and natural killer cells. TNF binds to two different receptors, each acting through specific intracellular signaling molecules, thus resulting in different effects of TNF. TNF can exist either as a membrane bound form or as a soluble secreted cytokine.
  • LT-I shares many activities with TNF, i.e. binding to the TNF receptors, but unlike TNF, appears to be secreted primarily by activated T cells and some β-lymphoblastoid tumors. The heteromeric complex of LT-α and LT-β is a membrane bound complex which binds to the LT-β receptor. The LT system (LTs and LT-R) appears to be involved in the development of peripheral lymphoid organs since genetic disruption of LT-β leads to disorganization of T and B cells in the spleen and an absence of lymph nodes. The LT-β system is also involved in cell death of some adenocarcinoma cell lines.
  • Fas-L, another member of the TNF family, is expressed predominantly on activated T cells. It induces the death of cells bearing its receptor, including tumor cells and HIV-infected cells, by a mechanism known as programmed cell death or apoptosis. Furthermore, deficiencies in either Fas or Fas-L may lead to lymphoproliferative disorders, confirming the role of the Fas system in the regulation of immune responses. The Fas system is also involved in liver damage resulting from hepatitis chronic infection and in autoimmunity in HIV-infected patients. The Fas system is also involved in T-cell destruction in HIV patients. TRAIL, another member of this family, also seems to be involved in the death of a wide variety of transformed cell lines of diverse origin.
  • CD40-L, another member of the TNF family, is expressed on T cells and induces the regulation of CD40-bearing B cells. Furthermore, alterations in the CD40-L gene result in a disease known as X-linked hyper-IgM syndrome. The CD40 system is also involved in different autoimmune diseases and CD40-L is known to have antiviral properties. Although the CD40 system is involved in the rescue of apoptotic B cells, in non-immune cells it induces apoptosis. Many additional lymphocyte members of the TNF family are also involved in costimulation.
  • Generally, the members of the TNF family have fundamental regulatory roles in controlling the immune system and activating acute host defense systems. Given the current progress in manipulating members of the TNF family for therapeutic benefit, it is likely that members of this family may provide unique means to control disease. Some of the ligands of this family can directly induce the apoptotic death of many transformed cells e.g. LT, TNF, Fas ligand and TRAIL. Nagata (1997) 88 Cell 355-365. Fas and possibly TNF and CD30 receptor activation can induce cell death in nontransformed lymphocytes which may play an immunoregulatory function. Amakawa et al. (1996) 84 Cell 551-562; Nagata (1997) 88 Cell 355-365; Sytwu et al. (1996); Zheng et al. (1995) 377 Nature 348-351. In general, death is triggered following the aggregation of death domains which reside on the cytoplasmic side of the TNF receptors. The death domain orchestrates the assembly of various signal transduction components which result in the activation of the caspase cascade. Nagata (1997) 88 Cell 355-365. Some receptors lack canonical death domains, e.g. LTb receptor and CD30 (Browning et al. (1996); Lee et al. (1996)) yet can induce cell death, albeit more weakly. It is likely that these receptors function primarily to induce cell differentiation and the death is an aberrant consequence in some transformed cell lines, although this picture is unclear as studies on the CD30 null mouse suggest a death role in negative selection in the thymus. Amakawa et al. (1996) 84 Cell 551-562. Conversely, signaling through other pathways such as CD40 is required to maintain cell survival. Thus, there is a need to identify and characterize additional molecules which are members of the TNF family thereby providing additional means of controlling diseased and manipulating the immune system.
  • Sjögren's syndrome (SS) is a chronic inflammatory disorder characterized by the destruction of exocrine glands such as salivary and lacrimal glands, leading to symptoms of dry mouth (xerostomia) and eyes (keratoconjuncitivitis sicca). Jonsson et al. (2000) Sjogren's syndrome in Arthritis and allied conditions 1826-1849. SS is regarded as an autoimmune disease characterized by the presence of large mononuclear cell infiltrates in exocrine glands, B cell hyper-reactivity and various serum autoantibodies. Jonsson et al. (2000); Manoussakis et al. (1998) Sjogren's sydrome in The autoimmune diseases, Academic Press, 381-404. SS can develop alone or in association with other autoimmune disorders such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Jonnson et al. (2000); Manoussakis et al. (1998).
  • Abnormal B cell activity is a predominant feature of SS, which is manifested by massive polyclonal B cell activation and elevated secretion of autoantibodies such as rheumatoid factors (RF), anti-Ro (SS-A), anti-La (SS-B) and anti-λ-fodrin autoantibodies. Jonnson et al. (2000); Manoussakis et al. (1998); MacSween et al. (1967) Ann. Rheum. Dis. 26:402-411; Haneji et al. (1997) Science 276; 604-607. Intense B cell activity such as germinal center reactions occur in exocrine glands of some patients, placing them in a high risk category for the development of lymphomas. Jonsson et al. (2000); Stott et al. (1998) J. Clin. Invest. 102:938-946. However, the role of B cells and autoantibodies in the pathogenesis of SS still remains unclear.
  • SUMMARY OF THE INVENTION
  • Here we characterize the functional properties of a new ligand of the TNF cytokine-family. The new ligand, termed BAFF (B cell activating factor belonging to the TNF family), appears to be expressed by T cells and dendritic cells for the purpose of B-cell co-stimulation and may therefore play an important role in the control of B cell function. In addition, we have generated transgenic mice overexpressing BAFF under the control of a liver-specific promoter. These mice have excessive numbers of mature B cells, spontaneous germinal center reactions, secrete autoantibodies, and have high plasma cell numbers in secondary lymphoid organs and Ig deposition in the kidney.
  • The BAFF Tg mice develop as they age a secondary condition to their lupus-like disease, showing interesting similarities with that of SS in humans. We also identified a new and potentially pathogenic B cell population with MZ-like features infiltrating salivary glands of BAFF Tg mice.
  • Accordingly, the present invention is directed to the use of BAFF-ligands, blocking agents and antibodies for the ligand, to either stimulate or inhibit the growth of B-cells and the secretion of immunoglobulin. The claimed invention may be used for therapeutic applications in numerous diseases and disorders, as discussed in more detail below, as well as to obtain information about, and manipulate, the immune system and its processes. Further, this invention can be used as a method of stimulating or inhibiting the growth of B-cells and the secretion of immunoglobulins. BAFF associated molecules, as described by this invention, may also have utility in the treatment of autoimmune diseases, disorders relating to B-cell proliferation and maturation, BAFF ligand regulation and inflammation. The invention may be involved in the regulation or prevention of hypertension and hypertension-related disorders of the renal and cardiovascular tissue.
  • Additional features and advantages of the invention will be set forth in the description which follows, and in part will be apparent from the description, or may be learned by practice of the invention. The objectives and other advantages of the invention will be realized and attained by the methods particularly pointed out in the written description and claims hereof, as well as in the appended drawings.
  • Thus, to achieve these and other advantages, and in accordance with the purpose of the invention, as embodied and broadly described herein, the invention includes a method of effecting B-cell growth and secretion of immunoglobulins through the administration of various BAFF ligands and related molecules.
  • The invention also contemplates stimulating B-cell growth through the use of BAFF ligands or active fragments of the polypeptide. The polypeptide may be use alone or with a CD40 ligand or an anti-murine antibody.
  • In other embodiments, the invention relates to methods of stimulation of dendritic cell-induced B-cell growth and maturation through the use of BAFF ligands or active fragments of BAFF. Again, the polypeptide may be used alone or with CD40 ligand or anti-μ antibodies.
  • In other embodiments, blocking agents of BAFF and the BAFF receptor have been used to inhibit B-cell growth and immunoglobulin secretion. These agents can be inoperative, recombinant BAFF, BAFF specific antibodies, BAFF-receptor specific antibodies or an anti-BAFF ligand molecule.
  • In yet other embodiments, the invention relates to the use of BAFF, BAFF related molecules and BAFF blocking agents to treat hypertension, hypertension related disorders, immune disorders, autoimmune diseases, inflammation and B-cell lympho-proliferate disorders. In another aspect, the invention relates to the use of BAFF, BAFF-related molecules and BAFF blocking agents to treat Sjogren's syndrome.
  • In a preferred embodiment, the invention relates to methods for treating or reducing the advancement, severity or effects of Sjogren's syndrome in a patient by administering a pharmaceutical preparation comprising a therapeutically effective amount of a BAFF blocking agent and a pharmaceutically acceptable carrier. In some embodiments, the BAFF blocking agent may be a soluble BAFF receptor molecule, an antibody directed against BAFF-ligand or an antibody directed against a BAFF receptor. The BAFF receptor may be BCMA, TACI, or BAFF R.
  • The invention encompasses the use of BAFF and BAFF-related molecules as either agonists or antagonists in effecting immune responses by effecting the growth and/or maturation of B-cells and secretion of immunoglobulin.
  • The invention relates in other embodiments to soluble constructs comprising BAFF which may be used to directly trigger BAFF mediated pharmacological events. Such events may have useful therapeutic benefits in the treatment of cancer, tumors or the manipulation of the immune system to treat immunologic diseases.
  • Additionally, in other embodiments the claimed invention relates to antibodies directed against BAFF ligand, which can be used, for example, for the treatment of cancers, and manipulation of the immune system to treat immunologic disease.
  • In yet other embodiments the invention relates to methods of gene therapy using the genes for BAFF.
  • The pharmaceutical preparations of the invention may, optionally, include pharmaceutically acceptable carriers, adjuvants, fillers, or other pharmaceutical compositions, and may be administered in any of the numerous forms or routes known in the art.
  • It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory, and are intended to provide further explanation of the invention as claimed.
  • The accompanying drawings are included to provide a further understanding of the invention, and are incorporated in, and constitute a part of this specification, illustrate several embodiments of the invention, and together with the description serve to explain the principles of the invention.
  • DESCRIPTION OF THE DRAWINGS
  • FIG. 1 (A) depicts the predicted amino acid sequence of human [SEQ. ID. NO.: 1] and mouse BAFF [SEQ. ID. NO.:2]. The predicted transmembrane domain (TMD, dashed line), the potential N-linked glycosylation sites (stars) and the natural processing site of human BAFF (arrow) are indicated. The double line above hBAFF indicates the sequence obtained by Edman degradation of the processed form of BAFF. (B) Depicts a comparison of the extracellular protein sequence of BAFF [SEQ. ID. NO.: 3] and some members of the TNF ligand family [SEQ. ID. NO.: 4 (hAPRIL); SEQ. ID. NO.: 5 (hTNF alpha); SEQ. ID. NO.: 6 (hFasL); SEQ. ID. NO.: 7 (hLT alpha); SEQ. ID. NO.: 8 (hRANKL)]. Identical and homologous residues are represented in black and shaded boxes, respectively. (C) Depicts dendrogram of TNF family ligands.
  • FIG. 2 is a schematic characterization of recombinant BAFF (A) Schematic representation of recombinant BAFF constructs. Soluble recombinant BAFFs starting at Leu83 and Gln136 are expressed fused to a N-terminal Flag tag and a 6 amino acid linker. The long form is cleaved between Arg133 and Ala134 (arrow) in 293 T cells, to yield a processed form of BAFF. Asn124 and Asn242 belong to N-glycosylation consensus sites. N-linked glycan present on Asn124 is shown as a Y. TMD: transmembrane domain. (B) Peptide N-glycanase F (PNGase F) treatment of recombinant BAFF. Concentrated supernatants containing Flag-tagged BAFFs and APRIL were deglycosylated and analyzed by Western blotting using polyclonal anti-BAFF antibodies or anti-Flag M2, as indicated. All bands except processed BAFF also reacted with anti-Flag M2 (data not shown). (C) Full length BAFF is processed to a soluble form. 293T cells were transiently transfected with full length BAFF. Transfected cells and their concentrated supernatants were analyzed by Western blotting using polyclonal anti-BAFF antibodies. Supernatants corresponding to 10× the amount of cells were loaded onto the gel. (D) Size exclusion chromatography of soluble BAFF on Superdex-200. Concentrated supernatants containing soluble BAFF/short were fractionated on a Superdex-200 column and the eluted fractions analyzed by Western blotting using anti-Flag M2 antibody. The migration positions of the molecular mass markers (in kDa) are indicated on the left-hand side for SDS-PAGE and at the top of the figure for size exclusion chromatography.
  • FIG. 3 depicts expression of BAFF (A) Northern blots (2 μg poly A+ RNA per lane) of various human tissues were probed with BAFF antisense mRNA. (B) Reverse transcriptase amplification of BAFF, IL-2 receptor alpha chain and actin from RNA of purified blood T cells at various time points of PHA activation, E-rosetting negative blood cells (B cells and monocytes), in vitro derived immature dendritic cells, 293 cells, and 293 cells sterilely transfected with full length BAFF (293-BAFF). Control amplifications were performed in the absence of added cDNA. IL-2 receptor alpha chain was amplified as a marker of T cell activation.
  • FIG. 4 depicts BAFF binding to mature B cells. (A) Binding of soluble BAFF to BJAB and Jurkat cell lines, and to purified CD19+ cells of cord blood. Cells were stained with the indicated amount (in ng/50 μl) of Flag-BAFF and analyzed by flow cytometry. (B) Binding of soluble BAFF to PBLS. PBLs were stained with anti-CD8-FITC or with anti-CD19-FITC (horizontal axis) and with Flag-BAFF plus M2-biotin and avidin-PE (vertical axis). Flag-BAFF was omitted in controls.
  • FIG. 5 depicts BAFF costimulates B cell proliferation. (A) Surface expression of BAFF in stably transfected 293 cells. 293-BAFF and 293 wild-type cells were stained with anti-BAFF mAb 43.9 and analyzed by flow cytometry. (B) Costimulation of PBLs by 293-BAFF cells. PBLs (105/well) were incubated with 15.000 glutaraldehyde-fixed 293 cells (293 wt or 293-BAFF) in the presence or absence of anti-B cell receptor antibody (anti-μ). Fixed 293 cells alone incorporated 100 cpm. (C) Dose dependent costimulation of PBL proliferation by soluble BAFF in the presence of anti-μ. Proliferation was determined after 72 h incubation by [3H]-thymidine incorporation. Controls include cells treated with BAFF alone, with heat-denatured BAFF or with an irrelevant isotype matched antibody in place of anti-μ. (D) Comparison of (co)stimulatory effects of sCD40L and sBAFF on PBL proliferation. Experiment was performed as describe in panel C. (EAFF costimulates Ig secretion of preactivated human B cells. Purified CD19+ B cells were activated by coculture with EL-4 T cells and activated T cell supernatants for 5-6 d, then re-isolated and cultured for another 7 days in the presence of medium only (−) or containing 5% activated T cell supernatants (T-SUP) or a blend of cytokines (IL-2, IL-4, IL-10). The columns represent means of Ig concentrations for cultures with or without 1 μg/ml BAFF. Means±SD in terms of “fold increase” were 1.23±0.11 for medium only, 2.06±0.18 with T cell supernatants (4 experiments) and 1.45±0.06 with IL-2, IL-4 and IL-10 (2 experiments). These were performed with peripheral blood (3 experiments) or cord blood B cells (one experiment; 2.3 fold increase with T cell supernatants, 1.5 fold increase with IL-2, IL-4 and IL-10). (F) Dose-response curve for the effect of BAFF in cultures with T cell supernatants, as shown in panel D. Mean±SD of 3 experiments.
  • FIG. 6 depicts that BAFF acts as a cofactor for B cell proliferation. The proliferation of human PBL was measured alone (500 cpm), with the presence of BAFF ligand alone, with the presence of goat anti-murine (mu) alone, and with both BAFF ligand and anti-mu. The combination of both anti-mu and BAFF significantly raised proliferation of PBL as the concentration of BAFF increased suggesting BAFF's cofactor characteristics.
  • FIG. 7 depicts increased B cell numbers in BAFF Tg mice.
  • (A) Increased lymphocytes counts in BAFF Tg mice. The graph compares 12 control littermates (left panel) with 12 BAFF Tg mice (right panel). Lymphocytes counts are shown with circles and granulocytes (including neutrophils, eosinophils, basophils) with diamonds.
  • (B) Increased proportion of B cells in PBL from BAFF Tg mice. PBL were stained with both anti-B220-FITC and anti-CD4-PE for FACS analysis and gated on live cells using the forward side scatter. Percentages of CD4 and B220 positive cells are indicated. One control mouse (left) and two BAFF Tg mice (right) are shown and the results were representative of 7 animals analysed in each group.
  • (C) FACS analysis of the ratio of B to T cells in PBL. The difference between control animals and BAFF Tg mice in (A) and (C) was statistically significant (P<0.001).
  • (D) Increased MHC class II expression on B cells from BAFF Tg mice PBL. MHC class II expression was analysed by FACS.
  • (E) Increased Bcl-2 expression in B cells from BAFF Tg mice PBL. Bcl-2 expression was measured by intracytoplasmic staining and cells were analysed by FACS. In both (D) and (E) Live cells were gated on the forward side scatter. Four control littermates (white bars) and 4 BAFF Tg mice are shown and are representative of at least 12 animals analysed for each group. MFI: mean of fluorescence intensity. The difference between control animals and BAFF Tg mice was statistically significant (P<0.005).
  • (F) Increased expression of effector T cells in BAFF Tg mice. PBL were stained with anti-CD4-Cychrome, anti-CD44-FITC and anti-L selectin-PE. Are shown CD4+-gated cells. Percentages of CD44hi/L-selectinlo cells are indicated. One control mouse (left) and two BAFF Tg mice (right) are shown and the results were representative of 8 animals analysed in each group.
  • FIG. 8 depicts increased B cell compartments in the spleen but not in the bone marrow of BAFF Tg mice.
  • (A) FACS staining for mature B cells using both anti-IgM-FITC and anti-B220-PE, in spleen (top panel), bone marrow (medium panel) and MLN (bottom panel). Percentages of B220+/IgM+mature B cells are indicated.
  • (B) FACS staining for preB cells (B220+/CD43−) and proB cells (B220+/CD43+) in the bone marrow using anti-CD43-FITC, anti-B220-Cy-chrome and anti-IgM-PE simultaneously. Are shown cells gated on the IgM negative population. Percentages of preB cells (B220+/CD43−) and proB cells (B220+/CD43+) cells are indicated.
  • For all figures (A and B) one control mouse (left) and two BAFF Tg mice (right) are shown and results are representative of 7 animals analysed for each group.
  • FIG. 9 depicts increased Ig, RF and CIC levels in BAFF Tg mice
  • (A) SDS-PAGE of two control sera (−) and 4 sera from BAFF Tg mice (+) side by side with the indicated amount of a purified mouse IgG for reference. The intensity of the albumin band in similar in all lanes indicating that the material loaded on the gel is equivalent for each sample. ELISA-based analysis of total mouse Ig (B), RF (C) and CIC (D) in the sera of 19 control littermates (white bars) and 21 BAFF Tg mice (Black bars). In the absence of a proper RF control, the titer (log base 2) for RF is defined as the dilution of the sera giving an O.D. 3 times higher than that of background. The quantity of CIC is defined as the quantity of PAP required to generate an O.D. equivalent to that obtained with the tested serum. The difference between control animals and BAFF Tg mice was statistically significant (P<0.001 in (B) and (C), P<0.003 in (D)).
  • FIG. 10 depicts the presence of anti-ssDNA and anti-dsDNA autoantibodies in some BAFF Tg mice.
  • (A) Analysis by ELISA of anti-ssDNA autoantibodies in 19 control littermates (gray bars) and 21 BAFF Tg mice (black bars).
  • (B) Analysis by ELISA of anti-ssDNA autoantibodies in 5 control littermates and the 5 animals showing levels of anti-ssDNA autoantibodies from (A).
  • (C) Paraffin sections of kidneys from a control mouse (left) and a BAFF Tg mouse (right), stained with goat anti-mouse Ig-HRP. Ig deposition is shown by a brown staining. These pictures are representative of 6 BAFF Tg mice analysed.
  • FIG. 11 depicts enlarged Peyer's patches in BAFF Tg mice. Photography of Peyers patches (indicated with an arrow) on the small intestine of a control mouse (left) and a BAFF Tg mouse (right). This pictures is representative of at least 12 mice sacrificed for each group. Magnification
  • FIG. 12 depicts disrupted T and B cell organization, intense germinal center reactions, decreased number of dendritic cells and increased number of plasma cells in the spleen of BAFF Tg mice.
