EP2139509A2 - Traitement de troubles auto-immuns - Google Patents

Traitement de troubles auto-immuns

Info

Publication number
EP2139509A2
EP2139509A2 EP08742126A EP08742126A EP2139509A2 EP 2139509 A2 EP2139509 A2 EP 2139509A2 EP 08742126 A EP08742126 A EP 08742126A EP 08742126 A EP08742126 A EP 08742126A EP 2139509 A2 EP2139509 A2 EP 2139509A2
Authority
EP
European Patent Office
Prior art keywords
ltbr
seq
extracellular domain
variant
amino acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP08742126A
Other languages
German (de)
English (en)
Inventor
Graham K. Farrington
Evan Beckman
Jeffrey L. Browning
Werner Meier
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 Inc
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
Application filed by Biogen Idec Inc, Biogen Idec MA Inc filed Critical Biogen Idec Inc
Publication of EP2139509A2 publication Critical patent/EP2139509A2/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • 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
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • Protein heterogeneity can be caused by a range of post-translational modifications. Protein heterogeneity often results from different types of post- translational modifications, including carboxylation, hydroxylation, proteolytic processing, suflation, and glycosylation, the latter of which is the most common modification (Walsh and Jefferis (2006) Nat Biotech 24(10): 1241). Post-translational modifications can potentially affect product production levels (by influencing, for example, the degree of proper protein folding), stability, and a range of pharmacokinetic and pharmacodynamic parameters, as well as safety and immunogenicity. Post-translational modifications of therapeutic biologies, or protein- based biopharmaceuticals, may affect protein properties relevant to their therapeutic application.
  • N and/or C-terminal heterogeneity is an example of protein heterogeneity which must be considered in the manufacture of protein-based biopharmaceuticals.
  • N-terminal heterogeneity results from proteolytic processing at the amino terminal portion of the protein, where such processing may result in a population of proteins having different sizes. Variations in N-terminal proteolysis may occur in proteins comprising a signal sequence.
  • N-terminal glutamine residues can undergo spontaneous cyclization to form pyroglutamic acid. Thus, obtaining a homogenous population of proteins which can be used for therapeutic purposes often presents a challenge.
  • Lymphotoxin beta receptor is a member of the tumor necrosis factor receptor (TNFR) family. The receptor is expressed on the surface of cells in the parenchyma and stroma of most lymphoid organs but is absent on T- and B- lymphocytes. Signaling through LTBR by the LT ⁇ / ⁇ heterotrimer (LT) is important during lymphoid development.
  • LTBR is also known to bind the ligand LIGHT (homologous to lymphotoxins, exhibits inducible expression, and competes with herpes simplex virus glycoprotein D for HVEM, a receptor expressed by T lymphocytes), which has been implicated in T-cell driven events, both in the periphery and in the thymus. LT and LIGHT are expressed on the surface of activated lymphocytes. Blocking the LT pathway with a soluble decoy LTBR has been shown to be effective to treat autoimmune disease in various animal models.
  • the invention pertains to a composition
  • a composition comprising a population of lymphotoxin- ⁇ receptor (LT- ⁇ -R)-Ig- fusion proteins which comprise a variant LT- ⁇ -R extracellular domain of 193 or 194 amino acids in length and a variant Ig portion of 227 amino acids in length, wherein at least 90% of the LT- ⁇ -R-Ig-fusion proteins are missing no more than 5 amino acids from the N-terminus of the mature form of the wild type LT- ⁇ -R extracellular domain set forth in SEQ ID NO:21 and wherein the LT- ⁇ -R-Ig-fusion proteins lack N-terminal pyroglutamic acid.
  • the N-terminal amino acid of the variant LT- ⁇ -R-Ig fusion protein is a non-polar amino acid.
  • the non polar amino acid is either a valine (amino acid six of the mature form of the wild type LT- ⁇ -R extracellular domain of SEQ ID NO:21) or an alanine (amino acid five of the mature form of the wild type LT- ⁇ -R extracellular domain of SEQ ID NO:21).
  • the N-terminal amino acid of at least 95% of the LT- ⁇ -R- Ig-fusion proteins is either a valine (amino acid six of the mature form of the wild type LT- ⁇ -R extracellular domain of SEQ ID NO:21) or an alanine (amino acid five of the mature form of the wild type LT- ⁇ -R extracellular domain of SEQ ID NO:21).
  • the composition is made by expressing a nucleic acid molecule comprising a nucleotide sequence encoding the extracellular domain of LTBR set forth in SEQ ID NO:4 in a mammalian cell.
  • nucleic acid molecule comprises the sequence set forth in SEQ ID NO:3.
  • variant Ig portion comprises Fc regions of an IgGl isotype.
  • the variant Ig portion comprises the amino acid sequence set forth in SEQ ID NO:2. [0014] In one embodiment, the Ig portion is non-glycosylated.
  • the composition is made by expressing a nucleic acid molecule encoding the LT- ⁇ -R- Ig fusion protein set forth in SEQ ID NO:5 in a mammalian cell.
  • the nucleic acid molecule comprises the sequence set forth in SEQ ID NO:7.
  • the step of expressing is done at manufacturing scale.
  • the invention pertain to a composition
  • a composition comprising a population of lymphotoxin- ⁇ receptor-immunoglobulin (LT- ⁇ -R-Ig)-fusion proteins comprising a variant LT- ⁇ -R extracellular domain and a variant Ig portion, wherein the variant LT- ⁇ -R extracellular domain is aglycosylated.
  • LT- ⁇ -R-Ig lymphotoxin- ⁇ receptor-immunoglobulin
  • the aglycosylated extracellular domain of LTBR comprises amino acids 1 to 194 of SEQ ID NO: 10.
  • the invention pertains to a composition
  • a composition comprising a population of lymphotoxin- ⁇ receptor-immunoglobulin (LT- ⁇ -R-Ig)-fusion proteins, the fusion proteins comprising a variant LT- ⁇ -R extracellular domain of 193 or 194 amino acids in length and a variant Ig portion, wherein the population has reduced N- terminal pyroglutamic acid formation, and reduced C-terminal heterogeneity compared to wild-type LT- ⁇ -R-Ig fusion proteins.
  • LT- ⁇ -R-Ig lymphotoxin- ⁇ receptor-immunoglobulin
  • At least 90% of the LT- ⁇ -R-Ig- fusion proteins comprise a variant LT- ⁇ -R extracellular domain as set forth the amino acid sequence of SEQ ID NO: 1
  • the variant Ig portion comprises a mutation in the hinge region.