  • A control mouse is shown in A, C, E and G and a BAFF Tg in B, D, F, and H. B cells are blue and T cells brown (A and B). Germinal centers are shown with an arrow (C and D). Only few residual germinal centers are seen in control mice (C). CD11c positive dendritic cells are brown and appear in the T cell zone, bridging channels and the marginal zone (E). Very few are present in BAFF Tg mice (F). Syndecan-1-positive plasma cells were only detectable in the red pulp of BAFF Tg mice (H) but not control mice (G).
  • These pictures are representative of at least 12 BAFF Tg mice analysed and 12 control mice. The magnification is 100× for all pictures except C and D which are 50×. B: B cell follicle, T: PALS, WP: white pulp, RP: red pulp.
  • FIG. 13 depicts disrupted T and B cells organization, intense germinal center reactions and large number of plasma cells in the MLN of BAFF Tg mice.
  • The control mouse is shown in A, C, E and G and the BAFF Tg mouse is shown in B, D, F, and H. The immunohistochemistry was performed as described in FIG. 6. T and B cell staining is shown in A and B, germinal centers in C and D, dendritic cells E and F and plasma cells in G and H. GC: germinal center. Magnification 100×.
  • FIG. 14 depicts enlarged and inflamed salivary glands BAFF Tg mice. (A) Mice 15-17 months old, 4 control littermates (white bar) and 4 BAFF Tg mice (grey bar) were sacrificed the same day for organ collection. Both right and left submaxillary glands were collected and weighted. The data shows the mean t standard deviation of weight for both glands. These results are representative of at least 4 separate groups of age-matched animals dissected. (B) Paraffin sections of submaxillary glands from a control littermate (left panel) and two BAFF Tg mice (middle and right panel) were stained with H&E. Arrows indicate ducts and acinar cells in the left and right panels. The arrow in the middie panel is showing acinar destruction. White stars indicate periductal infiltrates (foci). Magnification 100×. (C) Paraffin sections of submaxillary glands from 7 control mice and 22 BAFF Tg mice (age 12-17 months) were prepared as shown in (B) and scored for the disease as described in materials and methods. The mean score of disease is indicated with a bar. *p<0.05, **p<0.03.
  • FIG. 15 depicts tumor-like clusters of B-lymphoid cells in a submaxillary tumor or infiltrates present in submaxillary glands of BAFF Tg mice. Frozen sections of submaxillary glands (A and C) or a submaxillary tumor (B, D and E) were stained with H&E (A, B and C) or Biotin-labelled anti-mouse B220 (D) or anti-mouse syndecan-1 (E), follwed by Streptavidin-HRP brown staining in D and E). Arrows in A, B, C and D indicate the tumor-like clusters, arrow in E shows the presence of syndecan-1 positive plasma cells whereas the stars indicate the location of tumor-like clusters. Note that tumor-like clusters and plasma cells do not colocalize. Panels (A) and (C) are representative of 4 animals in which these clusters were detected as cells infiltrating submaxillary glands. Panels (B), (D) and (E) are representative of 3 submaxillary tumors analysed. Magnification is indicated for each panel. A germinal center (GC) is also indicated in panel (B).
  • FIG. 16 depicts identification of a Mz-like B cell population infiltrating salivary glands of BAFF Tg mice. Submaxillary glands from 13-17 months old BAFF Tg mice and age-matched control littermates were digested with collagenase for purification of infiltrating lymphocytes as described in materials and methods. Using four-color flow cytometry analysis, cells were stained with anti-B220, anti-CD5, anti-IgM and anti-CD43. (A) shows the gating on B220hi and B220lo/int B cells on control (left) and BAFF Tg mice (right) as indicated. (B) shows four dot plots for expression of IgM and CD5 on B220hi cells (left) and B220lo/int (right) from BAFF Tg mice and control mice as indicated. The IgMhi B cell gate on B220hi cells (left) and the B-1a and B-1b gates on B220lo/int cells are shown. (C) shows histograms for the expression of CD43 on gated IgMhi/B220hi cells (left) and gated B-1a and B-1b cells (right) from BAFF Tg mice as indicated. (D) Lymphocytes prepared from submaxillary glands (left panel) and inguinal lymph nodes (right panel) of a BAFF Tg mouse were stained with anti-B220, anti-IgM and anti-L-selectin, using a four-color flow cytometry procedure. Cells from submaxillary glands were gated on B220hi/IgMhi (left panel) and on IgM+(right panel). A histogram of L-selectin expression is shown in each panel. (E) Lymphocytes from a BAFF Tg mice (right) and a control littermate (left) were prepared as in A and stained with anti-B220, anti-IgM and either anti-IgD, anti-CD23, anti-CD21, anti-CD1, anti-HSA, using a three color flow cytometry procedure. Cells were gated on B220hi and IgMhi cells as indicated on the top histograms and expression of IgD, CD23, CD1, CD21 and HSA on these gated cells is shown on histograms for control (left) and Tg mice (right) as indicated. In (C), (D) and (E), mean of fluorescence intensity (MFI) and a bar delineating the area in which the negative control histogram is located are indicated in each histogram. (F) Schematic representation of the common and distinct markers expressed on MZ (right circle), B-1 (left circle) and T1 B cells (lower circle). The phenotype of the B cells infiltrating submaxillary glands of BAFF Tg mice is indicated by an oval with a dotted line. These results are representative of at least 12 BAFF Tg and 7 control mice analysed.
  • FIG. 17 depicts decreased saliva flow in older BAFF Tg mice. 13 BAFF Tg mice (diamonds) and 14 control littermates (circles) were injected with pilocarpine prior to saliva collection as described in materials and methods. Mice 13-15.5 months old are shown in (A) and mice 8-10 months old are shown in (B). Means of saliva flow are shown with a bar and p values are indicated in each panel.
  • FIG. 18 depicts elevated levels of BAFF in sera and salivary gland tissues from patients suffering from primary SS but lack of correlation of these levels with levels of total IgG, RF and presence of anti-Ro/La autoantibodies. (A) individual BAFF levels in 39 healthy controls (square), 41 patients with primary SS (diamond), 53 patients with SLE (circle) and 53 patients with RA (triangle) were measured in sera by ELISA and BAFF levels plotted on a log scale. Controls were drawn from normal healthy donors. The horizontal black bars indicates the average for each group: normal 10.4±13 (ng/ml), SS patients 53±67 (ng/ml), SLE patients 12.7±24.4 (ng/ml) and RA patients 23±47 (ng/ml). Some individuals did not have detectable amount of BAFF in their serum and do not appear on the log scale, this includes 16 normals, 6 SS, 20 SLE and 10 RA patients. The dotted line delineates the range of normal BAFF levels. *p<0.04, the p value was determined by ANOVA t test. (B) and (C) show the correlation of BAFF levels in the sera of patients with SS with the corresponding levels of IgG (B) and RF (C) in each serum. r and p values calculated by ANOVA are shown. (D) shows the levels of BAFF in patients with detected anti-Ro and anti-La (left), anti-Ro only (middle) or no precipitin detected (right). (E) Paraffin sections of a human labial salivary gland biopsy from a patient with SS were stained with a rat anti-human BAFF antibody (right) or an isotype-matched control antibody (left). Staining of normal human labial salivary gland with rat anti-human BAFF antibody is also shown (bottom left, magnification 100×). The staining was revealed using biotin-labelled anti-rat Ig followed by Streptavidin-HRP. The staining appears brown on the sections. Magnification 200×. These pictures are representative of 4 patients with primary SS and 3 control tissues analysed.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Reference will now be made in detail to the present preferred embodiments of the invention. This invention relates to the use of BAFF and BAFF related molecules to effect the growth and maturation of B-cells and the secretion of immunoglobulin. The invention relates to the use of BAFF and BAFF related molecules to effect responses of the immune system, as necessitated by immune-related disorders. Additionally, this invention encompasses the treatment of cancer and immune disorders through the use of a BAFF, BAFF blocking agents, or BAFF related gene through gene therapy methods.
  • The BAFF ligand and homologs thereof produced by hosts transformed with the sequences of the invention, as well as native BAFF purified by the processes known in the art, or produced from known amino acid sequences, are useful in a variety of methods for anticancer, antitumor and immunoregulatory applications. They are also useful in therapy and methods directed to other diseases.
  • “BAFF blocking agents” refers to agents that can diminish BAFF ligand binding to BAFF receptors, or can diminish BAFF receptor signalling, or that can influence how the BAFF receptor signal is interpreted within the cell.
  • A BAFF blocking agent that acts by diminishing ligand-receptor binding can inhibit BAFF ligand binding by at least 20%. Examples of BAFF blocking agents include soluble BAFF receptor-Fc molecules, anti-BAFF ligand antibodies and anti-BAFF receptor antibodies.
  • “BAFF receptors” have been identified and characterized and include TACI (see, e.g., U.S. Pat. No. 5,969,102 and WO98/39361, incorporated herein by reference), BCMA (see, e.g., WO01/12812, incorporated herein by reference), and BAFFR (see, e.g., Thompson et al. (2001) Science 293:2108, incorporated herein by reference).
  • Another aspect of the invention relates to the use of the polypeptide encoded by the isolated nucleic acid encoding the BAFF-ligand in “antisense” therapy. As used herein, “antisense” therapy refers to administration or in situ generation of oligonucleotides or their derivatives which specifically hybridize under cellular conditions with the cellular mRNA and/or DNA encoding the ligand of interest, so as to inhibit expression of the encoded protein, i.e. by inhibiting transcription and/or translation. The binding may be by conventional base pair complementarity, or, for example, in the case of binding to DNA duplexes, through specific interactions in the major groove of the double helix. In general, “antisense” therapy refers to a range of techniques generally employed in the art, and includes any therapy which relies on specific binding to oligonucleotide sequences.
  • An antisense construct of the present invention can be delivered, for example, as an expression plasmid, which, when transcribed in the cell, produces RNA which is complementary to at least a portion of the cellular mRNA which encodes Kay-ligand. Alternatively, the antisense construct can be an oligonucleotide probe which is generated ex vivo. Such oligonucleotide probes are preferably modified oligonucleotides which are resistant to endogenous nucleases, and are therefor stable in vivo. Exemplary nucleic acids molecules for use as antisense oligonucleotides are phosphoramidates, phosphothioate andmethylphosphonate analogs of DNA (See, e.g., U.S. Pat. No. 5,176,996; U.S. Pat. No. 5,264,564; and U.S. Pat. No. 5,256,775). Additionally, general approaches to constructing oligomers useful in antisense therapy have been reviewed, for example, by Van Der Krol et al., (1988) Biotechniues 6:958-976; and Stein et al. (1988) Cancer Res 48: 2659-2668, specifically incorporated herein by reference.
  • A. BAFF-LIGAND
  • The BAFF-ligand of the invention, as discussed above, is a member of the TNF family and is described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herein. The protein, fragments or homologs thereof may have wide therapeutic and diagnostic applications.
  • The BAFF-ligand is present primarily in the spleen and in peripheral blood lymphocytes, strongly indicating a regulatory role in the immune system. Comparison of the claimed BAFF-ligand sequences with other members of the human TNF family reveals considerable structural similarity. All the proteins share several regions of sequence conservation in the extracellular domain.
  • Although the precise three-dimensional structure of the claimed ligand is not known, it is predicted that, as a member of the TNF family, it may share certain structural characteristics with other members of the family.
  • The novel polypeptides of the invention specifically interact with a receptor, which has not yet been identified. However, the peptides and methods disclosed herein enable the identification of receptors which specifically interact with the BAFF-ligand or fragments thereof.
  • The claimed invention in certain embodiments includes methods of using peptides derived from BAFF-ligand which have the ability to bind to their receptors. Fragments of the BAFF-ligands can be produced in several ways, e.g., recombinantly, by PCR, proteolytic digestion or by chemical synthesis. Internal or terminal fragments of a polypeptide can be generated by removing one or more nucleotides from one end or both ends of a nucleic acid which encodes the polypeptide. Expression of the mutagenized DNA produces polypeptide fragments.
  • Polypeptide fragments can also be chemically synthesized using techniques known in the art such as conventional Merrifield solid phase f-moc or t-boc chemistry. For example, peptides and DNA sequences of the present invention may be arbitrarily divided into fragments of desired length with no overlap of the fragment, or divided into overlapping fragments of a desired length. Methods such as these are described in more detail below.
  • B. Generation of Soluble Forms of BAFF-Ligand and BAFF-Receptors
  • Soluble forms of the BAFF-ligand can often signal effectively and hence can be administered as a drug which now mimics the natural membrane form. It is possible that the BAFF-ligand claimed herein are naturally secreted as soluble cytokines, however, if not, one can reengineer the gene to force secretion. To create a soluble secreted form of BAFF-ligand or BAFF receptor, one would remove at the DNA level the N-terminus transmembrane regions, and some portion of the stalk region, and replace them with a type I leader or alternatively a type II leader sequence that will allow efficient proteolytic cleavage in the chosen expression system. A skilled artisan could vary the amount of the stalk region retained in the secretion expression construct to optimize both receptor binding properties and secretion efficiency. For example, the constructs containing all possible stalk lengths, i.e. N-terminal truncations, could be prepared such that proteins starting at amino acids 81 to 139 would result. The optimal length stalk sequence would result from this type of analysis.
  • Soluble forms of BAFF receptors can be prepared using techniques well-known to those of ordinary skill in the art. Immunoglobulin Fc portions may be fused to the BAFF receptor protein to increase the half-life of the soluble BAFF receptor. Production of such soluble BAFF receptor proteins is described, for example, in WO 01/12812, WO 01/24811, and PCT/US01/40626, the entire disclosures of which are incorporated herein by reference.
  • C. Generation of Antibodies Reactive with the BAFF-Ligand and BAFF Receptors
  • The invention also includes antibodies specifically reactive with the claimed BAFF-ligand or its receptors. Anti-protein/anti-peptide antisera or monoclonal antibodies can be made by standard protocols (See, for example, Antibodies: A Laboratory Manual ed. by Harlow and Lane (Cold Spring Harbor Press: 1988)). A mammal such as a mouse, a hamster or rabbit can be immunized with an immunogenic form of the peptide. Techniques for conferring immunogenicity on a protein or peptide include conjugation to carriers, or other techniques, well known in the art.
  • An immunogenic portion of BAFF-ligand or its receptors can be administered in the presence of an adjuvant. The progress of immunization can be monitored by detection of antibody titers in plasma or serum. Standard ELISA or other immunoassays can be used with the immunogen as antigen to assess the levels of antibodies.
  • In a preferred embodiment, the subject antibodies are immunospecific for antigenic determinants of BAFF-ligand or its receptors, (e.g. antigenic determinants of a polypeptide of SEQ. ID. NO.: 2, said sequence as described in PCT application number PCTI/US98/19037 (WO99/12964) and is incorporated in its entirety herewith), or a closely related human or non-human mammalian homolog (e.g. 70, 80 or 90 percent homologous, more preferably at least 95 percent homologous). In yet a further preferred embodiment of the present invention, the anti-BAFF-ligand or anti-BAFF-ligand-receptor antibodies do not substantially cross react (i.e. react specifically) with a protein which is e.g., less than 80 percent homologous to SEQ. ID. NO.: 2 or 6 said sequence as described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herewith; preferably less than 90 percent homologous with SEQ. ID. NO.: 2 said sequence as described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herewith; and, most preferably less than 95 percent homologous with SEQ. ID. NO.: 2 said sequence as described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herewith. By “not substantially cross react”, it is meant that the antibody has a binding affinity for a non-homologous protein which is less than 10 percent, more preferably less than 5 percent, and even more preferably less than 1 percent, of the binding affinity for a protein of SEQ. ID. NO.: 2 said sequence as described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herewith.
  • Production of exemplary anti-BAFF receptor antibodies is described, for example, in WO 01/12812, WO 01/24811 and PCT/US01/40626, the entire disclosures of which are incorporated herein by reference.
  • The term antibody as used herein is intended to include fragments thereof which are also specifically reactive with BAFF-ligand, or its receptors. Antibodies can be fragmented using conventional techniques and the fragments screened for utility in the same manner as described above for whole antibodies. For example, F(ab′)2 fragments can be generated by treating antibody with pepsin. The resulting F(ab′)2 fragment can be treated to reduce disulfide bridges to produce Fab′ fragments. The antibodies of the present invention are further intended to include biospecific and chimeric molecules having anti-BAFF-ligand or anti-BAFF-ligand-receptor activity. Thus, both monoclonal and polyclonal antibodies (Ab) directed against BAFF-ligand, Tumor-ligand and their receptors, and antibody fragments such as Fab′ and F(ab′)2, can be used to block the action of the Ligand and their respective receptor.
  • Various forms of antibodies can also be made using standard recombinant DNA techniques. Winter and Milstein (1991) Nature 349: 293-299, specifically incorporated by reference herein. For example, chimeric antibodies can be constructed in which the antigen binding domain from an animal antibody is linked to a human constant domain (e.g. Cabilly et al., U.S. Pat. No. 4,816,567, incorporated herein by reference). Chimeric antibodies may reduce the observed immunogenic responses elicited by animal antibodies when used in human clinical treatments.
  • In addition, recombinant “humanized antibodies” which recognize BAFF-ligand or its receptors can be synthesized. Humanized antibodies are chimeras comprising mostly human IgG sequences into which the regions responsible for specific antigen-binding have been inserted. Animals are immunized with the desired antigen, the corresponding antibodies are isolated, and the portion of the variable region sequences responsible for specific antigen binding are removed. The animal-derived antigen binding regions are then cloned into the appropriate position of human antibody genes in which the antigen binding regions have been deleted. Humanized antibodies minimize the use of heterologous (i.e. inter species) sequences in human antibodies, and thus are less likely to elicit immune responses in the treated subject.
  • Construction of different classes of recombinant antibodies can also be accomplished by making chimeric or humanized antibodies comprising variable domains and human constant domains (CH1, CH2, CH3) isolated from different classes of immunoglobulins. For example, antibodies with increased antigen binding site valencies can be recombinantly produced by cloning the antigen binding site into vectors carrying the human: chain constant regions. Arulanandam et al. (1993) J. Exp. Med., 177: 1439-1450, incorporated herein by reference.
  • In addition, standard recombinant DNA techniques can be used to alter the binding affinities of recombinant antibodies with their antigens by altering amino acid residues in the vicinity of the antigen binding sites. The antigen binding affinity of a humanized antibody can be increased by mutagenesis based on molecular modeling. Queen et al., (1989) Proc. Natl. Acad. Sci. 86: 10029-33 incorporated herein by reference.
  • D. Generation of Analogs: Production of Altered DNA and Peptide Sequences
  • Analogs of the BAFF-ligand can differ from the naturally occurring BAFF-ligand in amino acid sequence, or in ways that do not involve sequence, or both. Non-sequence modifications include in vivo or in vitro chemical derivatization of the BAFF-ligand. Non-sequence modifications include, but are not limited to, changes in acetylation, methylation, phosphorylation, carboxylation or glycosylation.
  • Preferred analogs include BAFF-ligand biologically active fragments thereof, whose sequences differ from the sequence given in SEQ. ID NO. 2 said sequence as described in PCT application number PCT/US98/19037 (WO99/12964) and is incorporated in its entirety herewith, by one or more conservative amino acid substitutions, or by one or more non-conservative amino acid substitutions, deletions or insertions which do not abolish the activity of BAFF-ligand. Conservative substitutions typically include the substitution of one amino acid for another with similar characteristics, e.g. substitutions within the following groups: valine, glycine; glycine, alanine; valine, isoleucine, leucine; aspartic acid, glutamic acid; asparagine, glutamine; serine, threonine; lysine, arginine; and, phenylalanine, tyrosine.
  • E. Materials and Methods of the Invention
  • The anti-Flag M2 monoclonal antibody, biotinylated anti-Flag M2 antibody and the anti-Flag M2 antibody coupled to agarose were purchased from Sigma. Cell culture reagents were obtained from Life Sciences (Basel, Switzerland) and Biowhittaker (Walkersville, Md.). Flag-tagged soluble human APRIL (residues K110-L250) was produced in 293 cells as described (10, 11). FITC-labeled anti-CD4, anti-CD8 and anti-CD19 antibodies were purchased from Pharmingen (San Diego, Calif.). Goat F(ab)2 specific for the Fc5, fragment of human IgM were purchased from Jackson ImmunoResearch (West Grove, Pa.). Secondary antibodies were obtained from either Pharmingen or from Jackson ImmunoResearch and used at the recommended dilutions.
  • Human embryonic kidney 293 T (12) cells and fibroblast cell lines (Table 1) were maintained in DMEM containing 10% heat-inactivated fetal calf serum (FCS). Human embryonic kidney 293 cells were maintained in DMEM-nutrient mix F12 (1:1) supplemented with 2% FCS. T cell lines, B cell lines, and macrophage cell lines (Table 1) were grown in RPMI supplemented with 10% FCS. Molt-4 cells were cultiv-atedin Iscove's medium-supplemented with 10% FCS. Epithelial cell lines were grown in MEM-alpha medium containing 10% FCS, 0.5 mM non-essential amino acids, 10 mM Na-Hepes and 1 mM Na pyruvate. HUVECs were maintained in M199 medium supplemented with 20% FCS, 100 μg/ml of epithelial cell growth factor (Collaborative Research, Inotech, Dottikon, Switzerland) and 100 μg/ml of heparin sodium salt (Sigma). All media contained penicillin and streptomycin antibiotics. Peripheral blood leukocytes were isolated from heparinized blood of healthy adult volunteers by Ficoll-Paque (Pharmacia, Uppsala, Sweden) gradient centrifugation and cultured in RPMI, 10% FCS.
  • T cells were obtained from non-adherents PBLs by rosetting with neuraminidase-treated sheep red blood cells and separated from non-rosetting cells (mostly B cells and monocytes) by Ficoll-Paque gradient centrifugation. Purified T cells were activated for 24 h with phytohemagglutinin (Sigma) (1 μg/ml), washed and cultured in RPMI, 10% FCS, 20 U/ml of IL 2. CD14+ monocytes were purified by magnetic cell sorting using anti-CD 14 antibodies, goat anti-mouse-coated microbeads and a Minimacs™ device (Miltenyi Biotech), and cultivated in the presence of GM-CSF (800 U/ml, Leucomax®, Essex Chemie, Luzern, Switzerland) and IL-4 (20 ng/ml, Lucerna Chem, Luzern, Switzerland) for 5 d, then with GM-CSF, IL4 and TNFα(200 U/ml, Bender, Vienna, Austria) for an additional 3 d to obtain a CD83+, dentritic cell-like population. Human B cells of >97% purity were isolated from peripheral blood or umbilical cord blood using anti-CD19 magnetic beads (M450, Dynal, Oslo, Norway) as described (13).
  • Northern Blot Analysis
  • Northern blot analysis was carried out using Human Multiple Tissue Northern Blots I and II (Clontech #7760-1 and #7759-1). The membranes were incubated in hybridization solution (50% formamide, 2.5× Denhardt's, 0.2% SDS, 10 mM EDTA, 2×SSC, 50 mM NaH2PO4, pH 6.5, 200 μg/ml sonicated salmon sperm DNA) for 2 h at 60° C. Antisense RNA probe containing the nucleotides corresponding to amino acids 136-285 of hBAFF was heat-denatured and added at 2×106 cpm/ml in fresh hybridization solution. The membrane was hybridized 16 h at 62° C., washed once in 2×SSC, 0.05% SDS (30 min at 25° C.), once in 0.1×SSC, 0.1% SDS (20 min at 65° C.) and exposed at −70° C. to X-ray films.
  • Characterization of BAFF cDNA.
  • A partial sequence of human BAFF cDNA was contained in several EST clones (e.g. GenBank Accession numbers T87299 and AA166695) derived from fetal liver and spleen and ovarian cancer libraries. The 5′ portion of the cDNA was obtained by 5′-RACE-PCR (Marathon-Ready cDNA, Clonetech, Palo Alto, Calif.) amplification with oligonucleotides AP1 and JT1013 (5′-ACTGTTTCTTCTGGACCCTGAACGGC-3′) [SEQ ID. NO.: 9] using the provided cDNA library from a pool of human leukocytes as template, as recommended by the manufacturer. The resulting PCR product was cloned into PCR-0 blunt (Invitrogen, Nev. Leek, The Netherlands) and subcloned as EcoRI/PstI fragment into pT7T3 Pac vector (Pharmacia) containing EST clone T87299. Full-length hBAFF cDNA was therefore obtained by combining 5′ and 3′ fragments using the internal PstI site of BAFF. Sequence has been assigned GenBank accession number AF116456.
  • A partial 617 bp sequence of murine BAFF was contained in two overlapping EST clones (AA422749 and AA254047). A PCR fragment spanning nucleotides 158 to 391 of this sequence was used as a probe to screen a mouse spleen cDNA library (Stratagene, La Jolla, Calif.).
  • Expression of Recombinant BAFF
  • Full length hBAFF was amplified using oligos JT1069 (5′-GACAAGCTTGCCACCATGGATGACTCCACA-3′) [SEQ. ID. NO.: 10] and JT637 (5′-ACTAGTCACAGCAGTITCAATGC-3) [SEQ. ID. NO.: 11]. The PCR product was cloned into PCR-0 blunt and re-subcloned as HindIII/EcoRI fragment into PCR-3 mammalian expression vector. A short version of soluble BAFF (amino acids Q136-L285) was amplified using oligos JT636 (5′-CTGCAGGGTCCAGAAGAAACAG-3) [SEQ. ID. NO.: 12] and JT637. A long version of soluble BAFF (aa L83-L285) was obtained from full length BAFF using internal PstI site. Soluble BAFFs were resubcloned as PstI/EcoRI fragments behind the haemaglutinin signal peptide and Flag sequence of a modified PCR-3 vector, and as PstI/SpeI fragments into a modified pQE16 bacterial expression vector in frame with a N-terminal Flag sequence (14). Constructs were sequenced on both strands. The establishment of stable 293 cell lines expressing the short soluble form or full length BAFF, and the expression and purification of recombinant soluble BAFF from bacteria and mammalian 293 cells was performed as described (14, 15).
  • Reverse Transcriptase PCR
  • Total RNA extracted from T cells, B cells, in vitro derived immature dendritic cells, 293 wt and 293-BAFF (full length) cells was reverse transcribed using the Ready to Go system (Pharmacia) according to the manufacturer's instructions. BAFF and β-actin cDNAs were detected by PCR amplification with Taq DNA polymerase (steps of 1 min each at 94° C., 55° C. and 72° C. for 30 cycles) using specific oligonucleotides: for BAFF, JT1322 5′-GGAGAAGGCAACTCCAGTCAGAAC-3′ [SEQ. ID. NO.: 13] and JT1323 5′-CAATTCATCCCCAAAGACATGGAC-3′ [SEQ. ID. NO.: 14]; for IL-2 receptor alpha chain, JT1368 5′-TCGGAACACAACGAAACAAGTC-3′ [SEQ. ID. NO.: 15] and JT1369 5′-CTTCTCCTTCACCTGGAAACTGACTG-3′ [SEQ. ID NO.: 16]; for β-actin, 5′-GGCATCGTGATGGACTCCG-3′ [SEQ. ID. NO.: 17] and 5′-GCTGGAAGGTGGACAGCGA-3′ [SEQ. ID. NO.: 18].
  • Gel Permeation Chromatography
  • 293T cells were transiently transfected with the short form of soluble BAFF and grown in serum-free Optimem medium for 7 d. Conditionned supernatants were concentrated 20×, mixed with internal standards catalase and ovalbumin, and loaded onto a Superdex-200 HR10/30 column. Proteins were eluted in PBS at 0.5 ml/min and fractions (0.25 ml) were precipitated with trichloroacetic acid and analyzed by Western blotting using anti-Flag M2 antibody. The column was calibrated with standard proteins: ferritin (440 kDa), catalase (232 kDa), aldolase (158 kDa), bovine serum albumine (67 kDa), ovalbumine (43 kDa), chymotrypsinogen A (25 kDa) and ribonuclease A (13.7 kDa).
  • PNGase F Treatment
  • Samples were heated in 20 μl of 0.5% SDS, 1% 2-mercaptoethanol for 3 min at 95° C., then cooled and supplemented with 10% Nonidet P40 (2 μl), 0.5 M sodium phosphate, pH 7.5 (2 μl) and Peptide N-glycanase F (125 units/μl, 1 μl, or no enzyme in controls). Samples were incubated for 3 h at 37° C. prior to analysis by Western blotting.
  • EDMAN Sequencing
  • 293 T cells were transiently transfected with the long form of soluble BAFF and grown in serum-free Optimem medium for 7 d. Conditioned supernatants were concentrated 20×, fractionated by SDS-PAGE and blotted onto polyvinylidene difluoride membrane (BioRad Labs, Hercules, Calif.) as previously described (16), and then sequenced using a gas phase sequencer (ABI 120A, Perkin Elmer, Foster City, Calif.) coupled to an analyzer (ABI 120A, Perkin Elmer) equipped with a phenylthiohydantoin C18 2.1×250 mm column. Data was analyzed using software ABI 610 (Perkin. Elmer).
  • Antibodies
  • Polyclonal antibodies were generated by immunizing rabbits (Eurogentec, Seraing, Belgium) with recombinant soluble BAFF. Spleen of rats immunized with the same antigen were fused to x63Ag8.653 mouse myeloma cells, and hybridoma were screened for BAFF-specific IgGs. One of these monoclonal antibodies, 43.9, is an IgG2a that specifically recognizes hBAFF.
  • Cells were stained in 50 μl of FACS buffer (PBS, 10% FCS, 0.02% NaN3) with 50 ng (or the indicated amount) of Flag tagged short soluble HBAFF for 20 min at 4° C., followed by anti-Flag M2 (1 μg) and secondary antibody. Anti-BAFF mAb 43.9 was used at 40 μg/ml. For two color FACS analysis, peripheral blood lymphocytes were stained with Flag tagged soluble BAFF/long (2 μg/ml), followed by biotinylated anti-Flag M2 (1/400) and PE-labeled streptavidin (1/100), followed by either FITC-labeled anti-CD4, anti-CD8 or anti-CD19.
  • PBL Proliferation Assay
  • Peripheral blood leukocytes were incubated in 96-well plates (105 cells/well in 100 μl RPMI supplemented with 10% FCS) for 72 h in the presence or absence of 2 μg/ml of goat anti-human p chain antibody (Sigma) or control F(ab′)2 and with the indicated concentration of native or boiled soluble BAFF/long. Cells were pulsed for an additional 6 h with [3H]thymidine (1 μCi/well) and harvested. [3H]thymidine incorporation was monitored by liquid scintillation counting. In some experiments, recombinant soluble BAFF was replaced by 293 cells stably transfected with full length BAFF (or 293 wt as control) that had been fixed for 5 min at 25° C. in 1% paraformaldeyde. Assay was performed as described (17). In further experiments, CD19+ cells were isolated form PBL with magnetic beads and the remaining CD19 cells were irradiated (3000 rads) prior to renconstitution with CD19+cells. Proliferation assay with sBAFF was then performed as described above.
  • B cell Activation Assay
  • Purified B cells were activated in the EL4 culture system as described (13). Briefly, 104 B cells mixed with 5×104 irradiated murine ELA thymoma cells (clone B5) were cultured for 5-6 d in 200 μl medium containing 5% v/v of culture supernatants from human T cells (106/ml) which had been activated for 48 h with PHA (1 μg/ml) and PMA (1 ng/ml). B cells were then reisolated with anti-CD19 beads and cultured for another 7 d (5×104 cells in 200 μl, duplicate or triplicate culture in flat bottomed 96 well plates) in medium alone or in medium supplemented with 5% T cell supernatants, or with 50 ng/ml IL-2 (a kind gift from the former Glaxo Institute for Molecular Biology, Geneva) and 10 ng/ml each IL4 and IL-10 (Peprotech, London, UK), in the presence or absence of sBAFF. The anti-Flag M2 antibody was added at a concentration of 2 μg/ml and had no effect by itself. IgM, IgG and IgA in culture supernatants were quantitated by ELISA assays as described (13).
  • Human BAFF was identified by sequence homology as a possible novel member of the TNF ligand family while we screened public databases using an improved profile search (18). A cDNA encoding the complete protein of 285 amino acids (aa) was obtained by combining EST-clones (covering the 3′ region) with a fragment (5′ region) amplified by PCR. The absence of a signal peptide suggested that BAFF was a type II membrane protein that is typical of the members of the TNF-ligand family. The protein has a predicted cytoplasmic domain of 46 aa, a hydrophobic transmembrane region, and an extracellular domain of 218 aa containing two potential N-glycosylation sites (FIG. 1A). The sequence of the extracellular domain of BAFF shows highest homology with APRIL (33% amino acid identities, 48% homology), whereas the identity with other members of the family such as TNF, FasL, LTa, TRAIL or RANKL is below 20% (FIG. 1B, C). The mouse BAFF cDNA clone isolated from a spleen library encoded a slightly longer protein (309 aa) due to an insertion between the transmembrane region and the first of several β-strands which constitute the receptor binding domain in all TNF ligand members (19). This β-strand rich ectodomain is almost identical in mouse and human BAFF (86% identity, 93% homology) suggesting that the BAFF gene has been highly conserved during evolution (FIG. 1A).
  • Although TNF family members are synthesized as membrane-inserted ligands, cleavage in the stalk region between transmembrane and receptor binding domain is frequently observed. For example, TNF or FasL are readily cleaved from the cell surface by metalloproteinases (20, 21). While producing several forms of recombinant BAFF in 293T cells, we noticed that a recombinant soluble 32 kDa form of BAFF (aa 83-285, sBAFF/long), containing the complete stalk region and a N-terminal Flag-tag in addition to the receptor binding domain, was extensively processed to a smaller 18 kDa fragment (FIG. 2A, B). Cleavage occurred in the stalk region since the fragment was detectable only with antibodies raised against the complete receptor interaction domain of BAFF but not with anti-Flag antibodies (data not shown). Also revealed was that only N124 (located in the stalk) but not N242 (located at the entry of the F-□ sheet) was glycosylated, since the molecular mass of the non-processed sBAFF/long was reduced from 32 kDa to 30 kDa upon removal of the N-linked carbohydrates with PNGase F whereas the 18 kDa cleaved form was insensitive to this treatment. Peptide sequence analysis of the 18 kDa fragment indeed showed that cleavage occurred between R133 and A134 (FIG. 1A). R133 lies at the end of a polybasic region which is conserved between human (R-N-K-R) and mouse (R-N-R-R). To test whether cleavage was not merely an artifact of expressing soluble, non-natural forms of BAFF, membrane-bound full length BAFF was expressed in 293T cells (FIG. 2C). The 32 kDa complete BAFF and some higher molecular mass species (probably corresponding to non-dissociated dimers and trimers) were readily detectable in cellular extracts, but more than 95% of BAFF recovered from the supernatant corresponded to the processed 18 kDa form, indicating that BAFF was also processed when synthesized as a membrane-bound ligand.
  • A soluble BAFF was engineered (Q136-L285, sBAFF/short) whose sequence started 2 aa downstream of the processing site (FIG. 1 B). As predicted, the Flag-tag attached to the N-terminus of this recombinant molecule was not removed (data not shown) which allowed its purification by an anti-Flag affinity column. To test its correct folding, the purified sBAFF/short was analyzed by gel filtration where the protein eluted at an apparent molecular mass of 55 kDa (FIG. 2D). The sBAFF/short correctly assembles into a homotrimer (3×20 kDa) in agreement with the quaternary structure of other TNF family members (19). Finally, unprocessed sBAFF/long was readily expressed in bacteria, indicating that the cleavage event was specific to eukaryotic cells.
  • Northern blot analysis of BAFF revealed that the 2.5 kb BAFF mRNA was abundant in the spleen and PBLs (FIG. 3A). Thymus, heart, placenta, small intestine and lung showed weak expression. This restricted distribution suggested that cells present in lymphoid tissues were the main source of BAFF. Through PCR analysis, we found that BAFF mRNA was present in T cells and peripheral blood monocyte-derived dendritic cells but not in B cells (FIG. 3B). Even naive, non-stimulated T cells appeared to express some BAFF mRNA.
  • A sequence tagged site (STS, SHGC-36171) was found in the database which included the human BAFF sequence. This site maps to human chromosome 13, in a 9 cM interval between the markers D13S286 und D13S1315. On the cytogenetic map, this interval corresponds to 13q32-34. Of the known TNF ligand family members, only RANKL (Trance) has been localized to this chromosome (22) though quite distant to BAFF (13q14).
  • In order for the ligand to exert maximal biological effects, it was likely that the BAFF receptor (BAFF-R) would be expressed either on the same cells or on neighboring cells present in lymphoid tissues. Using the recombinant sBAFF as a tool to specifically determine BAFF-R expression by FACS, we indeed found high levels of receptor expression in various B cell lines such as the Burkitt lymphomas Raji and BJAB (FIG. 4A, Table 1). In contrast, cell lines of T cell, fibroblastic, epithelial and endothelial origin were all negative. Very weak staining was observed with the monocyte line THP-1 which, however, could be due to Fc receptor binding. Thus, BAFF-R expression appears to be restricted to B cell lines. The two mouse B cell lines tested were negative using the human BAFF as a probe, although weak binding was observed on mouse splenocytes (data not shown). The presence of BAFF-R on B cells was corroborated by analysis of umbilical cord and peripheral blood lymphocytes. While CD8+ and CD4+ T cells lacked BAFF-R (FIG. 4B and data not shown), abundant staining was observed on CD19+ B cells (FIGS. 4A and 4B), indicating that BAFF-R is expressed on all blood B cells, including naive and memory ones.
  • Since BAFF bound to blood-derived B cells, experiments were performed to determine whether the ligand could deliver growth-stimulatory or -inhibitory signals. Peripheral blood lymphocytes (PBL) were stimulated with anti-IgM (μ) antibodies together with fixed 293 cells stably expressing surface BAFF (FIG. 5A). The levels of [3H]thymidine incorporation induced by anti-μ alone was not altered by the presence of control cells but was increased two-fold in the presence of BAFF-transfected cells (FIG. 5B). A dose-dependent proliferation of PBL was also obtained when BAFF-transfected cells were replaced by purified sBAFF (FIG. 5C), indicating that BAFF does not require membrane attachment to exert its activity. In this experimental setup, proliferation induced by sCD40L required concentrations exceeding 1 μg/ml but was less dependent on the presence of anti-μ than that mediated by BAFF (FIG. 5D). When purified CD19+ B cells were co-cultured with irradiated autologous CD19 PBL, costimulation of proliferation by BAFF was unaffected, demonstrating that [3H]thymidine uptake was mainly due to B cell proliferation and not to an indirect stimulation of another cell type (data not shown). The observed B cell proliferation in response to BAFF was entirely dependent on the presence of anti-μ antibodies, indicating that BAFF functioned as costimulator of B cell proliferation.
  • To investigate a possible effect of BAFF on immunoglobulin secretion, purified peripheral or cord blood B cells were preactivated by coculture with EL-4 T cells in the presence of a cytokine mixture from supernatants of PHA/PMA stimulated T cells (23). These B cells were reisolated to 98% purity and yielded a two-fold increase in Ig secretion during a secondary culture in the presence of BAFF and activated T cell cytokines as compared to cytokines alone. A very modest effect occurred in the absence of exogenous cytokines, and an intermediate (1.5-fold) effect was observed in the presence of the recombinant cytokines IL-2, IL4 and IL-10 (FIG. 5E, F).
  • The biochemical analysis of BAFF is also consistent with the typical homotrimeric structure of TNF family members. Among this family of ligands, BAFF exhibits the highest level of sequence similarity with APRIL which we have recently characterized as a ligand stimulating growth of various tumor cells (11). Unlike TNF and LTO which are two family members with equally high homology (33% identity) and whose genes are linked on chromosome 6, APRIL and BAFF are not clustered on the same chromosome. APRIL is located on chromosome 17 (J. L. B., unpublished data) whereas BAFF maps to the distal arm of human chromosome 13 (13q34). Abnormalities in this locus were characterized in Burkitt lymphomas as the second most frequent defect (24) besides the translocation involving the myc gene into the Ig locus (25). Considering the high expression levels of BAFF-R on all Burkitt lymphoma cell lines analyzed (see Table 1), this raises the intriguing possibility that some Burkitt lymphomas may have deregulated BAFF expression, thus stimulating growth in an autocrine manner.
  • The role of antigen-specific B lymphocytes during the different stages of the immune response is highly dependent on signals and contacts from helper T cells and antigen-presenting cells such as dendritic cells (20). B lymphocytes first receive these signals early on during the immune response when they interact with T cells at the edge of the B cell follicles in lymphoid tissues, leading to their proliferation and differentiation into low affinity antibody forming cells (18). At the same time some antigen-specific B cells also migrate to the B cell follicle and contribute to the formation of germinal centers, another site of B cell proliferation but also affinity maturation and generation of memory B cells and high affinity plasma cells (19).