  • the invention pertains to a pharmaceutical composition
  • a pharmaceutical composition comprising a population of lymphotoxin- ⁇ receptor (LT- ⁇ -R)-Ig-fusion proteins which comprise a variant LT- ⁇ -R extracellular domain of 193 or 194 amino acids in length and a variant Ig portion of 227 amino acids in length, wherein at least 90% of the LT- ⁇ -R-Ig-fusion proteins are missing no more than 5 amino acids from the N- terminus of the mature form of the wild type LT- ⁇ -R extracellular domain set forth in SEQ ID NO:21and wherein the LT- ⁇ -R-Ig-fusion proteins lack N-terminal pyroglutamic acid and a pharmaceutically acceptable carrier.
  • the N-terminal amino acid of the variant LT- ⁇ -R-Ig fusion protein is a non-polar amino acid.
  • the non polar amino acid is either a valine (amino acid six of the mature form of the wild type LT- ⁇ -R extracellular domain of SEQ ID NO.21) or an alanine (amino acid five of the mature form of the wild type LT- ⁇ -R extracellular domain of SEQ ID NO:21).
  • the N-terminal amino acid of at least 95% of the LT- ⁇ -R- Ig- fusion proteins is either a valine (amino acid six of the mature form of the wild type LT- ⁇ -R extracellular domain of SEQ ID NO:21) or an alanine (amino acid five of the mature form of the wild type LT- ⁇ -R extracellular domain of SEQ ID NO:21).
  • the composition is made by expressing a nucleic acid molecule comprising a nucleotide sequence encoding the extracellular domain of LTBR set forth in SEQ ID NO:4 in a mammalian cell.
  • the invention pertains to a method of treating an autoimmune disorder comprising administering the pharmaceutical composition of claim 23 to a subject in need thereof.
  • the autoimmune disorder is selected from the group consisting of rheumatoid arthritis, Crohn's disease, or systemic lupus erythematosus
  • the invention pertains to a pharmaceutical composition
  • a pharmaceutical composition comprising a population of lymphotoxin- ⁇ receptor-immunoglobulin (LT- ⁇ -R-Ig)- fusion proteins, the fusion proteins comprising a variant LT- ⁇ -R extracellular domain of 193 or 194 amino acids in length and a variant Ig portion, wherein the population has reduced N-terminal pyroglutamic acid formation and reduced C-terminal heterogeneity compared to wild-type LT- ⁇ -R-Ig fusion proteins and a pharmaceutically acceptable carrier.
  • LT- ⁇ -R-Ig lymphotoxin- ⁇ receptor-immunoglobulin
  • the LT- ⁇ -R-Ig-fusion proteins comprise a variant LT- ⁇ -R extracellular domain as set forth the amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 23.
  • the variant Ig portion comprises a mutation in the hinge region.
  • the invention pertains to a method of treating an autoimmune disorder comprising administering the pharmaceutical composition of claim 29 to a subject in need thereof.
  • the autoimmune disorder is selected from the group consisting of rheumatoid arthritis, Crohn's disease, or systemic lupus erythematosus
  • the autoimmune disorder is rheumatoid arthritis.
  • the pharmaceutical composition is administered to the subject at a dose of from about 0.6 to 3 mg/kg biweekly.
  • the pharmaceutical composition is administered subcutaneously.
  • the invention pertains to an isolated polypeptide comprising a variant LT- ⁇ -R extracellular domain of 193 or 194 amino acids in length and a variant
  • polypeptide is missing no more than 5 amino acids from the N-terminus of the mature form of the wild type LT- ⁇ -R extracellular domain set forth in SEQ ID NO:21 and wherein the polypeptide lacks N- terminal pyroglutamic acid.
  • the N-terminal amino acid is a non-polar amino acid.
  • the non polar amino acid is either a valine (amino acid six of the mature form of the wild type LT- ⁇ -R extracellular domain of SEQ ID NO:21) or an alanine (amino acid five of the mature form of the wild type LT- ⁇ -R extracellular domain of SEQ ID NO:21 ).
  • the polypeptide is made by expressing a nucleic acid molecule comprising a nucleotide sequence encoding the extracellular domain of
  • LTBR set forth in SEQ ID NO:4 in a mammalian cell.
  • the invention pertains to an isolated nucleic acid molecule encoding the polypeptide of the invention.
  • the nucleic acid molecule comprises of the nucleotide sequence set forth in SEQ ID NO:7.
  • the inention pertains to a vector comprising the nucleic acid molecule of the invention
  • the invention pertains to a host cell expressing the vector of the invention.
  • the cell is a Chinese Hamster Ovary (CHO) cell.
  • the invention pertains to a process for making a composition comprising a population of lymphotoxin- ⁇ receptor (LT- ⁇ -R)-Ig- fusion proteins which comprise a variant LT- ⁇ -R extracellular domain and a variant Ig portion, wherein at least 90% of the LT- ⁇ -R-Ig-fusion proteins are missing no more than 5 amino acids from the N-terminus of the mature form of the wild type LT- ⁇ -R extracellular domain set forth in SEQ ID NO:21, the process comprising, expressing a nucleic acid molecule encoding the LT- ⁇ -R- Ig fusion protein set forth in SEQ ID NO: 8 in a mammalian cell, obtaining the population from the culture supernatant, and, optionally, purifying the supernatant
  • the nucleic acid molecule consists of the nucleotide sequence set forth in SEQ ID NO:7.
  • the invention pertains to a method of treating rheumatoid arthritis in a human subject, the method comprising administering to the subject a dose of LT- ⁇ -R-Ig fusion protein, wherein the dose is sufficient to maintain an average concentration of from about 0.14 ug/ml to about 3.5 ug/ml in the serum of the subject.
  • the invention pertains to a method of treating rheumatoid arthritis in a human subject, the method comprising administering to the subject a dose of LT- ⁇ -R-Ig fusion protein, wherein the dose is sufficient to maintain an average a minimal average concentration of about 0.6 ug/ml in the serum of the subject.
  • the LTBR-Ig fusion protein comprises the amino acid sequence set forth in SEQ ID NO: 5.
  • the concentration is achieved by administering LT- ⁇ -R-Ig fusion protein at a dose of from about 0.01 to about 5 mg/kg once every 7-60 days.
  • the invention pertains to a method of treating rheumatoid arthritis in a human subject, the method comprising administering to the subject a dose of LT- ⁇ -R-Ig fusion protein of from about 0.6 to 3 mg/kg not more than twice every 7-30 days.
  • method comprising administering to the subject a dose of LT- ⁇ -R-Ig fusion protein of from about 0.6 to 3 mg/kg once every 7-14 days.
  • administration is once every 14-30 days.
  • administration is once every 28-60 days.
  • administration is once every 7-30 days.