  • Signals triggered by another member of the TNF super family CD40L have been shown to be critical for the function of B lymphocytes at multiple steps of the T cell-dependent immune response. However, several studies clearly showed that CD40UCD40 interaction does not account for all contact-dependent T-cell help for B cells. Indeed, CD40L-deficient T cells isolated from either knock-out mice or patients with X-linked hyper IgM syndrome have been shown to sucessfully induce proliferation of B cells and their differentiation into plasma cells. Studies using blocking antibodies against CD40L showed that a subset of surface IgD positive B cells isolated from human tonsils proliferate and differentiate in response to activated T cells in a CD40-independent manner. Other members of the TNF family such as membrane-bound TNF and CD30L have also been shown to be involved in a CD40- and surface Ig-independent stimulation of B cells. Similar to our results with BAFF, it has been shown that CD40-deficient B cells can be stimulated to proliferate and differentiate into plasma cells by helper T cells as long as the surface Ig receptors are triggered at the same time. BAFF as well as CD30L and CD40L is expressed by T cells but its originality resides in its expression by dendritic cells as well as the highly specific location of its receptor on B cells which is in contrast to CD40, CD30 and the TNF receptor which expression has been descrided on many different cell. This observation suggests independent and specific BAFF-induced functions on B cells.
  • In support of a role for BAFF in T cell- and dendritic cell-induced B cell growth and potential maturation, we found that BAFF costimulates proliferation of blood-derived B cells concomitantly with cross-linking of the B cell receptors, and, thus, independently of CD40 signalling. Moreover, using CD19 positive B cells differentiated in vitro into a pre-plasma cell/GC-like B cell (14), we observed a costimulatory effect of BAFF on Ig secretion by these B cells in the presence of supernatant from activated T cells or a blend of IL-2, IL-4 and IL-10. Interestingly, the costimulatory effect was stronger in presence of the activated T cell supernatant when compared to the cytokine blend, suggesting additional soluble factors secreted by activated T cells involved in antibody production which can synergize with BAFF or additional BAFF itself. It is, therefore, possible that BAFF actively contributes to the differentiation of these GC-like B cells into plasma.
  • It is clear that BAFF can signal in both naive B cells as well as GC-conunited B cells in vitro. Whether this observation will translate or not during a normal immune response will have to be addressed by proper in vivo experiments.
  • The biological responses induced in B cells by BAFF are distinct from that of CD40L, since proliferation triggered by CD40L was less dependent on an anti-μ costimulus (17) (and FIG. 5D). Morever, CD40L can counteract apoptotic signals in B cells following engagement of the B cell receptor (29), whereas BAFF was not able to rescue the B cell line Ramos from anti-μ-mediated apoptosis, despite the fact that Ramos cells do express BAFF-R (Table 1; F. M. and J. L. B., unpublished observations). It is therefore likely that CD40L and BAFF fulfill distinct functions. In this respect, it is noteworthy that BAFF did not interact with any of 16 recombinant receptors of the TNF family tested, including CD40 (P.S and J.T, unpublished observations).
  • B cell growth was efficiently costimulated with recombinant soluble BAFF lacking the transmembrane domain. This activity is in contrast to several TNF family members which are active only as membrane-bound ligand such as TRAIL, FasL and CD40L. Soluble forms of these ligands have poor biological activity which can be enhanced by their cross-linking, thereby mimicking the membrane-bound ligand (15). In contrast, cross-linking Flag-tagged sBAFF with anti-Flag antibodies or the use of membrane-bound BAFF expressed on the surface of epithelial cells did not further enhance the mitogenic activity of BAFF, suggesting that it can act systemically as a secreted cytokine, like TNF does. This is in agreement with the observation that a polybasic sequence present in the stalk of BAFF acted as a substrate for a protease. Similar polybasic sequences are also present at corresponding locations in both APRIL and TWEAK and for both of them there is evidence of proteolytic processing (30) (N.H. and J.T, unpublished observation). Although the protease responsible for the cleavage remains to be determined, it is unlikely to be the metalloproteinase responsible for the release of membrane-bound TNF as their sequence preferences differ completely (21). The multibasic motifs in BAFF (R-N-K-R), APRIL (R-K-R-R) and Tweak (R-P-R-R) are reminiscent of the minimal cleavage signal for furin (R-X-K/R-R), the prototype of a proprotein convertase family (31).
  • Practice of the present invention will employ, unless indicated otherwise, conventional techniques of cell biology, cell culture, molecular biology, microbiology, recombinant DNA, protein chemistry, and immunology, which are within the skill of the art. Such techniques are described in the literature. See, for example, Molecular Cloning: A Laboratory Manual, 2nd edition. (Sambrook, Fritsch and Maniatis, eds.), Cold Spring Harbor Laboratory Press, 1989; DNA Cloning, Volumes I and II (D. N. Glover, ed), 1985; Oligonucleotide Synthesis, (M. J. Gait, ed.), 1984; U.S. Pat. No. 4,683,195 (Mullis et al.,); Nucleic Acid Hybridization (B. D. Hames and S. J. Higgins, eds.), 1984; Transcription and Translation (B. D. Hames and S. J. Higgins, eds.), 1984; Culture of Animal Cells (R. I. Freshney, ed). Alan R. Liss, Inc., 1987; Immobilized Cells and Enzymes, IRL Press, 1986; A Practical Guide to Molecular Cloning (B. Perbal), 1984; Methods in Enzymology, Volumes 154 and 155 (Wu et al., eds), Academic Press, New York; Gene Transfer Vectors for Mammalian Cells (J. H. Miller and M. P. Calos, eds.), 1987, Cold Spring Harbor Laboratory; Immunochemical Methods in Cell and Molecular Biology (Mayer and Walker, eds.), Academic Press, London, 1987; Handbook of Experiment Immunology, Volumes I-IV (D. M. Weir and C. C. Blackwell, eds.), 1986; Manipulating the Mouse Embryo, Cold Spring Harbor Laboratory Press, 1986.
  • The following Examples are provided to illustrate the present invention, and should not be construed as limiting thereof.
  • EXAMPLES
  • The following experimental procedures were utilized in Examples 1-6.
  • DNA Construct for the Generation of Murine BAFF Tg Mice
  • Both human and murine cDNA sequences have been described previously (Schneider et al., 1999). A PCR fragment encoding full-length murine BAFF was generated by RT-PCR. First strand cDNA was synthesized from mouse lung polyA+ (Clontech, Palo Alto, Calif.) using oligo dT according to the manufacturer's protocol (GibcoBRL, (Grand Island, N.Y.). The PCR reaction contained 1×Pfu buffer (Stratagene, La Jola, Calif.), 0.2 mM dNTPs, 10% DMSO, 12.5 pM primers, 5 units Pfu enzyme (Stratagene) and the following primers with Not1 restriction sites 5′-TAAGAATGCGGCCGCGGAATGGATGAGTCTGCAAA-3′ [SEQ. ID. NO.: 19] and 5′-TAAGAATGCGGCCGCGGGATCACGCACTCCAGCAA-3′ [SEQ. ID. NO.: 20]. The template was amplified for 30 cycles at 94° C. for 1 min, 54° C. for 2 min and 72° C. for 3 min followed by a 10 min extension at 72° C. This sequence corresponds to nucleotides 214 to 1171 of the GenBank file AF119383. The PCR fragment was digested with Not1 and then cloned into a modified pCEP4 vector (Invitrogen, Carlsbad, Calif.). The fragment containing murine BAFF was removed with Xba1 in order to include the SV40 polyA addition site sequence. This fragment was cloned into a pUC based vector where the promoter sequence was added. The promoter, a 1 Kb blunt Bgl2-Not1 fragment containing the human ApoE enhancer and AAT (alpha anti-trypsin) promoter was purified from the plasmid clone 540B (a kind gift from Dr. Katherine Parker Ponder, Washington University, St. Louis, Mo.). An EcoRV/Bgl2 fragment was purified from the final vector and used for the generation of transgenic mice. The injected offspring of C57BL/6J female×DBA/2J male F1 (BDF1) mice were backcrossed onto C57BL/6 mice. Techniques of microinjection and generation of transgenic mice have been previously described (Mcknights et al., 1983).
  • Analytical Methods:
  • Serum samples were subject to reduced SDS-PAGE analysis using a linear 12.5% gel. Total RNA from mouse liver was prepared and processed for Northern Blot analysis using an isolation kit from Promega (Madison, Wis.) according to the manufacturer's guidelines. BAFF transgene-specific mRNA was detected using a probe spanning the SV40 poly A tail of the transgene construct and obtained by digestion of the modified pCEP4 vector with Xba1 and BamH1. The probe recognizes a 1.8-2 Kd band corresponding to mRNA from the BAFF transgene. PCR analysis of tail DNA from BAFF Tg mice was carried using 12.5 pM of the following primers 5′-GCAGTTTCACAGCGATGTCCT-3′ [SEQ. ID. NO.: 21] and 5′-GTCTCCGTTGCGTGAAATCTG-3′ [SEQ. ID. NO.: 22] in a reaction containing 1× Taq polymerase buffer (Strata gene), 0.2 nM dNTPs, 10% DMSO and 5 units of Taq polymerase (Stratagene). A 719 bp of the transgene was amplified for 35 cycles at 94° C. for 30 sec., 54° C. for 1 min. and 72° C. for 1.5 min. followed by a 10 min. extension at 72° C.
  • The presence of proteins in mouse urine was measured using Multistix 10 SG reagent strips for urinalysis (Bayer Corporation, Diagnostics Division, Elkhart, Ind.).
  • Cell-dyn and Cytofluorimetric Analysis (FACS).
  • Differential WBC counts of fresh EDTA anticoagulated whole blood were performed with an Abbott Cell Dyne 3500 apparatus (Chicago, Ill.). For FACS analysis, Fluorescein (FITC)-, Cy-chrome- and Phycoerythrin-(PE)-labeled rat anti-mouse antibodies: anti-B220, anti-CD4, anti-CD8, anti-CD43, anti-IgM, anti-CD5, anti-CD25, anti-CD24, anti-CD38, anti-CD21, anti-CD44, anti-L-selectin and hamster anti-Bcl-2/control hamster Ig kit were purchased from Pharmingen (San Diego, Calif.). Production of recombinant E. coli as well as mammalian cell-derived human and mouse Flag-tagged BAFF were previously described (Schneider et al., 1999). All antibodies were used according to the manufacturer's specifications. PBL were purified from mouse blood as follows: mouse blood was collected in microtubes containing EDTA and was diluted ½ with PBS. Five hundred μl of diluted blood was applied on top of 1 ml of ficoll (Celardane, Hornby, Ontario, Canada) in a 4 ml glass tube, the gradient was performed at 2000 rpm for 30 min at room temperature and the interface containing the lymphocytes was collected and washed twice in PBS prior to FACS staining. Spleen, bone marrow and mesenteric lymph nodes were ground into a single cell suspension in RPMI medium (Life Technologies, Inc., Grand Island, N.Y.) and washed in FACS buffer (PBS supplemented with 2% fetal calf serum (JRH Biosciences, Lenexa, Kans.). Cells were first suspended in FACS buffer supplemented with the following blocking reagents: 10 μg/ml human Ig (Sandoz, Basel, Switzerland) and 10 μg/ml anti-mouse Fc blocking antibody (Pharmingen) and incubated 30 min on ice prior to staining with fluorochrome-labeled antibodies. All antibodies were diluted in FACS buffer with the blocking reagent mentioned above. Samples were analyzed using a FACScan cytofluorometer (Becton Dickinson).
  • Detection of Total Mouse Ig and Rheumatoid Factors in Mouse Sera by ELISA Assays.
  • ELISA plates (Corning glass works, Corning, N.Y.) were coated overnight at 4° C. with a solution of 10 μg/ml of goat anti-total mouse Ig (Southern Biotechnology Associates, Inc. Birmingham, Ala.) in 50 mM sodium bicarbonate buffer pH 9.6. Plates were washed 3 times with PBS/0.1% Tween and blocked overnight with 1% gelatin in PBS. One hundred μl/well of serum serial dilutions or standard dilutions was added to the plates for 30 min at 37° C. Mouse Ig were detected using 100 μl/well of a 1 μg/ml solution of an Alkaline Phosphatase (AP)-labeled goat anti-total mouse Ig (Southern Biotechnology Associates) for 30 min at 37° C. After a last wash, 3 times with PBS/0.1% Tween, the enzymatic reaction was developed using a solution of 10 μg/ml of p-nitrophenyl phosphate (Boehringer Mannheim, Indianapolis, Ind.) in 10% diethanolamine. The reaction was stopped by adding 100 μl of 3N NaOH/well. The optical density (O.D.) was measured at 405 nm using a spectrophotometer from Molecular Devices (Sunnyvale, Calif.). Standard curves were obtained using purified mouse Ig purchased from Southern Biotechnology Associates. In the case of detection of rheumatoid factors (RF), the plates were coated with normal goat Ig (Jackson ImmunoResearch laboratories, Inc., West Grove, Pa.) instead of goat anti-mouse Ig and detection of mouse Ig was performed as described above. Detection of mouse isotypes in the RF assay was done using AP-labeled goat anti-mouse IgA, IgM, IgG2a, IgG2b and IgG3, as well as purified mouse IgA, IgM, IgG2a, IgG2b and IgG3 for standard curves (Southern Biotechnology Associates Inc.). All statistical comparisons were performed by analysis of variance.
  • Detection of Circulating Immune Complexes (CIC) and Precipitation of Cryoglobulins in Mouse Sera.
  • The assay was performed as previously described (June et al., 1979; Singh and Tingle, 1982) with the following modifications: ELISA plates (Corning glass works) were coated overnight at 4° C. with 5 μg/ml of human C1q (Quidel, San Diego, Calif.) in 50 mM sodium bicarbonate buffer pH 9.6. The plates were washed 3 times with PBS/0.1% Tween. Fifty μl/well of 0.3 M EDTA was added to the plates plus 50 μl/well of serum serial dilutions or solutions of known concentrations of a standard immune complex (peroxidase-mouse anti-peroxidase (PAP) from DAKO (Carpinteria, Calif.). The plates were incubated 30 min at 37° C. The plates were washed 3 times with PBS/0.1% Tween. Mouse Ig in the immune complexes were detected using 100 μl/well of a 1 μg/ml solution of an AP-labeled goat anti-mouse Ig (Southern Biotechnology Associates, Inc.) as described above for the ELISA assays. Cryoglobulins were detected by incubating overnight at 4° C. mouse serum diluted 1/15 in water and precipitates were scored visually.
  • Anti-Double Stranded (ds) and Single Stranded (ss) DNA Assays.
  • Anti-ssDNA were performed using NUNC-immuno Plate MaxiSorp plates (NUNC A/S, Denmark). Plates were coated overnight at 4° C. first with 100 μg/ml methylated BSA (Calbochem Corp., La Jolla, Calif.), then with 50 μg/ml grade I calf thymus DNA (Sigma, St. Louis, Mo.). The calf thymus DNA was sheared by sonication and then digested with S1 nuclease before use. For the anti-ssDNA assay, the DNA was boiled for 10 min and chilled on ice before use. After blocking, serial dilutions of the serum samples were added and incubated at room temperature for 2 h. Autoantibodies were detected with goat anti-mouse IgG-AP (Sigma) and develop as described above for the ELISA assays. Standard curves were obtained using known quantities of anti-DNA mAb 205, which is specific for both ss- and dsDNA (Datta et al., 1987).
  • Immunohistochemistry
  • Spleen and lymph nodes were frozen in O.C.T. embedding medium (Miles, Elkhart, Ind.) and mounted for cryostat sectioning. Sections 7-10 μm thick were dried and fixed in acetone. All Ab incubations (10 μg/ml) were done for 1 hr at room temperature in a humidified box after dilution in Tris-buffered saline A (TBS-A, 0.05M Tris, 0.15M NaCl, 0.05% Tween-20 (v/v), 0.25% BSA), rinsed in TBS-B (0.05M Tris, 0.15M NaCl, 0.05% Tween-20) and fixed 1 min in methanol before initiating the enzymatic reaction. Horseradish peroxidase (HRP) and alkaline phosphatase (AP) activities were developed using the diaminobenzidine (DAB) tablet substrate kit (Sigma) and the 5-bromo-4-chloro-3-indolyl phosphate/nitro blue tetrazolium (BCIP/NBT, Pierce, Rockford, Ill.), respectively. Stained tissue sections were finally fixed 5 min in methanol and counter stained with Giemsa (Fluka, Buchs, Switzerland). Biotin-labeled antibodies rat anti-B220, anti-CD11c, anti-syndecan-1 as well as unlabeled rat anti-CD4, anti-CD8α and anti-CD8β were purchased from Pharmingen. Biotin-labeled peanut agglutinin (PNA) was obtained from Vector laboratories (Burlingame, Calif.). (HRP)-labeled mouse anti-rat Ig and (HRP)-streptavidin were purchased from Jackson ImmunoResearch laboratories, Inc. and AP-labeled streptavidin from Southern Biotechnology Associates, Inc. In the case of immunohistochemistry on kidney tissue to detect Ig deposition, paraffin section were used, dewaxed and blocked using diluted horse serum from Vector (Burlingame, Calif.), followed by staining with HRP-goat anti-mouse Ig from Jackson Immunoresearch. Detection was performed as described above.
  • Example 1
  • BAFF Transgenic (BAFF Tg) Founder Mice have an Abnormal Phenotype.
  • Full length murine BAFF was expressed in transgenic mice using the liver specific alpha-1 antitrypsin promoter with the APO E enhancer. The full length version was chosen with the expectation that BAFF would be either cleaved and act systemically or if retained in a membrane bound form that local liver specific abnormalities would be observed possibly providing functional clues. We obtained 13 founder mice positive for the BAFF transgene (Table 2). Four of these mice died at a young age. Routine pathology was carried out on mice 811 and 816 (Table 2). There was no obvious infection in these mice; however, cardiovascular and renal abnormalities were apparent and similar to those described for severe hypertension (Fu, 1995) (Table 2). Hematoxylin and eosin (H&E)-stained kidney tissue sections of founder 816 showed that the morphology of glomeruli in that mouse was abnormal, whereas the rest of the kidney tissue seemed normal (data not shown). Many BAFF transgenic founder mice had proteinuria (Table 2). Immunohistochemistry on spleen frozen tissue sections from mouse 816, revealed an abnormal and extensive B cell staining and reduced staining for T cells and this observation was confirmed in the progeny (see below, FIG. 12).
  • Using two color FACS analysis, the ratio of % B220 positive B cells over % CD4 positive T cells was calculated. This ratio was two to seven times higher in BAFF Tg founder mice when compared to control negative BDF1 mice (Table 2), suggesting an increase of the B cell population in BAFF Tg mice. We selected nine of these founder mice to generate our different lines of transgenic mice as underlined in Table 2. None of the remaining BAFF Tg founder mice or the derived progeny showed any signs of ill health months after the early death of founders 696, 700, 811 and 816, suggesting that these 4 mice might have expressed higher levels of BAFF which caused their death. BAFF overexpression in the liver of transgenic mice was confirmed by Northern blot analysis (data not shown). In all BAFF-Tg mice examined histologically, the livers showed no abnormalities indicating that local overexpression of BAFF did not induce any immunological or pathological events. An ELISA assay for murine BAFF is not available; however, we showed that 2% serum from BAFF Tg mice, but not from control mice, blocked the binding of mammalian cell-derived mouse soluble Flag-tagged BAFF to BJAB cells. Moreover, 5% serum from BAFF Tg mice but not from control mice increased the proliferation of human B cells from PBL in the presence of anti-μ (data not shown). These data suggest that substantial amounts of soluble BAFF are present in the blood of BAFF Tg.
  • Example 2
  • Peripheral Lymphocytosis in BAFF Tg Mice is Due to Elevated B Cell Numbers
  • The transgenic mice population was found to have more lymphocytes in the blood when compared to control negative littermates, reaching values as high as 13000 lymphocytes/μl of blood (FIG. 7A). In contrast, the number of granulocytes per μl of blood in both BAFF Tg mice and control mice remained within normal limits (FIG. 7A). Since FACS analysis, using anti-CD4 and anti-B220 antibodies, of peripheral blood cells (PBL) from 18 BAFF Tg mice issued from six different founder mice showed increased B/T ratios (FIGS. 7B and 7C), the elevated lymphocyte levels resulted from an expanded B cell subset. Likewise, using this method, calculation of absolute numbers of CD4 circulating T cells revealed a 50% reduction of this T cell subset in BAFF Tg mice when compared to control mice, and the same observation was made for the CD8 T cell subset (data not shown). All B cells from the PBL of BAFF Tg mice have increased MHC class II and Bcl-2 expression when compared to B cells from control mice (FIGS. 7D and 7E, respectively), indicating some level of B cell activation in PBL of BAFF Tg mice. T cells in the blood of BAFF Tg mice did not express the early activation markers CD69 or CD25; however, 40 to 56% of CD4 or CD8 T cells were activated effector T cells with a CD44hi, L-selectinlo phenotype versus only 8% to 12% in control littermates (FIG. 7F). Thus BAFF Tg mice clearly show signs of B cell lymphocytosis and global B cell activation along with T cell alterations.
  • Example 3
  • Expanded B Cell Compartments are Composed of Mature Cells.
  • To see whether overexpression of BAFF in the transgenic mice was affecting the B cell compartment centrally in the bone marrow and peripherally in secondary lymphoid organs, we examined by FACS the spleen, bone marrow and mesenteric lymph nodes from a total of seven BAFF Tg mice and seven control littermates derived from four different founder mice. The mature B cell compartment was analyzed by staining with both anti-B220 and anti-IgM antibodies. Two representative BAFF Tg mice and one representative control littermate are shown in FIG. 8. The mature B cell compartment (IgM+. B220+) was increased in both the spleen and the mesenteric lymph nodes (FIG. 8A, top and bottom panels, respectively). Analysis of B220+/IgM+B cells (FIG. 7A, middle panel) or the proB cell (CD43+/B220+) and the preB cell (CD43−/B220+) compartments in the bone marrow (FIG. 8B) showed that BAFF Tg mice and control littermates were similar. These data indicate that overexpression of BAFF is affecting the proliferation of mature B cells in the periphery but not progenitor B cells in the bone marrow. Analysis by FACS of the B cell subpopulations in the spleen, revealed an increased proportion of marginal zone (MZ) B cells in BAFF Tg mice when compared to control mice (Table 3). The population of follicular B cells remained proportional in both BAFF Tg and control mice whereas the fraction of newly formed B cells is slightly decreased in BAFF Tg mice (Table 3). This result was also confirmed on B220+splenic B cells using anti-CD38 versus anti-CD24 antibodies and anti-IgM versus anti-IgD antibodies and analyzing for at the CD38hi/CD24+ and IgMhi/IgDlo for the MZ B cell population, respectively, as previously described (Oliver et al., 1997)(data not shown). Immunohistochemical analysis using an anti-mouse IgM antibody revealed the expansion of the IgM-bright MZ B cell area in the spleen of BAFF Tg mice when compared to control mice (data not shown). All BAFF Tg B220+splenic B cells also express higher levels of MHC class II (Table 3) and Bcl-2 (data not shown) compared to splenic B cells from control mice, indicating that splenic B cells as well as B cells from PBL are in an activated state.
  • Example 4
  • BAFF Tg Mice have High Levels of Total Immunoglobulins, Rheumatoid Factors and Circulating Immune Complexes in their Serum.
  • The increased B cell compartment in BAFF Tg mice suggested that the level of total Ig in the blood of these animals might also be increased. SDS-PAGE, analysis of serum from BAFF Tg mice and control littermates showed that the heavy and light chains IgG bands were at least 10 fold more intense in 3 out of 4 BAFF Tg mice compared to the control sera (FIG. 9A). Likewise, an ELISA determination on the sera from BAFF Tg mice show significantly higher total Ig levels when compared to that of the control mice (FIG. 9B).
  • Despite the high levels seen by SDS-PAGE, the excessively high levels of Ig seen by ELISA determination in some mice, e.g., 697-5, 816-8-3 and 823-20, led us to suspect the presence of rheumatoid factors (RF) in the sera, or autoantibodies directed against antigenic determinants on the Fc fragment of IgG (Jefferis, 1995). These antibodies could bind to the goat anti-mouse Ig used to coat the ELISA plates and give erroneously high values. ELISA plates were coated with normal irrelevant goat Ig and the binding of BAFF Tg Ig to normal goat Ig was measured. FIG. 9C shows that sera from most BAFF Tg mice contained Ig reacting with normal goat Ig, whereas only two out of 19 control mice exhibited reactivity in the same assay. These RF were mainly of the IgM, IgA and IgG2a isotypes (data not shown).
  • Presence of RF can be associated with the presence of high levels of circulating immune complexes (CIC) and cryoglobulin in the blood (Jefferis, 1995). To verify whether or not BAFF Tg mice have abnormal serum levels of CIC, a C1q-based binding assay was used to detect CIC in the 21 BAFF Tg mice analyzed above. Only 5 BAFF Tg showed significantly high levels of CIC when compared to control mice, nonetheless these mice corresponded to the animals having the highest total Ig and rheumatoid factor levels (FIG. 9D). We also observed precipitate formation when BAFF Tg mice sera were diluted 1/15 in water but not control sera indicating the presence of cryoglobulin in these mice (data not shown). Thus, in addition to B cell hyperplasia, BAFF Tg mice display severe hyperglobulinemia associated with RF and CIC.
  • Example 5
  • Some BAFF Tg Mice have High Levels of Anti-Single Stranded (ss) and Double-Stranded (ds) DNA Autoantibodies.
  • Initially, we observed kidney abnormalities reminiscent of a lupus-like disease in two of our founder mice (Table II). The presence of anti-DNA autoantibodies have also been described in SLE patients or the SLE-like (SWR×NZB)F1 (SNF1) mouse (Datta et al., 1987). Anti-ssDNA autoantibody levels were detected in BAFF Tg mice previously shown to have the highest level of total serum Ig (FIG. 10A). We analyzed the serum of two BAFF Tg mice negative for antibodies against ssDNA (697-5 and 816-1-1) and three transgenic mice secreting anti-ssDNA antibodies (820-14, 816-8-3 and 820-7) for the presence of anti-dsDNA antibodies in parallel with five control littermates. BAFF Tg mice also secreted anti-dsDNA, however, the levels of secretion did not always correlate with that of anti-ssDNA antibodies, as serum from BAFF Tg mouse 697-5 which did not contain detectable levels of anti-ssDNA antibodies, was clearly positive for the presence of anti-dsDNA (FIG. 10B). Therefore, BAFF Tg mice showing the most severe hyperglobulinemia secrete pathological levels of anti-DNA autoantibodies. Additionally, and also reminiscent of a lupus-like problem in these mice we detected immunoglobulin deposition in the kidney of six BAFF Tg mice analyzed (FIG. 10C), three of these mice did not secrete detectable levels anti-DNA antibodies (data not shown).
  • Example 6
  • BAFF Tg Mice have Enlarged B Cell Follicles, Numerous Germinal Centers, Reduced Dendritic Cell Numbers and Increased Plasma Cell Numbers in both the Spleen and Mesenteric Lymph Nodes (MLN).
  • BAFF Tg mice had large spleens, MLN (data not shown) and Peyer's patches (FIG. 11). Immunohistochemistry showed the presence of enlarged B cell follicles and reduced peripheral arteriolar lymphoid sheets (PALS or T cell area) in BAFF Tg mice (FIG. 12B). Interestingly, few germinal centers were observed in non-immunized control littermates (and is typical of this colony in general) and those present were small (FIG. 12C), whereas BAFF Tg mice possessed numerous germinal centers in the absence of immunization (FIG. 12D). Staining with anti-CD11c for dendritic cells in the T cell zone and the marginal zone of control mice (FIG. 12E) was considerably reduced in BAFF Tg mice (FIG. 12F). Syndecan-1-positive plasma cells were almost undetectable in the spleen from control littermates (FIG. 12G), yet the red pulp of BAFF Tg mice was strongly positive for syndecan-1 (FIG. 12H). Very similar observations were made for the MLN (FIG. 13). In the MLN of BAFF Tg mice the B cell areas were dramatically expanded (FIG. 13B) in contrast to the normal node where B cell follicles were easily recognizable at the periphery of the node under the capsule with a typical paracortical T cell zone (FIG. 13A). The medulla of MLN from BAFF Tg mice were filled with syndecan-1 positive cells which presumably are plasma cells (FIG. 13H). In conclusion, analysis of secondary lymphoid organs in BAFF Tg mice was consistent with the expanded B cell phenotype showing multiple cellular abnormalities and intense immune activity.
  • The Following Experimental Procedures were used in Examples 7-13.
  • Mice and Reagents.
  • Full length murine BAFF was expressed in transgenic mice using the liver-specific alpha 1 antitrypsin promoter with the Apo E enhancer as previously described. (MacKay et al. (1999) J. Exp. Med. 190:1697-1710). C57BL/6 mice were purchased from ARC (Perth, Australia). BAFF transgenic mice are maintained as heterozygotes for the transgene by backcrossing onto C57BL/6 mice. BAFF Tg mice are screened for the presence of the transgene, both by PCR and southern blot analysis using genomic DNA isolated from 2-3 mm long tail snips. (MacKay et al.). We used animals from two separate lines of BAFF Tg mice issued after 10-12 backcrossings to C57BL/6. Age-matched negative littermates were used as controls. Animals between 8 and 17 months of age were used. Animals were housed under conventional barrier protection and handled in accordance with the Animal Experimentation and Ethic Committee (AEEC), which complies with the Australian code of practice for the care and use of animals for scientific purposes. Flag-tagged soluble human BAFF (amino acids (aa) 83-285) was expressed by E. coli and purified as described previously. (Schneider et al. (1999) J. Exp. Med. 189:1747-1756.) Anti-human BAFF antibodies Buffy-2 (rat IgM), Buffy-5 (Rat IgG1) and A21G3.3 (mouse IgG1, K) were obtained after immunization of rats or mice with recombinant soluble human BAFF as previously detailed. (Schneider et al.). These antibodies recognize the TNF homology domain of soluble BAFF. The antibody A21G3.3 was purified as follows: 500 ml of medium from hybridoma cultures were diluted 1 to 1 with 0.1 M Sodium Phosphate (pH 7.2) buffer containing 150 mM of NaCl. The diluted media was loaded onto a protein-L column (Clontech, Palo Alto, Calif.) at 1 ml/minute and eluted with 0.1 M Glycine (pH 2.8). The eluted solution was neutralized with 1 M Sodium Phosphate buffer (pH 7.2). The peak fractions were confirmed by SDS-PAGE. Centricon Plus-20 (Millipore, Bedford, Mass.) was used to exchange the buffer to PBS and to concentrate the purified antibody. The purified A21G3.3 antibody was labelled with 10× mole of EZ link sulfo-NHS-LC Biotin (Pierce, Rockford, Ill.) and incubated at room temperature for 30 minutes. The biotinylation reaction was stopped with 150 mM glycine. The sample was further purified with a desalting column to remove the free biotin (Amersham Pharmacia, Uppsala, Sweden). Flow cytometry.
  • Mice were sacrificed and spleen and submaxillary glands were collected. Spleens and lymph nodes were dissociated by grinding between frosted glass slides (Menzel-Glaser, Braunschweig, Germany). Cells were filtered through a 70 μm nylon Cell Strainer (Falcon, Becton Dickinson, Franklin Lakes, N.J.) and erythrocytes (only for spleen) removed by osmotic lysis with red blood cell lysis solution (8.34 mg/ml ammonium chloride, 0.84 mg/ml sodium bicarbonate, 1 mM EDTA, pH 8.0). Submaxillary glands were cut into pieces of 2-3 millimeters and incubated in a sterile collagenase solution (Roche Diagnostics, Mannheim, Germany), 1 mg/ml in PBS (Ca2+ and Mg2+ free), for 1 h at 37° C., until leukocytes were released from the tissue. After digestion, the cell suspension was filtered through a 70 μm nylon cell strainer (Falcon) and filtered cells were washed twice with PBS. Leukocytes obtained from spleens or submaxillary glands were resuspended in FACS buffer (1% BSA, 0.05% sodium azide in PBS) at a concentration of 5×106 cells/ml. Surface staining was done using various combinations of FITC-, PE-, Cy5- and Cychome™-labelled antibodies. Fluorescent-labelled anti-mouse antibodies anti-CD4 (L3T4), anti-CD8a (Ly-2) anti-CD45R/B220 (RA3-6B2), anti-Ly6-G (GR1), anti-IgD (11-26c.2a), anti-CD11b (Mac1), anti-Ly55 (NK1.1, NKR-P1C), anti-IgM (R6-60.2), anti-CD23 (IgE Fc Receptor, clone B3B4), anti-CD24 (Heat Stable Antigen, 30F1), anti-CD43 (S7), anti-L-selectin (MEL-14), anti-CD1 (1B1) and anti-CD21 (7G6) were supplied by BD PharMingen (San Diego, Calif.). Cy5-conjugated anti-IgM antibody was purchased from Jackson ImmunoResearch laboratories Inc. (West Grove, Pa.). FITC-labelled antibodies were used diluted 1/100 whereas other fluorochrome-labelled antibodies were used at 1/200 final dilution. For flow cytometry we acquired 30,000-100,000 events per sample. Data was collected on a FACSCalibur flow cytometer and analysed using CELLQues™ software (Becton Dickinson).
  • Immunohistochemistry.
  • Spleen (removed comma) and submaxillary glands were collected from both control and BAFF Tg mice. Tissues were either frozen in OCT compound (Tissue-Tek, Sakura Finetek, Torrance, Calif., USA) or fixed in 10% buffered formalin and embedded in paraffin. Biopsies of human parotid glands were processed into paraffin blocks by pathologists at the Flinders Medical center in Adelaide. Paraffin sections, 5 μm thick, were re-hydrated in successive baths of xylene, 100% ethanol and H2O. Slides were cooked under pressure in citrate buffer (8.2 mM trisodium citrate, 1.7 mM citrate acid, pH 6.0). Slides were either stained with hematoxylin-eosin (H&E) for histologic examination or used for immunohistochemical staining. Prior to immunohistochemical staining tissue sections were pre-incubated with human Ig (Sandoz, Basel, Switzerland) 10 μg/ml in TBS-Triton (0.5% Triton) to block non-specific binding and washed twice with TBS-Triton. Sections were incubated with rat anti-human BAFF (Buffy-2) or an isotype-matched control rat antibody (Jackson ImmunoResearch Laboratories, Inc.), 5 μg/ml for 30 min at room temperature, and washed with TBS-Triton. Slides were then incubated with biotin-labelled rabbit anti-rat Ig (1/100, DAKO (Australia) PTY LTD, Botany, Australia) for 30 min at room temperature, followed by horseradish peroxidase-labelled (HRP)-Streptavidin (Jackson ImmunoResearch Laboratories Inc.) 30 min and visualized using the substrate 3,3′ diaminobenzidine (DAB) (Vector laboratories, Inc., Burlingame, Calif.). Sections were counterstained using hematoxylin (Sigma) and Scott's blueing solution and dehydrated in successive baths of H2O, 100% ethanol and xylene. Slides were mounted with cover slips and Eukitt mounting solution (Calibrated Instruments Inc., Hawthorne, N.Y.). Endogenous peroxidase activity was blocked using 2% hydrogen peroxide in methanol for 20 min before staining with the primary antibody. Frozen sections of spleen and submaxillary glands were subjected to immunohistochemical analysis as previously described (32). Biotin-labelled anti-mouse B220 and anti-mouse Syndecan-1 were purchased from BD PharMingen and the staining was detected using HRP-streptavidin (Jackson ImmunoResearch Laboratories Inc.) and visualized using DAB. All slides were observed under a Leica light microscope and images were captured using a Leica DC 200 camera (Leica, Bannockburn, Ill.). Scoring of SS disease activity in mice.
  • Tissue sections of mouse submaxillary glands stained with H&E were examined at 100× under the microscope and scored as previously described. (White et al. (1974) J. Immunol. 112:178-185). The degree of inflammatory infiltrates is graded as follows: 1 indicated that 1 to 5 foci of mononuclear cells were seen (more than 20 cells per focus), 2 indicated that more than 5 foci of mononuclear cells were seen but without significant parenchymal destruction, 3 indicated that multiple confluent foci were seen with moderate degeneration of parenchymal tissue and 4 indicated extensive infiltration of the gland with mononuclear cells and extensive parenchymal destruction.
  • Measurement of Salivary Flow.
  • Mice were anaesthetized and injected ip with 50 μg of sterile pilocarpine in PBS (Sigma, St Louis, Mo.) per 100 g body weight. After 4 min, saliva was collected for 5 min on a cotton swab. The weight of the cotton swab was measured before and after saliva collection. The amount of saliva collected was normalized to 0 g of saliva per g of body weight. Patients with primary SS and sera.
  • Sera were collected from 41 patients followed between 1995 and 2001 at Flinders Medical Centre who fulfilled at least four of six European consensus criteria for the diagnosis of primary SS. (Vitali et al. (1993) Arthritis Rheum. 36:340-347. No patient was treated with corticosteroids or immunosupressive agents. Control sera were collected from 39 healthy donors. Labial salivary gland biopsies with lymphocyte focus scores of >1 per 4 mm2 of salivary gland tissue were obtained from 4 patients with primary SS (Chisholm, et al. (1968) J. Clin. Pathol. 21:656-660), and histologically normal labial salivary gland tissues obtained from 3 controls.
  • ELISA Assays for Detection of Human BAFF in Sera from Patients with Primary SS.
  • Sera from patients were diluted 1/10 and pre-cleared from human Ig on protein-A-Sepharose beads (10% beads (pelleted beads) v/v, Amersham Pharmacia) overnight at 4° C. ELISA plates (NUNC Nalge International, Rochester, N.Y.) were coated with 2 μg/ml rat anti-human BAFF antibody (Buffy-5), overnight at 4° C. Following blocking, serial dilutions of the pre-cleared sera were added, followed by the detection antibody, biotin-conjugated mouse anti-human BAFF (0.5 μg/ml, clone A21G3.3). Alkaline phosphatase (AP)-labelled streptavidin (AP-SA, Jackson ImmunoResearch Laboratories Inc.) and the corresponding AP substrate Sigma 104 (Sigma) were used for detection. The reaction was stopped using 3N NaOH. Plates were read at an OD of 405 nm, and a standard curve was generated using known quantities of recombinant human BAFF diluted in human serum and treated as described above for patients' samples.
  • Statistical Analyses were Done using the StatView Software and ANOVA.
  • Example 7
  • BAFF Tg Mice Develop a Sjögren-Like Syndrome with Age.
  • Following routine dissections of more than 50 BAFF Tg mice and over 20 age-matched littermate controls, we observed that many mice over age 13 months had enlarged salivary (submaxillary) glands (FIG. 14A). Histological preparations from 22 BAFF Tg mice 12-17 months-old and 7 age-matched control mice were examined and scored for disease severity. The scoring system used (White et al.) takes into consideration the number of foci of infiltrating leukocytes in the gland and the extent of destruction of the parenchymal tissue.
  • A massive destruction of epithelial duct/acinar cells with small periductal foci, as well as large leukocytic infiltrates, was observed in submaxillary glands from BAFF tg animals (FIG. 14B, middle and right panel). Although small foci of infiltrating leukocytes were detected in the submaxillary glands of age-matched control mice, disease grades remained between 0 and 2, while the majority of BAFF tg mice scored 3 or greater. Overall, our analysis revealed that 40% of BAFF Tg mice over 12 months of age exhibited severe disease (grade 3 or above, FIG. 14C), an incidence which is significantly greater than that observed for control mice (FIG. 14C). Furthermore, the pathology observed in the BAFF tg mice was independent of the sex of the animals (data not shown). Lacrimal glands were not examined as mice did not develop obvious ocular disorders (keratoconjonctivitis).
  • Example 8
  • Presence of Tumor-Like Infiltrates in Salivary Glands of BAFF Tg Mice.
  • Interestingly. 3 BAFF Tg mice between 13 and 15 months-old, developed a large submaxillary tumor (over 1 cm of diameter), while no such tumors were observed in age-matched controls. Histological analysis revealed that these tumors contained hyperplastic lymphoid tissue composed mainly of activated B-lymphocytes (FIG. 15D) and numerous germinal centers (FIG. 15B). It is not known whether the tumors arose from infiltrates within submaxillary glands or from neighbouring lymph nodes, as no remaining glandular tissue was evident by histology. However, as only one submaxillary gland was found in these mice, it is likely that the tumors arose from infiltrates within the gland which ultimately overwhelmed and destroyed all glandular tissue. Unusual aggregates of cells organised in diffused foci (small lymphocytes in sinusoids) (FIG. 15A, B and C) were detected in these tumors (FIG. 15B), as well as in the large leukocyte infiltrates found in salivary glands of 4 independent BAFF Tg mice (two examples are shown in FIGS. 15A and C). Cells in these infiltrates were B-lymphoid cells expressing high levels of B220 (FIG. 15D), however they were not plasma cells since they were not positive for the plasma cell marker, syndecan-1 (FIG. 15E). A consulting pathologist indicated that because no lympho-epithelial aggregates were seen, the infiltrating B cells could not be definitively classified as malignant. Studies to examine the clonality of these unusual B cells are in progress to determine whether they are simply tumor-like clusters of lymphoid cells that do not meet criteria for malignancy or cells with real neoplastic potential.
  • Example 9
  • Submaxillary Glands from Older BAFF Tg Mice are Infiltrated by a Large Number of B-lymphocytes.
  • Seven BAFF Tg mice that had significantly larger submaxillary glands compared to age-matched control mice were selected for a detailed assessment of infiltrating cells. One gland from each mouse was digested with collagenase, after which mononuclear cells were purified and analysed by flow cytometry. Absolute counts of various cell types showed a consistent and significant increase in the number of B-lymphocytes infiltrating submaxillary glands of aging BAFF Tg mice (Table 1). Numbers of T cells, NK cells, macrophages and granulocytes also increased, however a large variation was seen between animals (Table 1). Hematoxylin and eosin staining done on the second gland collected from each Tg animal confirmed that the disease score on these tissues was at least 3 (data not shown). Mice with severe lesions in their submaxillary glands (disease grade above 3) did not secrete anti-Ro/SSA and/or anti-La/SSB autoantibodies, which are often associated with human SS (data not shown).
    TABLE 1
    Analysis of leukocytes present in salivary glands of
    BAFF transgenic mice and control littermates by flow cytometry.
    Total CD4+ CD8+ Mac-1+
    Age Leukocytes B cells T cells T cells NK cells cells GR1+ cells
    (months) (×106) (×106) (×106) (×106) (×106) (×106) (×106)
    Control
    Littermates
    Control
    1 14 8.4 0.048 ND 0.04 0.22 ND
    Control
    2 14 5.6 0.045 ND 0.03 0.17 ND
    Control
    3 14 5.9 0.028 ND 0.02 0.23 ND
    Control
    4 15 1.5 0.0014 0.0082 0.0011 0.02 0.57 0.062
    Mean ± SD 14.25 ± 0.5  5.35 ± 2.8  0.03 ± 0.02 0.027 ± 0.009 0.29 ± 0.18
    BAFF Tg
    mice
    BAFF Tg
    1 14 30 1.1 ND 0.27 0.7 ND
    BAFF Tg
    2 14 31.7 0.14 ND 0.31 1.03 ND
    BAFF Tg
    3 16 42.3 0.6 ND 0.02 0.23 ND
    BAFF Tg
    4 8 6 0.35 0.039 0.028 0.13 0.29 0.45
    BAFF Tg 5 16 11 4.6 0.084 1.6 0.64 0.27 4.3
    BAFF Tg 6 17 2.5 0.03 0.027 0.022 0.087 0.17 0.23
    BAFF Tg 7 16 22 9.3 0.34 1.6 1.4 0.5 8.7
    Mean ± SD 14.4 ± 3   21 ± 14 2.3 ± 3.5 0.41 ± 0.5  0.46 ± 0.31
    p values P < 0.05 P < 0.05