  • the invention pertains to a method of treating an autoimmune disorder in a human subject, the method comprising administering to the subject a dose of a pharmaceutical composition comprising a population of LT- ⁇ -R-Ig fusion proteins comprising a variant LT- ⁇ -R extracellular domain of 193 or 194 amino acids in length, wherein at least 90% of the LT- ⁇ -R-Ig-fusion proteins are missing no more than 5 amino acids from the N-terminus of the mature form of the wild type LT- ⁇ -R extracellular domain set forth in SEQ ID NO:21.and wherein the dose is sufficient to maintain a minimal average concentration of about 0.6 ug/ml in the serum of the subject.
  • the LT- ⁇ -R-Ig fusion protein further comprises a variant
  • the autoimmune disorder is selected from the group consisting of rheumatoid arthritis, Crohn's disease, or systemic lupus erythematosus
  • the pharmaceutical composition comprises the amino acid sequence set forth in SEQ ID NO: 5. [0064] In one embodiment, administration is twice monthly.
  • administration once monthly.
  • administration is subcutaneous.
  • the dose is about 1 mg/kg. [0068] In one embodiment, the dose is about 3 mg/kg.
  • the dose is about 1 mg/kg administered about every 7 to
  • the dose is about 3 mg/kg administered about every 14 to 30 days.
  • the dose is about 1 mg/kg administered about every 14 days.
  • the autoimmune disorder is rheumatoid arthritis and the subject has been treated with a rheumatoid arthritis drug after being diagnosed with rheumatoid arthritis and prior to administration of the LT- ⁇ -R-Ig fusion protein.
  • the rheumatoid arthritis drug is chosen from the group consisting of a DMARD, an NSAID, and a corticosteroid.
  • the human is a DMARD-inadequate responder.
  • the rheumatoid arthritis drug is a TNF inhibitor.
  • the rheumatoid arthritis drug is adalimumab (Humira®), etanercept (Enbrel®), or infliximab (Remicade®).
  • LT- ⁇ -R-Ig is administered in combination with the rheumatoid arthritis drug.
  • the human is evaluated to determine if the response to the rheumatoid arthritis drug is inadequate prior to administration of LT- ⁇ -R-Ig.
  • the human is determined to have an inadequate response to the rheumatoid arthritis drug, and then the human is administered LT- ⁇ -R-Ig.
  • the human is asymptomatic for a first manifestation of rheumatoid arthritis and is symptomatic for a second manifestation of rheumatoid arthritis.
  • LT- ⁇ -R-Ig is administered in place of the rheumatoid arthritis drug.
  • administration is in combination with a tumor necrosis factor (TNF) inhibitor.
  • TNF tumor necrosis factor
  • the TNF inhibitor is adalimumab (Humira®), etanercept
  • the human is an anti-TNF-inadequate responder.
  • administration is in combination with a non-steroidal antiinflammatory agent (NSAID), a corticosteroid, or a disease modifying antirheumatic drug (DMARD).
  • NSAID non-steroidal antiinflammatory agent
  • corticosteroid corticosteroid
  • DMARD disease modifying antirheumatic drug
  • administration is in combination with methotrexate.
  • the human is a DMARD-inadequate responder.
  • Figure 1 describes the changes made from LTBROl to LTBR06 to improve heterogeneity.
  • the bold letters indicate the secretion sequence, and the italicized / underlines letters indicate the amino acids which are removed in LTBR06 relative to LTBROl, i.e., amino acids 1-4 and the last amino acid (lysine).
  • Three consensus sites for N-linked glycosylation are located at Asnl3, 150 and 276.
  • Amino acid positions refer to full length LTBR, i.e., amino acids 1-4 are those which are removed in
  • Figure 2 provides an alignment of LTBROl, LTBR05, LTBR06, and LTBR09. The secretion sequence is omitted from the LTBR sequences.
  • Figure 3 describes LTBR06 in comparison to wild type and provides a schematic of the protein.
  • Figure 4 describes the amino acid sequence of the LTB R06 construct (mature form of LTBR06 shown in SEQ ID NO: 5).
  • LTBR06 is a disulfide-linked, glycosylated, dimeric protein. There are 28 cysteine residues and 6 glycosylation sites, the latter of which is indicated in bold.
  • Figure 5 describes aglycosylated hLT ⁇ R hlgGl (mature form of protein).
  • the huLT ⁇ R is residues 1-204, the hulgGl Fc is residues 205-431 above (the Fc's glycosylation site is intact).
  • Figure 6 describes hinges of which may be used in the LTBR IgG fusion proteins of the invention.
  • Figure 7 describes a table which describes a summary of results from a hinge expression analysis.
  • Figure 8 is a graph depicting the percent improvement in RA symptom scores
  • FIG. 9 provides a graph which depicts a decrease in Tender Joint Counts (TJC) in patients following treatment with LTBR-Fc (LTBR06).
  • Figure 10 provides a graph which depicts a decrease in Swollen Joint Counts n (SJC) in patients following treatment with LTBR-Fc (LTBR06).
  • Figure 11 provides a graph which depicts the median % change in Swollen Joint Counts n (SJC) in patients following treatment with LTBR-Fc (LTBR06).
  • Figure 12 provides a graph which depicts ACR20 improvements in patients following treatment with LTBR-Fc (LTBR06).
  • Figure 13 graphically depicts serum levels of differentially sialylated LFA3TIP and hu-LTBR-Ig (LTBR05) in mice 24 hours post administration with 100 ug/animal.
  • Figure 14 illustrates a comparison of LTBR-Fc and a typical antibody and shows that at identical dosing significant differences in efficacy are observed for prolonged time frames.
  • Arrows A and B indicate the typical alpha and beta phases for an antibody or Fc-fusion protein, respectively.
  • the gray line indicated by arrow C shows typical lower limit concentration for efficacy
  • arrow D shows LTBR-Fc has efficacy at significantly lower concentrations.
  • fusion protein refers to a molecule comprising two or more proteins or fragments thereof linked by a covalent bond via their individual peptide backbones, most preferably generated through genetic expression of a polynucleotide molecule encoding those proteins.
  • the fusion protein includes an immunoglobulin domain.
  • immunoglobulin comprises five distinct classes of antibody that can be distinguished biochemically. All five classes of antibodies are clearly within the scope of the present invention, the following discussion will generally be directed to the IgG class of immunoglobulin molecules.
  • immunoglobulin fusion protein refers to a fusion of a functional portion of a polypeptide (generally comprising the extracellular domain of a cell surface protein) with one or more portions of an immunoglobulin constant region, e.g. the hinge, CHl, CH2 or CH3 domains or portions or combinations thereof.
  • the polypeptide is a member of the TNF family of receptors.
  • the portions of the Ig molecule may derive from any of the various immunoglobulin isotypes, including, for example, IgGl, IgG2, IgM, IgA etc.