    Animals were sacrificed and submaxillary glands dissected. Tissues were digested and leukocytes purified as described in materials and methods. The Total number of leukocytes per gland was counted using an hemocytometer. Cells were stained with anti-B220 (B cells), anti-CD4 and anti-CD8 (T cells), anti-NK1.1 (NK cells), anti-Mac1 (macrophages) and anti-GR1 (granulocytes) fluorochrome-labelled antibodies. Flow cytometry analysis provided percentage values for each subpopulation
    # of cells and absolute numbers were calculated relative to the total number of cells collected per gland.

    ND: not detected,

    —: not done.

    Only significant p values are indicated.
  • Example 10
  • A Large Proportion of B Cells Infiltrating Submaxillary Glands of BAFF Tg Mice have a Marginal Zone (MZ)-Like Phenotype.
  • A flow cytometric profiling analysis of B cells infiltrating the submaxillary glands of BAFF Tg mice revealed two B cell subsets based on their level of expression of B220 (FIG. 16A). Analysis of the B-1 B cell population in the B220 low/intermediate (B220lo/int) gate revealed a 3-fold increase of B-1b (CD5) B cells and presence of B-1a (CD5+) B cells not detected in control mice (FIG. 16B). In the B220 high (B220hi) gate, cells were further analysed based on their level of IgM expression. The IgM dull (IgMdull) subset of B cells is present in both control and BAFF Tg mice, however this population is larger in BAFF Tg mice compared to control animals (FIG. 16B). The IgMdull subset in both control and BAFF Tg mice contains B-2-like cells, which are CD21lo/int, IgD+ and CD23+ (data not shown). The B220hi/IgMhi B cell subset is greatly increased in BAFF Tg mice compared to control mice (FIG. 16B). Moreover, the IgMhi B cells found in BAFF Tg mice are phenotypically different than IgMhi B cells from control mice. Indeed, the small IgMhi B cell subset detected in control mice had characteristics of B-2 B cells (CD21lo/int, IgD+ and CD23+, add HSA and L-selectin (FIG. 16D, 16E), whereas the large IgMhi B cell subset of BAFF Tg mice had no counterpart in control mice and resembled MZ B cells in many respects. These cells were IgDlo, CD21hi, CD23lol−, CD1hi, HSA++ (FIG. 16E) and L-selectinlo (FIG. 16D). These cells also express lower levels of CD43 when compared to B-1 B cells (FIG. 16C). We therefore-refer to these-cells as M-Z-like-B-cells-because their profile is closer to that of MZ B cells than to other phenotypically-related B cell subsets such as B-1 and splenic transitional T1 B cells (FIG. 16F). (Wells et al. (1994) J. Immunol. 153:5503-5515; Amano et al. (1998) J. Immunol. 161:1710-1717). However, these cells also share some similarities with B-1b cells, such as low expression of CD43.
  • Example 11
  • Older BAFF Tg Mice Exhibit Impaired Saliva Production.
  • The inflammation seen in the salivary glands of BAFF Tg mice was reminiscent of the inflammation described for SS patients. Therefore, we aimed to determine if the inflammation in BAFF Tg mice caused an impairment of normal saliva production. To this end, 13 BAFF Tg mice and 14 control littermates between 8 and 15.5 months of age received pilocarpine, a known stimulant for salivary flow, 4 min prior to the collection and measurement of saliva. We found that mice between 13 and 15 months of age had a significantly reduced production of saliva when compared to age-matched control animals (FIG. 17A). However, this difference was not seen with mice between 8 and 10 months of age (FIG. 17B). Interestingly, reduction in saliva flow correlated with the highest numbers of B-lymphocytes detected in submaxillary glands of these Tg animals (data not shown).
  • Example 12
  • Elevated Levels of BAFF Detected in the Serum of Patients Suffering from Primary Sjögren's Syndrome.
  • As the Sjögren's-like pathology observed in some BAFF Tg mice showed similar features to human SS, we measured BAFF levels in sera of patients suffering from this disease. Sera from 41 patients with primary SS and 39 healthy individuals were analysed using a human BAFF-specific ELISA assay. This assay showed that at least 15 patients out of 41 (36%) clearly had higher levels of serum BAFF when compared to healthy individuals (FIG. 18A). Two patients had very high BAFF levels (over 200 ng/ml, FIG. 18A). BAFF levels in patients' sera did not correlate with the levels of total IgG or RF (FIG. 18B and C, respectively). These BAFF levels also did not correlate with the presence of precipitins such as anti-Ro and/or anti-La (FIG. 18D). We confirmed human BAFF levels in patients' sera using BAFF-specific immunoprecipitation procedures followed by western blotting techniques (data not shown).
  • Example 13
  • Detection of BAFF Expressing Cells in Tissue Sections from Labial Glands of Patients with SS.
  • We examined the expression of BAFF at the site of abnormal lymphocyte infiltrates in biopsies of labial glands from SS patients. A strong BAFF+ signal was detected on leukocytic infiltrates within patient tissues, although clearly not all cells were positive for BAFF within the infiltrate (FIG. 18E, right panel). The positive signal detected was specific since it could be competed away by the addition of recombinant human BAFF (data not shown). The antibody did not stain normal labial salivary gland tissue (FIG. 18E, bottom left). Preliminary experiments using 2-color staining on tissues indicate that infiltrating macrophages/monocytes are the likely source of BAFF in these inflamed tissues (data not shown).
  • REFERENCES
    • 1. Smith et al. (1994) Cell 76:959-962.
    • 2. Vassalli (1992) Annu. Rev. Immunol. 10:411-452.
    • 3. De Togni et al. (1994) Science 264:703-707.
    • 4. Koni et al. (1997) Immunity 6:491-500.
    • 5. Amakawa et al. (1996) Cell 84:551-562.
    • 6. Russell et al. (1993) Proc. Natl. Acad. Sci. USA 90:4409-4413.
    • 7. Zheng et al. (1995) Nature 377:348-351.
    • 8. van Kooten and Banchereau (1997) Curr. Opin. Immunol. 9:330-337.
    • 9. Stuber and Strober (1996). J. Exp. Med. 183:979-989.
    • 10. Schneider et al. (1997) J. Biol. Chem. 272:18827-18833.
    • 11. Hahne et al. (1998) J. Exp. Med. 188:1185-1190.
    • 12. Hahne et al. (1996) Science 274:1363-1366.
    • 13. Grimaitre et al. (1997) Eur. J. Immunol. 27:199-205.
    • 14. Thome et al. (1997) Nature 386:517-521.
    • 15. Schneider et al. (1998) J. Exp. Med. 187:1205-1213.
    • 16. Matsudaira, P. (1987) J. Biol. Chem. 262:10035-10038.
    • 17. Armitage et al. (1992) Nature 357:80-82.
    • 18. Bucher et al. (1996) Computer Chem. 20:3-24.
    • 19. Banner et al. (1993) Cell 73:431-445.
    • 20. Nagata (1997) Cell 88:355-365.
    • 21. Black et al. (1997) Nature 385:729-733.
    • 22. Wong et al. (1997) J. Biol. Chem. 272:25190-25194.
    • 23. Kindler and Zubler. (1997) J. Immunol. 159:2085-2090.
    • 24. Sonoki et al. (1995) Leukemia 9:2093-2099.
    • 25. Magrath, I. (1990) Adv Cancer Res 55:133-270.
    • 26. Garside et al. (1998) Science 281:96-99.
    • 27. MacLennan et al. (1997) Immunol. Rev. 156:53-66.
    • 28. Dubois et al. (1997). J. Exp. Med. 185:941-951.
    • 29. Tsubata et al. (1993) Nature 364:645-648.
    • 30. Chicheportiche et al. (1997) J. Biol. Chem. 272:32401-32410.
    • 31. Nakayama (1997) Biochem. J. 327:625-635.
    • 32. Jefferis, R. (1995). Rheumatoid factors, B cells and immunoglobulin genes. Br. Med. Bull. 51, 312-331.
    • 33. Schneider et al. (1999) J. Exp. Med. 189, 1747-1756.
    • 34. Mcknights et al. (1983) Cell 34, 335-341.
    • 35. Datta et al. (1987) J. Exp. Med. 165, 1252-1261.