  • Immunoglobulin fusion proteins are referred to herein as Ig for Fc fusion proteins.
  • the protein used in the methods and compositions of the invention is an immunoglobulin fusion protein.
  • the fusion protein may comprise a receptor, or ligand binding portion thereof, and a dimerization domain, e.g., an Fc domain.
  • variant LTBR extracellular domain refers the mature form of a polypeptide (or protein) having an amino acid sequence that differs from the sequence presented in SEQ ID NO: 1 and SEQ ID NO: 21 (wild type hLTBR) at one or more amino acid positions.
  • variant Ig portion refers to a polypeptide (or protein) having an amino acid sequence that differs from Fc regions known in the art, including the sequence provided herein as SEQ ID NO: 22, at one or more amino acid positions.
  • the term "reduced N-terminal heterogeneity" refers to a decrease in the number of proteins in a population having different N-terminal amino acid residues or that occur in different forms relative to a control protein population.
  • expression of a control protein may result in a protein population comprising proteins with missing N-terminal amino acid residues ranging from one amino acid to three amino acids (relative to the predicted translated protein), also described as N-I, N-2, and N-3.
  • the population of proteins would include three different types, i.e., N-I, N-2, and N-3.
  • a population of proteins having reduced N-terminal heterogeneity would include a population of proteins having less than three different N-terminal amino acid residues, e.g., N-I and N-2, N- 2 and N-3, or N-I and N-3 types of proteins or reduced percentage of one or more of these types of variant molecules.
  • N-termial amino acids themselves may not be different, e.g., for a protein having an N-terminus of N-I relative to the wild-type form, the N-terminal amino acid may be an alanine and for a protein having an N-terminus of N-3 relative to the wild-type form, the N-terminal amino acid may be also an alanine.
  • N-terminal heterogeneity has to do with the overall difference in lengths among the proteins within a population (expressed in relative terms to the predicted translated protein).
  • N-# refers to the number of amino acids deleted from the amino terminus of the wild-type translated protein. For example, if a wild-type predicted protein sequence was AAGTY, an N-I protein would not comprise the initial A and would instead begin with AGTY, although both proteins begin with A. Similarly, an N-2 protein would begin with GTY.
  • N-terminal heterogeneity can be reduced by reducing the potential number of variant forms in a population of proteins.
  • N-terminal glutamine residues can undergo spontaneous cyclization to form pyroglutamic acid which can lead to further heterogeneity.
  • the formation of pyroglutamic acid is reduced or eliminated compared to that present in wild-type proteins.
  • the term “reduced C terminal heterogeneity” likewise refers to a decrease in the number of proteins in a population having different C-terminal amino acid residues relative to a control protein population.
  • C-# refers to the number of amino acids deleted from the carboxy terminus of a protein.
  • glycosylation refers to the covalent linking of one or more carbohydrates to a polypeptide. Typically, glycosylation is a posttranslational event which can occur within the intracellular milieu of a cell or extract therefrom.
  • glycosylation includes, for example, N-linked glycosylation (where one or more sugars are linked to an asparagine residue) and/or O-linked glycosylation (where one or more sugars are linked to an amino acid residue having a hydroxyl group (e.g., serine or threonine).
  • N-linked glycosylation where one or more sugars are linked to an asparagine residue
  • O-linked glycosylation where one or more sugars are linked to an amino acid residue having a hydroxyl group (e.g., serine or threonine).
  • TNF family of receptors refers to any receptor, whether naturally membrane bound or secreted (as in the case of osteoprotegerin), which has the canonical TNF family cysteine bridging patterns or any receptor which binds to a defined member of the TNF family of ligands (e.g. Banner et al 1993).
  • the claimed invention in other embodiments relates to TNF family receptor-Ig fusions obtained by the methods discussed herein, as well as to pharmaceutical preparations comprising them.
  • a "signal peptide” or “signal sequence” is a peptide sequence that directs a newly synthesized polypeptide to which the signal peptide is attached to the endoplasmic reticulum (ER) for further post-translational processing and distribution.
  • the mature form of a protein refers to the protein without the signal sequence.
  • a "soluble LTBR” is a polypeptide that includes all or a portion of the extracellular domain of human LTBR (e.g., an LTBR immunoglobulin fusion).
  • Preferred soluble LTBRs are soluble molecules which include sufficient sequence from the extracellular region of LTBR that they can bind to a ligand, e.g., LT or LIGHT, with at least 10% and preferably at least 50% of the affinity of the molecule of SEQ ID NO: 1.
  • a ligand e.g., LT or LIGHT
  • ligand binding domain or "ligand binding portion” as used herein refers to any native receptor (e.g., cell surface receptor) or any region or derivative thereof retaining at least a qualitative ligand binding ability, and preferably the biological activity of a corresponding native receptor.
  • conservative amino acids substitutions denotes the replacement of an amino acid residue by another, biologically similar residue. For example, one would expect conservative amino acid substitutions to have little or no effect on the biological activity, particularly if they represent less than 10% of the total number of residues in the polypeptide or protein.
  • conservative amino acids substitutions represent changes in less than 5% of the polypeptide or protein, most preferably less than 2% of the polypeptide or protein (e.g., when calculated in accordance with SEQ ID NO 5, most preferred conservative substitutions would represent fewer than 9 amino acid substitutions in the wild type amino acid sequence).
  • substitutions of particularly conservative substitutions include the substitution of one hydrophobic residue such as isoleucine, valine, leucine or methionine for one another, or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamic for aspartic acid, or glutamine for asparagine, and the like.
  • Genetically encoded amino acids generally may be divided into four families: (1) acidic: aspartate, glutamate; (2) basic: lysine, arginine, histidine; (3) nonpolar: alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan; and (4) uncharged polar: glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine. Phenylalanine, tryptophan, and tyrosine may be classified jointly as aromatic amino acids.
  • the amino acid at the N-terminal of the polypeptide of the invention is a nonpolar, amino acid (i.e., alanine, valine, leucine, isoleucine, phenylalanine, metionine, tryptophan, glycine, and cysteine), excluding imino acids, i.e., proline.
  • a nonpolar, amino acid i.e., alanine, valine, leucine, isoleucine, phenylalanine, metionine, tryptophan, glycine, and cysteine
  • imino acids i.e., proline.
  • treating refers to administering a therapy in an amount, manner, and/or mode effective to improve or prevent a condition, symptom, or parameter associated with a disorder or to prevent onset, progression, or exacerbation of the disorder (including secondary damage caused by the disorder), to either a statistically significant degree or to a degree detectable to one skilled in the art. Accordingly, treating can achieve therapeutic and/or prophylactic benefits.
  • An effective amount, manner, or mode can vary depending on the subject and may be tailored to the subject.
  • inadequate response refers to a patient who, as assessed by the patient or a clinician of ordinary skill, exhibits insufficient efficacy or intolerable or unacceptable toxicity to a particular treatment. Insufficient efficacy can mean a failure to meet a predetermined level of response to treatment.
  • insufficient efficacy may be defined as failure to exhibit at least a 10%, 20%, 25%, 30%, 40%, 50% or more decrease in a clinical parameter of RA, such as tender joint count (TJC), swollen joint count (SJC), patient global assessment of disease activity [PGA visual analog scale (VAS) 0-10 cm], physician global assessment of disease activity (MDGA VAS 0-10 cm) and C-reactive protein (CRP in mg/dl).
  • TJC tender joint count
  • SJC swollen joint count
  • C-reactive protein C-reactive protein
  • Intolerable toxicity can be an adverse reaction to an agent that results in medical need or recommendation to discontinue use of the first agent.
  • intolerable or unacceptable toxicity may include hepatic injury or dysfunction, severe allergic reaction, severe depression or suicidal ideation, anaphylaxis, or injection site reaction.
  • administered in combination means that two or more agents (e.g., the soluble LTBR and the second agent) are administered to a subject at the same time or within an interval, such that there is overlap of an effect of each agent on the patient.
  • the administrations of the first and second agent are spaced sufficiently close together such that a combinatorial effect, e.g., an additive or synergistic effect, is achieved.
  • the interval can be an interval of hours, days or weeks.
  • the agents can be concurrently bioavailable, e.g., detectable, in the subject.
  • at least one administration of one of the agents, e.g., the first agent is made while the other agent, e.g., the soluble LTBR, is still present at a therapeutic level in the subject.
  • week means not more than once within a particular 6 to 8 day period, e.g., once every 7 days.
  • the term "biweekly” means not more than once within a particular 12 to 16 day period, e.g., once every 14 days.
  • the term “monthly” means once a month, e.g, once every 28 to 31 days.
  • the subject is typically a mammal, e.g., human, non- human primate (such as a monkey or ape), dog, cat, rabbit, or agriculture mammal (e.g., horse, cow, pig, and so on).
  • the subject is a human, e.g., a human male or female.
  • the subject can be at least about 18, 25, 30, 45, 50, 55, 60, or 70 years old.
  • minimal average concentration refers to the mean minimal concentration of drug present in the circulation or in the serum of a subject.
  • the invention pertains to compositions comprising LBTR-Ig fusion proteins that are improved for therapeutic use in that the population of LTBR-Ig fusions proteins has reduced molecular heterogeneity.
  • the invention provides compositions, including pharmaceutical compositions, comprising LTBR-Ig fusion proteins, as well as proteins, nucleic acids, vectors, host cells, and methods of making the same.
  • lymphotoxin- ⁇ receptor-immunoglobulin (LT ⁇ R-Ig) fusion protein can block signaling between the surface LT ligand and the receptor with consequences on the functional state of follicular dendritic cells (Mackay and
  • soluble LTBRs of the invention are fusion proteins.
  • a soluble LTBR, as defined herein, is a molecule that includes an LT- binding fragment of the extracellular domain of LTBR.
  • a soluble LTBR can include all or a substantial portion of the extracellular domain of LTBR (e.g., it can include residues 40-200, 35-200, 40-210; 35-220, 32-225, or 28-225 of SEQ ID NO:1).
  • a soluble LTBR includes residues 32-225 of SEQ ID NO: 1.
  • a soluble LTBR can be modified by covalent attachment of a moiety, e.g., a heterologous polypeptide (e.g., to make an LTBR fusion protein) or a non-polypeptide moiety.
  • LTBR fusion proteins can include all or part of the constant region of an antibody (e.g., an Fc domain), transferrin, or albumin, such as human serum albumin (HSA) or bovine serum albumin (BSA).
  • HSA human serum albumin
  • BSA bovine serum albumin
  • the fusion protein can include a linker region between the LTBR sequence and the non-LTBR protein domain.
  • a soluble LTBR is modified by covalent attachment to a polymer such as a polyethylene glycol (PEG). While not wishing to be bound by theory or mechanism, such soluble LTBRs can act as decoy receptors to reduce (block) LTBR activity.
  • PEG polyethylene glycol
  • An exemplary LTBR-Fc has the amino acid sequence of SEQ ID NO:5, 6, 8, 9, or 10.
  • the soluble LTBR can include all or a fragment of LTBR, e.g., a soluble fragment of LTBR, fused to one or more heterologous protein domains (which domain(s) may increase solubility or lifetime in the blood).
  • An exemplary LTBR moiety is the LTBR sequence of SEQ ID NO: 1 , or a sequence which differs therefrom by no more than 1, 2, 3, 5, or 10 amino acid residues. The differences can be any difference, e.g., a substitution, deletion or insertion, but is preferably a substitution, e.g., a conservative substitution.
  • non-LTBR proteins or domains include all or part of the constant region of an antibody, e.g., an Fc domain, transferrin, or albumin, such as human serum albumin (HSA) or bovine serum albumin (BSA).
  • HSA human serum albumin
  • BSA bovine serum albumin
  • the polypeptide of the invention is an Fc fusion protein containing a polypeptide such as an antibody, and preferably an IgG immunoglobulin, e.g., of the subtype IgGl, IgG2, IgG3, or IgG4, and preferably, of the subtype IgGl or IgG4.
  • the foregoing polypeptide binds to a ligand of LTBR.
  • Amino acid numberings herein for portions of an Fc region of a polypeptide correspond to the Kabat numbering system as described, e.g., by Kabat et al., in "Sequences of Proteins of Immunological Interest", U.S. Dept.
  • a fusion protein of the invention comprises at least a portion of a hinge region, a CHl, a CH2, and a CH3 region of an immunoglobulin.
  • Heterogeneity within a protein population may result from post- translational modifications such as variable glycosylation patterns, N-terminal proteolysis, C-terminal proteolysis, and pyroglutamate formation (also referred to herein as pyroglu formation).
  • the invention provides a composition comprising a population of lymphotoxin- ⁇ receptor-immunoglobulin (LT- ⁇ -R-Ig)-fusion proteins having reduced heterogeneity, including, but not limited to, reduced N-terminal heterogeneity (e.g., with respect to variations in size of molecules or variantion in the form of molecules (e.g., pyroglutamic acid containing proteins) or reduced C-terminal heterogeneity, as well as combinations thereof.
  • LT- ⁇ -R-Ig lymphotoxin- ⁇ receptor-immunoglobulin
  • the reductions in heterogeneity may be attributed to deletions and/or mutations made within the sequence of the LTBR-Ig protein, such that heterogeneity is reduced relative to the wild-type LTBR-Ig fusion protein, i.e., unmutated and/or undeleted LTBR-Ig.
  • An example of a wild-type LTBR-Ig fusion protein i.e., unmutated and/or undeleted LTBR-Ig.
  • LTBR-Ig fusion protein is described in SEQ ID NO: 11 (mature form of protein), also referred to as LTBROl . It should be noted that the terms LT ⁇ R-Ig and LTBR-Fc are used interchangeably herein.
  • an LT ⁇ R-Ig fusion protein comprises a variant LTBR extracellular domain and/or a variant Ig portion, e.g., Fc portion of an Ig.
  • the LTBR-Ig fusion protein comprises either a LTBR extracellular domain variant, a variant Ig portion, or a combination thereof.
  • the amino acid and nucleic acid sequences of wild type LTBR are described in the NCBI database as AAH26262 and P36941.
  • the wild type human amino acid sequence of LTBR is also describe as SEQ ID NO: 1.
  • a soluble LTBR can be an LTBR-Fc polypeptide having the sequence of SEQ ID NO:1, or preferably a variant thereof.
  • the soluble LTBR is an LTBR-Fc polypeptide which differs from the sequence of SEQ ID NO:1 by no more than 1, 2, 3, 5, or 10 amino acid residues.
  • Human LTBR sequence (GenPept ID No. P36941) SEQ ID NO: 1 is the immature or nonprocessed human LTBR sequence, i.e., contains the signal sequence. Amino acids in italics indicate signal sequence. Amino acids 28-225 are the extracellular region of LTBR.
  • wild type LTBR-Ig refers to a fusion protein comprising the extracellular domain of human wild type LTBR, e.g., the extracellular domain of the LTBR sequence presented in SEQ ID NO: 1, and any immunoglobulin sequence known in the art which is not modified, for example, by mutations, deletions, etc.
  • An exemplary wild type Ig amino acid sequence is provided in SEQ ID NO: 22.
  • An example of a wild type LTBR-Ig fusion protein having no modifications is described in SEQ ID NO: 6.
  • the invention pertains to LTBR-Ig fusion proteins comprising a variant LTBR extracellular domain.
  • amino acids "SQPQ” may be deleted from the amino terminal of the wild-type LTBR protein (mature form), as shown in SEQ ID NO: 4 (amino acid sequence of the LTBR extracellular domain of LTB R06). As demonstrated in the Examples provided below, deletion of "SQPQ" from the amino terminal of LTBR improves the overall heterogeneity within an LTBR-Ig protein population, including N-terminal heterogeneity.
  • LTB R06 provides a population of LTBR-Ig fusion proteins where at least 90% of the LT- ⁇ -R-Ig- fusion proteins are missing no more than 5 amino acids from the N-terminus of the mature form of the wild type LT- ⁇ -R extracellular domain set forth in SEQ ID NO:21.
  • expression of LTBR06 results in a population of LTBR-Ig proteins, where 90% of the proteins are either N-4 or N-5.
  • LTBR06 has reduced pyroglutamic acid formation.
  • the LTBR-Ig of the invention may include a variant Ig portion having a deletion in the c-terminal amino acid of the Fc portion, i.e., "K".
  • the deletion of the last amino acid of an Fc portion of an LTBR-Ig fusion protein reduces C-terminal heterogeneity.
  • An example of a variant Ig portion where the last amino acid has been deleted to improve, i.e., reduce, C-terminal heterogeneity is described in SEQ ID NO: 2.
  • Examples of LTBR- Ig fusion proteins having variant Ig portions with a deleted last amino acid, i.e., the last lysine are described in SEQ ID NOs: 5, 8, 9, and 12.
  • the variant Ig portion may comprise an Fc region of an IgG isotype, including, but not limited to, an IgGl isotype.
  • the invention also includes an LTBR-Ig fusion protein having both a variant LTBR extracellular domain and a variant Ig portion.
  • LTBR06 is an LTBR-Ig fusion protein comprising a variant LTBR extracellular domain having the first four amino acids removed and a variant Ig portion, wherein the last amino acid (K) is deleted.
  • K last amino acid
  • the immunoglobulin portion of an Ig fusion protein of the invention comprises at least a portion of an immunoglobulin hinge region.
  • a variant Fc portion may comprise at least one mutation in the hinge region, e.g., a mutation of the cysteine (at amino acid position 220 (Kabat numbering) also shown as amino acid position one of SEQ ID NO:22) of the Ig upper hinge to a valine.
  • the subject valine is bolded/underlined/italicized below.
  • the underlined sequence is a substantial part of the extracellular domain of LTBR and corresponds to amino acids 32 to 225 of SEQ ID NO:1 (above).
  • the signal sequence of the LTBR protein is cleaved.
  • the final LTBR protein product for LTBR06 is described in SEQ ID NO: 5.
  • an immunoglobulin hinge be used to link an LTBR extracellular domain to, e.g., the CHl, CH2, and CH3 domains of an immunoglobulin molecule.
  • an IgG hinge region having the sequence CDKTHTCPPCPAPELLGGP may be used.
  • a hinge region having the sequence VDKTHTCPPCP APELLGGP may be used.
  • Other exemplary upper and middle hinge constructs are shown in Figures 6 to 8, as well as SEQ ID NOs: 13 to 20.
  • the variant Ig portion of the LTBR-Ig fusion protein may include an upper and middle hinge region comprising at least 90% to 95% identity to the hinges described in SEQ ID NOs: 13 to 20.
  • the upper and middle hinge region set forth in SEQ ID NO: 13 was used in the variant LTBR-Ig fusion proteins exemplified herein.
  • LTBR-Ig fusion proteins comprising variant hinges, e.g., those described in SEQ ID NOs: 14 to 21, would be constructed by replacing the sequence set forth in SEQ ID NO: 13 with the variant hinge.
  • variant hinge molecules that can optionally be used to link an LTBR extracellular domain to, e.g., the CHl, CH2, and CH3 domains of an immunoglobulin molecule are disclosed in 20050163782A1, the contents of which are incorporated by reference herein.
  • an LTBR-Ig fusion protein such as LTBR06
  • the population of proteins which results includes a variety of overall lengths due, at least in part, to N-terminal and/or C-terminal proteolysis, as well as heterogeneity of pyroglu formation.
  • An important aspect of the invention is the discovery that by deleting the first four amino acids of the LTBR-Ig fusion protein (specifically the first four amino acids of the LTBR extracellular domain), heterogeneity can be reduced, including N-terminal heterogeneity.