Claims (27)

1-59. (canceled)
60. A method of treating a mammal, the method comprising administering to a mammal having Sjögren's syndrome a composition comprising a BAFF blocking agent, thereby reducing immunoglobulin production or B cell growth in the mammal.
61. The method of claim 60, wherein the mammal is a human.
62. The method of claim 60, wherein the mammal is a mouse.
63. The method of claim 62, wherein the mouse is a BAFF Tg mouse.
64. The method of claim 60, wherein the salivary gland of the mammal is infiltrated by MZ-like B cells.
65. The method of claim 60, wherein the BAFF blocking is selected form the group consisting of:
(a) a soluble BAFF receptor;
(b) an antibody against BAFF ligand;
(c) an antibody against BAFF receptor; and
(d) an inoperative BAFF ligand molecule.
66. The method of claim 65, wherein soluble BAFF receptor comprises an immunoglobulin Fc domain.
67. The method of claim 65, wherein the BAFF receptor is BAFF-R.
68. The method of claim 65, wherein the BAFF receptor is TACI.
69. The method of claim 65, wherein the BAFF receptor is BCMA.
70. The method of claim 67, wherein BAFF-R is human.
71. The method of claim 65, wherein the soluble BAFF receptor comprises a portion of SEQ ID NO:27 that binds to BAFF.
72. The method of claim 71, wherein the soluble BAFF receptor further comprises a human immunoglobulin Fc domain.
73. The method of claim 67, wherein BAFF-R is murine.
74. The method of claim 70, wherein the soluble BAFF receptor comprises a portion of SEQ ID NO:28 that binds to BAFF.
75. The method of claim 65, wherein the antibody against BAFF ligand or BAFF receptor is a monoclonal antibody.
76. The method of claim 75, wherein the antibody is recombinantly produced.
77. The method of claim 75, wherein the antibody is a chimeric antibody.
78. The method of claim 75, wherein the antibody is a humanized antibody.
79. The method of claim 75, wherein the antibody comprises human constant domains.
80. The method of claim 75, wherein the antibody is a F(ab′)2 fragment.
81. The method of claim 60, further comprising detecting the level of B cell growth in the mammal.
82. The method of claim 60, further comprising detecting the level of immunoglobulin production in the mammal.
83. The method of claim 60, further comprising detecting the levels of B cell growth and immunoglobulin production in the mammal.
84. The method of claim 60, further comprising detecting circulating levels of a rheumatoid factor in the mammal.
85. The method of claim 60, further comprising detecting circulating levels of anti-DNA autoantibody in the mammal.
US11/065,669 1999-01-25 2005-02-24 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders Abandoned US20050244411A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/065,669 US20050244411A1 (en) 1999-01-25 2005-02-24 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US12/061,398 US9545086B2 (en) 1999-01-25 2008-04-02 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US11716999P 1999-01-25 1999-01-25
US14322899P 1999-07-09 1999-07-09
PCT/US2000/001788 WO2000043032A2 (en) 1999-01-25 2000-01-25 Baff, inhibitors thereof and their use in the modulation of b-cell response
US09/911,777 US6869605B2 (en) 1999-01-25 2001-07-24 BAFF, inhibitors thereof and their use in the modulation of B-cell response
US10/045,574 US20030095967A1 (en) 1999-01-25 2001-11-07 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US11/065,669 US20050244411A1 (en) 1999-01-25 2005-02-24 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/045,574 Continuation US20030095967A1 (en) 1999-01-25 2001-11-07 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/061,398 Division US9545086B2 (en) 1999-01-25 2008-04-02 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders

Publications (1)

Publication Number Publication Date
US20050244411A1 true US20050244411A1 (en) 2005-11-03

Family

ID=46150035

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/045,574 Abandoned US20030095967A1 (en) 1999-01-25 2001-11-07 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US11/065,669 Abandoned US20050244411A1 (en) 1999-01-25 2005-02-24 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US12/061,398 Active - Reinstated 2027-04-11 US9545086B2 (en) 1999-01-25 2008-04-02 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/045,574 Abandoned US20030095967A1 (en) 1999-01-25 2001-11-07 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/061,398 Active - Reinstated 2027-04-11 US9545086B2 (en) 1999-01-25 2008-04-02 BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders

Country Status (1)

Country Link
US (3) US20030095967A1 (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050186637A1 (en) * 1999-03-02 2005-08-25 Guo-Liang Yu Neutrokine-alpha and neutrokine-alpha splice variant
US20060062789A1 (en) * 2000-06-16 2006-03-23 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to B lymphocyte stimulator protein
US20060084608A1 (en) * 2000-08-18 2006-04-20 Human Genome Sciences, Inc. Binding polypeptides and methods based thereon
US20060198784A1 (en) * 1996-10-25 2006-09-07 Human Genome Sciences, Inc. Methods of treatment using antibodies to Neutrokine-alpha
US20090081213A1 (en) * 2005-10-13 2009-03-26 Human Genome Sciences, Inc. Methods and compositions for use in treatment of patients with autoantibody positive disease
US20090148462A1 (en) * 2005-10-13 2009-06-11 Human Genome Sciences, Inc. Methods and compositions for use in treatment of patients with autoantibody positive disease
US20090297504A1 (en) * 2004-01-29 2009-12-03 Genentech, Inc. Bcma polypeptides and uses thereof
US7879328B2 (en) 2000-06-16 2011-02-01 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to B lymphocyte stimulator
WO2012047093A1 (en) * 2010-10-05 2012-04-12 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Treatment of the sjögren's syndrome
US8211649B2 (en) 2006-03-31 2012-07-03 Human Genome Sciences, Inc. Methods of diagnosing and prognosing hodgkin's lymphoma

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030095967A1 (en) * 1999-01-25 2003-05-22 Mackay Fabienne BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US20050100548A1 (en) * 2001-07-24 2005-05-12 Biogen Idec Ma Inc. BAFF, inhibitors thereof and their use in the modulation of B-cell response
IL150755A0 (en) * 2000-02-16 2003-02-12 Genentech Inc Uses of agonists and antagonists to modulate activity of tnf-related molecules
UA83458C2 (en) * 2000-09-18 2008-07-25 Байоджен Айдек Ма Інк. The isolated polypeptide baff-r (the receptor of the factor of activation of b-cells of the family tnf)
ATE549354T1 (en) * 2001-05-11 2012-03-15 Amgen Inc PEPTIDES AND ASSOCIATED TALL-1 BINDING MOLECULES
WO2003014294A2 (en) * 2001-08-03 2003-02-20 Genentech, Inc. Tacis and br3 polypeptides and uses thereof
WO2003072713A2 (en) * 2002-02-21 2003-09-04 Biogen Idec Ma Inc. Use of bcma as an immunoregulatory agent
JP2005533863A (en) * 2002-07-25 2005-11-10 ジェネンテック・インコーポレーテッド TACI antibodies and their uses
AU2004233164B2 (en) 2003-03-28 2009-10-08 Biogen Ma Inc. Truncated BAFF receptors
ES2537738T3 (en) * 2003-06-05 2015-06-11 Genentech, Inc. Combination therapy for B cell disorders
US20050163775A1 (en) * 2003-06-05 2005-07-28 Genentech, Inc. Combination therapy for B cell disorders
US7850967B2 (en) 2003-10-20 2010-12-14 Biogen Idec Ma Inc. Method of treating a patient having an autoimmune disorder by administering an antibody that binds human BAFFR
WO2006012508A2 (en) * 2004-07-22 2006-02-02 Genentech, Inc. Method of treating sjögren's syndrome
AU2005295713B2 (en) 2004-10-13 2011-06-16 The Washington University Use of BAFF to treat sepsis
CA2596276A1 (en) * 2005-01-28 2006-08-03 Biogen Idec Ma Inc. Use of baff to treat th2-mediated conditions
JP6088723B2 (en) * 2005-11-23 2017-03-01 ジェネンテック, インコーポレイテッド Compositions and methods for B cell assays.
US8435510B2 (en) * 2007-08-08 2013-05-07 Sutter West Bay Hospitals Platelet derived growth factor receptor supports cytomegalovirus infectivity
EP2396035A4 (en) * 2009-02-12 2012-09-12 Human Genome Sciences Inc Use of b lymphocyte stimulator protein antagonists to promote transplantation tolerance
US9458246B2 (en) 2013-03-13 2016-10-04 Amgen Inc. Proteins specific for BAFF and B7RP1
JOP20140087B1 (en) 2013-03-13 2021-08-17 Amgen Inc Proteins specific for baff and b7rp1 and uses thereof
KR102480433B1 (en) 2014-02-07 2022-12-21 맥마스터 유니버시티 Trifunctional t cell-antigen coupler and methods and uses thereof
WO2019035938A1 (en) 2017-08-16 2019-02-21 Elstar Therapeutics, Inc. Multispecific molecules that bind to bcma and uses thereof
JP7227630B2 (en) 2017-10-12 2023-02-22 マックマスター ユニバーシティー T cell-antigen coupler with Y182T mutation and methods and uses thereof
EP3818083A2 (en) 2018-07-03 2021-05-12 Elstar Therapeutics, Inc. Anti-tcr antibody molecules and uses thereof
US11110123B2 (en) 2018-07-17 2021-09-07 Triumvira Immunologics Usa, Inc. T cell-antigen coupler with various construct optimizations
CN112516303A (en) * 2020-11-18 2021-03-19 华中科技大学同济医学院附属协和医院 Application of BAFF antibody in preparation of sepsis drug
BR112023020832A2 (en) 2021-04-08 2023-12-19 Marengo Therapeutics Inc TCR-BINDED MULTIFUNCTIONAL MOLECULES AND THEIR USES
US11453723B1 (en) 2021-06-25 2022-09-27 Mcmaster University BCMA T cell-antigen couplers and uses thereof

Citations (43)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US640377A (en) * 1899-06-09 1900-01-02 Mathias Haentges Crib for building piers, walls, &c.
US5595721A (en) * 1993-09-16 1997-01-21 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20
US5969102A (en) * 1997-03-03 1999-10-19 St. Jude Children's Research Hospital Lymphocyte surface receptor that binds CAML, nucleic acids encoding the same and methods of use thereof
US20010010925A1 (en) * 1997-11-17 2001-08-02 Steven R. Wiley Methods of detecting target nucleic acids of tnf-delta
US6297367B1 (en) * 1997-12-30 2001-10-02 Chiron Corporation Polynucleotide encoding TNFL1
US20020004587A1 (en) * 2000-04-11 2002-01-10 Genentech, Inc. Multivalent antibodies and uses therefor
US20020037852A1 (en) * 1999-01-25 2002-03-28 Jeffrey Browning BAFF, inhibitors thereof and their use in the modulation of B-cell response
US20020055624A1 (en) * 1997-11-17 2002-05-09 Steven R. Wiley Tnf-delta ligand and uses thereof
US20020064829A1 (en) * 1996-03-14 2002-05-30 Human Genome Sciences, Inc. Human tumor necrosis factor delta and epsilon
US20020115112A1 (en) * 1999-02-23 2002-08-22 Human Genome Sciences, Inc. Neutrokine-alpha and Neutrokine-alpha splice variant
US6475986B1 (en) * 1999-02-02 2002-11-05 Research Development Foundation Uses of THANK, a TNF homologue that activates apoptosis
US6475987B1 (en) * 1999-05-06 2002-11-05 National Jewish Medical And Research Center Tall-1 receptor homologues
US20020165156A1 (en) * 1999-08-17 2002-11-07 Jeffrey Browning BAFF receptor (BCMA), an immunoregulatory agent
US20030022239A1 (en) * 1997-06-18 2003-01-30 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030023038A1 (en) * 2000-02-11 2003-01-30 Paul Rennert Heterologous polypeptide of the TNF family
US20030053984A1 (en) * 1999-12-30 2003-03-20 Jurg Tschopp Bimer or an oligomer of a dimer, trimer, quadromer or pentamer of recombinant fusion proteins
US20030082175A1 (en) * 1999-10-06 2003-05-01 Pascal Schneider April receptor (BCMA) and uses thereof
US20030095967A1 (en) * 1999-01-25 2003-05-22 Mackay Fabienne BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US20030100074A1 (en) * 2001-08-16 2003-05-29 Guo-Liang Yu Methods and compositions for treating metabolic bone diseases relating to human endokine alpha
US20030100068A1 (en) * 2001-10-12 2003-05-29 Jonathan Lam RANKL mimics and uses thereof
US20030108992A1 (en) * 2000-02-11 2003-06-12 Lenardo Michael J. Identification of a novel domain in the tumor necrosis factor receptor family that mediates pre-ligand receptor assembly and function
US20030119149A1 (en) * 2001-12-21 2003-06-26 Pranhitha Reddy Recombinant polypeptides
US6587906B2 (en) * 1999-12-22 2003-07-01 Intel Corporation Parallel multi-threaded processing
US20030148454A1 (en) * 2001-11-02 2003-08-07 Trustees Of Boston University Virus-free vesicles for delivery of functional membrane bound proteins
US20030166864A1 (en) * 1996-03-14 2003-09-04 Guo-Liang Yu Human tumor necrosis factor delta and epsilon
US20030175208A1 (en) * 1996-10-25 2003-09-18 Human Genome Sciences, Inc. Neutrokine-alpha and neutrokine-alpha splice variant
US6623941B1 (en) * 2000-05-05 2003-09-23 Human Genome Sciences, Inc. Nucleic acids encoding human tumor necrosis factor TR20
US20030194743A1 (en) * 2000-08-18 2003-10-16 Beltzer James P. Binding polypeptides for B lymphocyte stimulator protein (BLyS)
US20030194745A1 (en) * 1998-06-26 2003-10-16 Mcdowell Robert S. Cysteine mutants and methods for detecting ligand binding to biological molecules
US20030198640A1 (en) * 1994-11-07 2003-10-23 Human Genome Sciences, Inc. Methods and compositions for treating inflammatory bowel diseases relating to human tumor necrosis factor-gamma-beta
US20030211509A1 (en) * 2002-03-26 2003-11-13 Wiley Steven R. TNF-delta ligand and uses thereof
US6667920B2 (en) * 1999-12-28 2003-12-23 Intel Corporation Scratchpad memory
US20040013674A1 (en) * 2001-04-27 2004-01-22 Christine Ambrose Taci as an anti-tumor agent
US6689579B1 (en) * 1996-10-25 2004-02-10 Human Genome Sciences, Inc. Polynucleotides encoding neutrokine-α
US20040028658A1 (en) * 2002-06-27 2004-02-12 Denise Faustman Methods of organ regeneration
US20040033511A1 (en) * 2000-09-15 2004-02-19 Klaus Pfizenmaier Site-specific antibody-mediated activation of proapoptotic cytokines: AMAIZEe (antibody-mediated apoptosis inducing cytokines)
US20040038349A1 (en) * 2001-07-27 2004-02-26 Hilbert David M. Heteromultimeric TNF ligand family members
US20040039895A1 (en) * 2000-01-05 2004-02-26 Intel Corporation, A California Corporation Memory shared between processing threads
US20040054880A1 (en) * 1999-08-31 2004-03-18 Intel Corporation, A California Corporation Microengine for parallel processor architecture
US20040072188A1 (en) * 2000-09-18 2004-04-15 Ambrose Christine M. Novel receptor nucleic acids and polypeptides
US6728845B2 (en) * 1999-08-31 2004-04-27 Intel Corporation SRAM controller for parallel processor architecture and method for controlling access to a RAM using read and read/write queues
US20040098496A1 (en) * 1999-12-28 2004-05-20 Intel Corporation, A California Corporation Thread signaling in multi-threaded network processor
US20060193859A1 (en) * 1996-10-25 2006-08-31 Human Genome Sciences, Inc. Methods of Treatment Using Antibodies to Neutrokine-alpha