  • the invention features a composition comprising a population of LT ⁇ R-Ig-fusion proteins which comprise a variant LT ⁇ -R extracellular domain of 193 or 194 amino acids in length and a variant Ig portion of 227 amino acids in length, wherein at least 90% of the LT- ⁇ -R-Ig-fusion proteins are missing no more than 5 amino acids from the N-terminus of the mature form of the wild type LT- ⁇ -R extracellular domain set forth in SEQ ID NO:21 and wherein the LT- ⁇ -R-Ig-fusion proteins lack pyroglutamic acid.
  • the invention also features a composition comprising a population of LT- ⁇ -R-Ig fusion proteins comprising a variant LT- ⁇ -R extracellular domain of 193 or 194 amino acids in length and a variant Ig portion, wherein the population has reduced N-terminal pyroglutamic acid formation, and reduced C-terminal heterogeneity compared to wild-type LT- ⁇ -R- Ig fusion proteins.
  • the amino terminal amino acid of the variant LTBR-Ig fusion protein may be an alanine, as set forth in SEQ ID NOs: 5 and 12, or, alternatively, may be a non-polar amino acid.
  • non polar amino acids which may be used as the first amino acid of the LTBR-Ig fusion protein include a valine (amino acid six of the mature form of the wild type LT- ⁇ -R portion SEQ ID NO:1) or an alanine (amino acid five of the mature form of the wild type LT- ⁇ -R portion SEQ ID NO: 1).
  • non-polar amino acids serine or threonine may be used.
  • the composition of the invention includes a population of LTBR-Ig fusion proteins whereby the N-terminus of at least 95% of the LT- ⁇ -R-Ig-fusion proteins is either a valine (amino acid six of the mature form of wild type LT- ⁇ -R) or an alanine (amino acid five of the mature form of wild type LT- ⁇ -R).
  • heterogeneity may also result from variable glycosylation.
  • the LTBR and/or the Ig may be altered to reduce the amount of glycosylation.
  • the Ig portion of the LTBR-Ig fusion protein is non-glycosylated.
  • the invention also contemplates a composition comprising a population of LT ⁇ R-Ig fusion proteins comprising a variant LT- ⁇ -R extracellular domain and a variant Ig portion, wherein variant LT- ⁇ -R extracellular domain is aglycosylated.
  • An example of an aglycosylated extracellular domain of LTBR is provided in the Examples, and is also described in SEQ ID NO: 10 (see amino acids 1 to 194 of SEQ ID NO: 10).
  • examples of post-translational heterogeneity that may be decreased using the methods and compositions described herein include N-terminal heterogeneity, C-terminal heterogeneity, and glycosylation heterogeneity. It is within the scope of the invention to provide LTBR-Ig fusion proteins having reduced heterogeneity in each of these parameters, as well as combinations thereof.
  • the invention includes compositions comprising a population of LT- ⁇ -R-Ig fusion proteins, wherein the population has reduced N-terminal heterogeneity and reduced C-terminal heterogeneity, compared to a population of wild-type LT- ⁇ -R-Ig fusion proteins.
  • An LTBR-Ig fusion polypeptide of the invention retains the ligand binding activity of LTBR, and includes those polypeptides which have an amino acid sequence that has at least 70% homology to the LTBR-Ig polypeptides set forth in SEQ ID NOs: 5, 6, 8, 9, 10, 11, 12, and 23 and variants and derivatives of each of the foregoing.
  • the invention also includes isolated polypeptides described in SEQ ID NOs: 3, 4, and 7, which provide either LTBR extracellular domains of the invention or Ig variants of the invention.
  • the LTBR-Ig polypeptide (or portion thereof) has an amino acid sequence greater than 85% homology, more preferably greater than 90% homology, more preferably greater than 95% homology, most preferably greater than 99% homology, to the foregoing sequences.
  • the invention features an isolated polypeptide comprising an amino acid sequence which is at least 90% identical to the full length of the mature form of SEQ ID NO:8 or SEQ ID NO:23, wherein the amino acid sequence of the ligand binding domain is unchanged, the polypeptide further comprising a truncated Fc region.
  • the polypeptide of the invention comprises an amino acid sequence is at least 95% identical to the full length of the mature form of the polypeptide set forth in SEQ ID NO: 8.
  • the polypeptide of the invention comprises an amino acid sequence of the mature form of the polypeptide set forth in SEQ ID NO: 8.
  • the polypeptide of the invention comprises the amino acid sequence set forth in SEQ ID NO: 5.
  • variants of the polypeptides described herein are also contemplated by the invention.
  • variants of the polypeptide having an amino acid sequence that differs from the sequence presented in SEQ ID NO: 5, SEQ ID NO: 6, SEQ ID NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ ID NO: 12, and SEQ ID NO: 23, as well as portions thereof described in SEQ ID NO: 3, SEQ ID NO: 4, and SEQ ID NO: 7, at one or more amino acid positions are included in the invention.
  • variant polypeptides include the modified polypeptides described above, as well as conservative substitutions, splice variants, isoforms, homologues from other species, and polymorphisms, the
  • Modifications of an LTBR-Ig primary amino acid sequence may result in proteins which have substantially equivalent activity as compared to the unmodified counterpart polypeptide, and thus may be considered functional analogous of the parent proteins. Such modifications may be deliberate, e.g. as by site-directed mutagenesis, or they may occur spontaneous, and include splice variants, isoforms, homologues from other species, and polymorphisms. Such functional analogs are also contemplated according to the invention. [00153] Moreover, modifications of the primary amino acid sequence may result in proteins which do not retain the biological activity of the parent protein, including dominant negative forms, etc. A dominant negative protein may interfere with the wild-type protein by binding to, or otherwise sequestering regulating agents, such as upstream or downstream components, that normally interact functionally with the polypeptide. Such dominant negative forms are also contemplated according to the invention.
  • the LTBR-Ig proteins of the invention may be made according to standard methods known in the art.
  • a nucleic acid molecule encoding LTBR-Ig may be used to express a LTBR-Ig polypeptide, e.g., by expressing a LTBR-Ig polypeptide in vivo, or by administering a nucleic acid molecule encoding LTBR-Ig to an animal for in vivo expression.
  • Nucleic acid molecules encoding LTBR-Ig may be included within a nucleic acid vector, e.g., an expression vector or a cloning vector.
  • a nucleic acid molecule encoding a LTBR-Ig may, but need not of necessity, be maintained, reproduced, transferred, or expressed as part of a nucleic acid vector.
  • another aspect of the invention is isolated nucleic acid molecules which encode the LTBR-Ig proteins of the invention described herein.
  • the invention includes a nucleic acid molecule comprising the nucleotide sequence set forth in SEQ ID NO: 7 (mature form of LTBRO ⁇ ).
  • a recombinant expression vector containing a LTBR-Ig polynucleotide sequence can be introduced into and/or maintained within a cell. Cells hosting a LTBR-Ig vector may be prokaryotic.
  • a LTBR-Ig nucleic acid can be introduced into a eukaryotic cell, e.g., a eukaryotic cell that contains the appropriate apparati for post-translational processing of a polypeptide into a mature protein, and/or the appropriate apparati for secreting a polypeptide into the extracellular environment of the cell.
  • vectors comprising nucleic acid molecules encoding the LTBR-Ig fusion proteins of the invention.
  • LTBR-Ig proteins of the invention are known in the art and are described, for example, in WO 97/03687, WO 98/17313, WO 00/21558, WO 99/38525, WO 00/36092.
  • an LTBR immunoglobulin fusion protein can be expressed in cell culture (e.g., mammalian cell culture (such as monkey cos cells or Chinese hamster ovary cells) or yeast cell culture) at a reduced temperature, e.g, to produce an increased amount of properly folded fusion protein.
  • host cells expressing LTBR-Ig fusions proteins of the invention, where the host cell comprises a vector comprising a nucleic acid encoding an LTBR-Ig fusion protein.
  • the host cell is a Chinese Hamster Ovary (CHO) cell.
  • the expressed fusion protein can be purified, e.g., by affinity or conventional chromatography techniques. See WO 00/36092. Expression of the LTBR-Ig fusion protein may range in scale, including manufacturing scale.
  • the invention further provides a process for making a composition comprising a population of LT ⁇ R-Ig-fusion proteins which comprise a variant LT- ⁇ - R extracellular domain and a variant Ig portion, wherein at least 90% of the LT ⁇ R-Ig- fusion proteins are missing no more than 5 amino acids from the N-terminus of the mature form of the wild type LT- ⁇ -R portion set forth in SEQ ID NO:1, the process comprising, expressing a nucleic acid molecule encoding the LT- ⁇ -R- Ig fusion protein set forth in SEQ ID NO: 8 in a mammalian cell, obtaining the population from the culture supernatant, and, optionally, purifying the supernatant, to thereby obtain a composition comprising a population of LT ⁇ R-Ig-fusion proteins which comprise a variant LT- ⁇ -R extracellular domain and a variant Ig portion, wherein at least 90% of the LT- ⁇ -R-Ig-fusion proteins are missing no more than 5 amino
  • the process comprises expressing a nucleic acid molecule set forth in SEQ ID NO:7 in a mammalian cell. In one embodiment, the process comprises expressing a nucleic acid molecule set forth in SEQ ID NO: 7 in a mammalian cell.
  • the invention also features expressing a nucleic acid molecule comprising a nucleotide sequence encoding the extracellular domain of LTBR set forth in SEQ ID NO:4, or the nucleic acid sequence set forth in SEQ ID NO: 3 in a mammalian cell.
  • the invention further features a composition which is made by expressing a nucleic acid molecule encoding the LT- ⁇ -R- Ig fusion protein set forth in SEQ ID NO:8, or the nucleic acid sequence set forth in SEQ ID NO: 7, in a mammalian cell.
  • LTBR-IgG of Invention are lymphotoxin (LT) pathway inhibitors useful for treating autoimmune disorders.
  • Autoimmune disorders include, for example, autoimmune arthritides (including rheumatoid arthritis (RA) and Sjogren's syndrome) psoriasis, multiple sclerosis, inflammatory bowel disease (IBD) (including ulcerative colitis and Crohn's disease), insulin-dependent diabetes, uveitis, systemic lupus erythematosus (SLE, or lupus), polychondritis, and transplant rejection.
  • RA rheumatoid arthritis
  • IBD inflammatory bowel disease
  • SLE systemic lupus erythematosus
  • polychondritis and transplant rejection.
  • the agents and methods described herein are particulary suitable for treatment of RA.
  • Rheumatoid arthritis is marked by tenderness in the joints. Synovial thickening eventually occurs in most effected joints. Stiffness lasting >30 minuts upon arising in the morning or after prolonged inactivity is common, as is early afternoon fatigue and malaise. Deformities, particularly flexion contractures may develop rapidly. Carpal tunnel syndrome can result from wrist synovitis. X-rays may reveal soft-tissue swelling in the first months of disease, and subsequently, perarticular osteoporosis, joint space narrowing, and marginal erosions may be present. [00160] Various tests can be employed to assay the efficacy of a soluble LTBR to treat autoimmune disorders.
  • insufficient efficacy may be defined as failure to exhibit at least a 10%, 20%, 25%, 30%, 40%, 50% or more decrease in a clinical parameter of RA, such as tender joint count (TJC), swollen joint count (SJC), patient global assessment of disease activity [PGA visual analogue scale (VAS) 0-10 cm, for measurement of pain], physician global assessment of disease activity (MDGAVAS 0-10 cm), levels of C-reactive protein (CRP in mg/dl), and erythrocyte sedimentation rate (ESR).
  • C-reactive protein is produced by the liver during episodes of acute inflammation or infection. CRP levels in blood serum are an indicator of the severity of RA.
  • a decrease in CRP levels can be an indication of effective treatment of autoimmune disorder, such as RA, in response to treatment with a soluble LTBR, such as LTBR-Fc.
  • Erythrocyte sedimentation rate (ESR) is also elevated in 90% of RA cases.
  • ESR Erythrocyte sedimentation rate
  • soluble LTBR e.g., an LTBR-Fc
  • autoimmune arthritides including rheumatoid arthritis and Sjogren's syndrome
  • psoriasis multiple sclerosis
  • IBD inflammatory bowel disease
  • IBD insulin-dependent diabetes
  • uveitis systemic lupus erythematosus (SLE, or lupus)
  • polychondritis and transplant rejection.
  • the invention is based, in part, on the discovery that low dosage or frequency regimens of a soluble form of LTBR (e.g., LTBR-Fc) can effectively treat symptoms of an autoimmune disorder, and rheumatoid arthritis (RA) in particular.
  • a soluble form of LTBR e.g., LTBR-Fc
  • RA rheumatoid arthritis
  • the invention provides methods of treating autoimmune disease in a subject.
  • the method includes administering to the subject an LTBR blocking agent (e.g., a soluble LTBR fusion protein, e.g., LTBR-Fc) at a low dose and/or frequency.
  • an LTBR blocking agent e.g., a soluble LTBR fusion protein, e.g., LTBR-Fc
  • a soluble LTBR fusion protein is administered to achieve an average concentration of between about 0.14 ug/ml to about 3.5 ug/ml. In one embodiment, a soluble LTBR fusion protein is administered to achieve a minimal average concentration of about 0.3 to about 1.0 