Family Cites Families (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE69630710T2 (en) 1996-03-14 2004-09-23 Human Genome Sciences Inc. HUMAN TUMORNE CROSS FACTOR DELTA AND EPSILON
JP2001503263A (en) 1996-10-25 2001-03-13 ヒューマン ジノーム サイエンシーズ,インコーポレイテッド Neutrokine α
AU5705898A (en) 1996-12-17 1998-07-15 Schering Corporation Mammalian cell surface antigens; related reagents
CA2232743A1 (en) 1997-04-02 1998-10-02 Smithkline Beecham Corporation A tnf homologue, tl5
IL133315A0 (en) 1997-06-06 2001-04-30 Regeneron Pharma Ntn-2 member of tnf ligand family
WO1998055621A1 (en) 1997-06-06 1998-12-10 Regeneron Pharmaceuticals, Inc. Ntn-2 member of tnf ligand family
WO1999004001A1 (en) 1997-07-21 1999-01-28 Zymogenetics, Inc. Tumor necrosis factor receptor ztnfr-5
WO1999011791A2 (en) 1997-09-05 1999-03-11 University Of Washington Tumor necrosis factor family receptors and ligands, encoding nucleic acids and related binding agents
CN1195849C (en) 1997-09-12 2005-04-06 阿普泰克R&D公司 APPIL-a novel protein with growth effects
EA200000311A1 (en) 1997-09-12 2000-10-30 Апотек Р Энд Д Са NEW PROTEIN IMMUNE SYSTEM - KAU
JP2002503444A (en) 1997-11-26 2002-02-05 イーライ・リリー・アンド・カンパニー Ligand family genes
AU2093499A (en) 1997-12-30 1999-07-19 Chiron Corporation Members of tnf and tnfr families
WO2000026244A2 (en) 1998-11-04 2000-05-11 The Government Of The United States Of America, As Represented By The Secretary, Department Of Health And Human Services A novel tumor necrosis factor family member, drl, and related compositions and methods
US7300774B1 (en) 1999-12-09 2007-11-27 The Regents Of The University Of California Multimeric fusion proteins of the TNF superfamily ligands
GB9828628D0 (en) 1998-12-23 1999-02-17 Glaxo Group Ltd Novel ligand
ATE453712T1 (en) 1999-01-07 2010-01-15 Zymogenetics Inc METHOD FOR THERAPEUTIC USE OF SOLUBLE RECEPTORS BR43X2
US7833529B1 (en) * 1999-01-07 2010-11-16 Zymogenetics, Inc. Methods for inhibiting B lymphocyte proliferation with soluble ztnf4 receptor
WO2000047740A2 (en) 1999-02-12 2000-08-17 Amgen Inc. Tnf-related proteins
CA2363112A1 (en) 1999-02-23 2000-08-31 Craig A. Rosen Neutrokine-alpha and neutrokine-alpha splice variant
JP2002541779A (en) 1999-02-24 2002-12-10 ザ ジュネラル ホスピタル コーポレーション Cloning method of signaling intermediate
AU3633000A (en) 1999-03-26 2000-10-16 Human Genome Sciences, Inc. Neutrokine-alpha binding proteins and methods based thereon
AU5308200A (en) 1999-06-11 2001-01-02 Human Genome Sciences, Inc. 48 human secreted proteins
US20030166003A1 (en) 1999-06-14 2003-09-04 Cochran Andrea G. Structured peptide scaffold for displaying turn libraries on phage
EP1690872A3 (en) 1999-12-01 2006-08-23 Genentech, Inc. Composition and methods for the diagnosis of tumours
JP4723782B2 (en) 2000-01-03 2011-07-13 ティーアール アソシエイツ,エル.エル.シー. Novel chimeric protein and method of using the protein
AU2001274888A1 (en) 2000-05-19 2001-12-03 Human Genome Sciences, Inc. Nucleic acids, proteins, and antibodies
WO2001096528A2 (en) 2000-06-15 2001-12-20 Human Genome Sciences, Inc. Human tumor necrosis factor delta and epsilon
KR101287395B1 (en) 2000-06-16 2014-11-04 휴먼 게놈 사이언시즈, 인코포레이티드 Antibodies that immunospecifically bind to BLyS
CA2416538A1 (en) 2000-07-20 2002-01-31 Genentech, Inc. Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
WO2002009262A1 (en) 2000-07-20 2002-01-31 Koninklijke Philips Electronics N.V. Switched mode power supply control
CA2419661A1 (en) 2000-08-15 2002-03-07 Guo-Liang Yu Neutrokine-alpha and neutrokine-alpha splice variant
US20020052318A1 (en) 2000-08-25 2002-05-02 Duke University Adjuvant
ES2329012T3 (en) 2000-11-07 2009-11-20 Zymogenetics, Inc. RECEIVER OF THE HUMAN TUMOR NECROSIS FACTOR.
WO2003001877A2 (en) 2001-06-26 2003-01-09 Gene Logic, Inc. Methods for the diagnosis and treatment of cardiac tissue rejection
WO2003014294A2 (en) 2001-08-03 2003-02-20 Genentech, Inc. Tacis and br3 polypeptides and uses thereof
CA2459890A1 (en) 2001-09-06 2003-03-20 Biogen Idec Ma Inc. Crystal structure of baff, and use thereof in drug design
US7256015B2 (en) 2001-09-21 2007-08-14 Amgen Inc. TALL-1 receptor molecules and uses thereof
WO2003033658A2 (en) 2001-10-17 2003-04-24 Human Genome Sciences, Inc. Neutrokine-alpha and neutrokine-alpha splice variant
CA2465268A1 (en) 2001-10-24 2003-05-01 National Jewish Medical And Research Center Three-dimensional structures of tall-1 and its cognate receptors and modified proteins and methods related thereto
AU2002364696A1 (en) 2001-11-07 2003-06-23 Human Genome Sciences, Inc. Crystalline neutrokine-alpha protein, method of preparation thereof, and method of use thereof
AU2002364954A1 (en) 2001-11-16 2003-07-15 Human Genome Sciences, Inc. ANTIBODIES THAT IMMUNOSPECIFICALLY BIND TO BLyS
EP1997829A1 (en) 2001-12-21 2008-12-03 Human Genome Sciences, Inc. Albumin fusion proteins
AU2003254001A1 (en) 2002-08-14 2004-03-03 Eli Lilly And Company Novel htnfsf13b protein variants
ES2537738T3 (en) 2003-06-05 2015-06-11 Genentech, Inc. Combination therapy for B cell disorders

Patent Citations (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US640377A (en) * 1899-06-09 1900-01-02 Mathias Haentges Crib for building piers, walls, &c.
US5595721A (en) * 1993-09-16 1997-01-21 Coulter Pharmaceutical, Inc. Radioimmunotherapy of lymphoma using anti-CD20
US20030198640A1 (en) * 1994-11-07 2003-10-23 Human Genome Sciences, Inc. Methods and compositions for treating inflammatory bowel diseases relating to human tumor necrosis factor-gamma-beta
US20020064829A1 (en) * 1996-03-14 2002-05-30 Human Genome Sciences, Inc. Human tumor necrosis factor delta and epsilon
US20030166864A1 (en) * 1996-03-14 2003-09-04 Guo-Liang Yu Human tumor necrosis factor delta and epsilon
US6541224B2 (en) * 1996-03-14 2003-04-01 Human Genome Sciences, Inc. Tumor necrosis factor delta polypeptides
US6689579B1 (en) * 1996-10-25 2004-02-10 Human Genome Sciences, Inc. Polynucleotides encoding neutrokine-α
US20060198784A1 (en) * 1996-10-25 2006-09-07 Human Genome Sciences, Inc. Methods of treatment using antibodies to Neutrokine-alpha
US20030175208A1 (en) * 1996-10-25 2003-09-18 Human Genome Sciences, Inc. Neutrokine-alpha and neutrokine-alpha splice variant
US20060193859A1 (en) * 1996-10-25 2006-08-31 Human Genome Sciences, Inc. Methods of Treatment Using Antibodies to Neutrokine-alpha
US6316222B1 (en) * 1997-03-03 2001-11-13 St. Jude Children's Research Hospital Nucleic acids encoding a lymphocyte surface receptor that binds CAML
US5969102A (en) * 1997-03-03 1999-10-19 St. Jude Children's Research Hospital Lymphocyte surface receptor that binds CAML, nucleic acids encoding the same and methods of use thereof
US20030022239A1 (en) * 1997-06-18 2003-01-30 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20020055624A1 (en) * 1997-11-17 2002-05-09 Steven R. Wiley Tnf-delta ligand and uses thereof
US20010010925A1 (en) * 1997-11-17 2001-08-02 Steven R. Wiley Methods of detecting target nucleic acids of tnf-delta
US6297367B1 (en) * 1997-12-30 2001-10-02 Chiron Corporation Polynucleotide encoding TNFL1
US20030194745A1 (en) * 1998-06-26 2003-10-16 Mcdowell Robert S. Cysteine mutants and methods for detecting ligand binding to biological molecules
US20030095967A1 (en) * 1999-01-25 2003-05-22 Mackay Fabienne BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US6869605B2 (en) * 1999-01-25 2005-03-22 Biogen Idec Ma Inc. BAFF, inhibitors thereof and their use in the modulation of B-cell response
US20020037852A1 (en) * 1999-01-25 2002-03-28 Jeffrey Browning BAFF, inhibitors thereof and their use in the modulation of B-cell response
US6475986B1 (en) * 1999-02-02 2002-11-05 Research Development Foundation Uses of THANK, a TNF homologue that activates apoptosis
US20030166546A1 (en) * 1999-02-02 2003-09-04 Research Development Foundation Uses of THANK, a TNF homologue that activates apoptosis
US20020115112A1 (en) * 1999-02-23 2002-08-22 Human Genome Sciences, Inc. Neutrokine-alpha and Neutrokine-alpha splice variant
US6475987B1 (en) * 1999-05-06 2002-11-05 National Jewish Medical And Research Center Tall-1 receptor homologues
US20030148445A1 (en) * 1999-05-06 2003-08-07 National Jewish Medical And Research Center TALL-1 nucleic acid molecules, proteins, receptors and methods of use thereof
US20020165156A1 (en) * 1999-08-17 2002-11-07 Jeffrey Browning BAFF receptor (BCMA), an immunoregulatory agent
US20020172674A1 (en) * 1999-08-17 2002-11-21 Browning Jeffrey BAFF receptor (BCMA), an immunoregulatory agent
US6728845B2 (en) * 1999-08-31 2004-04-27 Intel Corporation SRAM controller for parallel processor architecture and method for controlling access to a RAM using read and read/write queues
US20040054880A1 (en) * 1999-08-31 2004-03-18 Intel Corporation, A California Corporation Microengine for parallel processor architecture
US20030082175A1 (en) * 1999-10-06 2003-05-01 Pascal Schneider April receptor (BCMA) and uses thereof
US6587906B2 (en) * 1999-12-22 2003-07-01 Intel Corporation Parallel multi-threaded processing
US6667920B2 (en) * 1999-12-28 2003-12-23 Intel Corporation Scratchpad memory
US20040098496A1 (en) * 1999-12-28 2004-05-20 Intel Corporation, A California Corporation Thread signaling in multi-threaded network processor
US20030053984A1 (en) * 1999-12-30 2003-03-20 Jurg Tschopp Bimer or an oligomer of a dimer, trimer, quadromer or pentamer of recombinant fusion proteins
US20040039895A1 (en) * 2000-01-05 2004-02-26 Intel Corporation, A California Corporation Memory shared between processing threads
US20030108992A1 (en) * 2000-02-11 2003-06-12 Lenardo Michael J. Identification of a novel domain in the tumor necrosis factor receptor family that mediates pre-ligand receptor assembly and function
US20030023038A1 (en) * 2000-02-11 2003-01-30 Paul Rennert Heterologous polypeptide of the TNF family
US20020004587A1 (en) * 2000-04-11 2002-01-10 Genentech, Inc. Multivalent antibodies and uses therefor
US6623941B1 (en) * 2000-05-05 2003-09-23 Human Genome Sciences, Inc. Nucleic acids encoding human tumor necrosis factor TR20
US20030194743A1 (en) * 2000-08-18 2003-10-16 Beltzer James P. Binding polypeptides for B lymphocyte stimulator protein (BLyS)
US20040033511A1 (en) * 2000-09-15 2004-02-19 Klaus Pfizenmaier Site-specific antibody-mediated activation of proapoptotic cytokines: AMAIZEe (antibody-mediated apoptosis inducing cytokines)
US20040072188A1 (en) * 2000-09-18 2004-04-15 Ambrose Christine M. Novel receptor nucleic acids and polypeptides
US20040013674A1 (en) * 2001-04-27 2004-01-22 Christine Ambrose Taci as an anti-tumor agent
US20040038349A1 (en) * 2001-07-27 2004-02-26 Hilbert David M. Heteromultimeric TNF ligand family members
US20030100074A1 (en) * 2001-08-16 2003-05-29 Guo-Liang Yu Methods and compositions for treating metabolic bone diseases relating to human endokine alpha
US20030100068A1 (en) * 2001-10-12 2003-05-29 Jonathan Lam RANKL mimics and uses thereof
US20030148454A1 (en) * 2001-11-02 2003-08-07 Trustees Of Boston University Virus-free vesicles for delivery of functional membrane bound proteins
US20030119149A1 (en) * 2001-12-21 2003-06-26 Pranhitha Reddy Recombinant polypeptides
US20030211509A1 (en) * 2002-03-26 2003-11-13 Wiley Steven R. TNF-delta ligand and uses thereof
US20040028658A1 (en) * 2002-06-27 2004-02-12 Denise Faustman Methods of organ regeneration

Cited By (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8173122B2 (en) 1996-10-25 2012-05-08 Human Genome Sciences, Inc. Methods of treatment using antibodies to neutrokine-alpha
US8303951B2 (en) 1996-10-25 2012-11-06 Human Genome Sciences, Inc. Neutrokine-alpha antibodies and methods of use thereof
US8231873B2 (en) 1996-10-25 2012-07-31 Human Genome Sciences, Inc. Methods of treatment using antibodies to Neutrokine-alpha
US20060198784A1 (en) * 1996-10-25 2006-09-07 Human Genome Sciences, Inc. Methods of treatment using antibodies to Neutrokine-alpha
US20090068201A1 (en) * 1996-10-25 2009-03-12 Human Genome Sciences, Inc. Methods of treatment using antibodies to neutrokine-alpha
US20100196360A9 (en) * 1996-10-25 2010-08-05 Human Genome Sciences, Inc. Neutrokine-alpha and neutrokine-alpha splice variant
US20090104189A1 (en) * 1996-10-25 2009-04-23 Human Genome Sciences, Inc. Neutrokine-alpha and neutrokine-alpha splice variant
US8071092B1 (en) 1996-10-25 2011-12-06 Human Genome Sciences, Inc. Methods of inhibiting B lymphocytes using antibodies to Neutrokine-alpha
US20090169565A1 (en) * 1996-10-25 2009-07-02 Human Genome Sciences, Inc. Neutrokine-alpha antibodies and methods of use thereof
US20110135639A1 (en) * 1996-10-25 2011-06-09 Human Genome Sciences, Inc. B lymphocyte stimulator assays
US20110052590A1 (en) * 1996-10-25 2011-03-03 Human Genome Sciences, Inc. Neutrokine-alpha antibodies and methods of use thereof
US8212004B2 (en) 1999-03-02 2012-07-03 Human Genome Sciences, Inc. Neutrokine-alpha fusion proteins
US20050186637A1 (en) * 1999-03-02 2005-08-25 Guo-Liang Yu Neutrokine-alpha and neutrokine-alpha splice variant
US7879328B2 (en) 2000-06-16 2011-02-01 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to B lymphocyte stimulator
US8101181B2 (en) 2000-06-16 2012-01-24 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to B lymphocyte stimulator protein
US9187548B2 (en) 2000-06-16 2015-11-17 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to B lymphocyte stimulator protein
US20110117093A1 (en) * 2000-06-16 2011-05-19 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to b lymphocyte stimulator protein
US7605236B2 (en) 2000-06-16 2009-10-20 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to B lymphocyte stimulator protein
US20060062789A1 (en) * 2000-06-16 2006-03-23 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to B lymphocyte stimulator protein
US20100003259A1 (en) * 2000-06-16 2010-01-07 Human Genome Sciences, Inc. Antibodies that immunospecifically bind to b lymphocyte stimulator protein
US20060084608A1 (en) * 2000-08-18 2006-04-20 Human Genome Sciences, Inc. Binding polypeptides and methods based thereon
US20070293434A9 (en) * 2000-08-18 2007-12-20 Human Genome Sciences, Inc. Binding polypeptides and methods based thereon
US8062906B2 (en) 2000-08-18 2011-11-22 Human Genome Sciences, Inc. B-lymphocyte stimulator binding polypeptides and methods based thereon
US8105603B2 (en) * 2004-01-29 2012-01-31 Genentech, Inc. Polypeptides that bind APRIL
US20090297504A1 (en) * 2004-01-29 2009-12-03 Genentech, Inc. Bcma polypeptides and uses thereof
US20090081213A1 (en) * 2005-10-13 2009-03-26 Human Genome Sciences, Inc. Methods and compositions for use in treatment of patients with autoantibody positive disease
US20090148462A1 (en) * 2005-10-13 2009-06-11 Human Genome Sciences, Inc. Methods and compositions for use in treatment of patients with autoantibody positive disease
US9168286B2 (en) 2005-10-13 2015-10-27 Human Genome Sciences, Inc. Methods and compositions for use in treatment of patients with autoantibody positive disease
US8211649B2 (en) 2006-03-31 2012-07-03 Human Genome Sciences, Inc. Methods of diagnosing and prognosing hodgkin's lymphoma
WO2012047093A1 (en) * 2010-10-05 2012-04-12 Academisch Ziekenhuis Bij De Universiteit Van Amsterdam Treatment of the sjögren's syndrome

Also Published As

Publication number Publication date
US9545086B2 (en) 2017-01-17
US20090110676A1 (en) 2009-04-30
US20030095967A1 (en) 2003-05-22

Similar Documents

Publication Publication Date Title
US9545086B2 (en) BAFF, inhibitors thereof and their use in the modulation of B-cell response and treatment of autoimmune disorders
US6869605B2 (en) BAFF, inhibitors thereof and their use in the modulation of B-cell response
US20100233179A1 (en) BAFF, inhibitors thereof and their use in the modulation of B-cell response
Mackay et al. Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations
Gross et al. TACI-Ig neutralizes molecules critical for B cell development and autoimmune disease: impaired B cell maturation in mice lacking BLyS
Han et al. Overexpression of human decoy receptor 3 in mice results in a systemic lupus erythematosus–like syndrome
ES2369265T3 (en) A BAFF SOLUBLE SPECIFIC ANTIBODY FOR USE IN CANCER TREATMENT.

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION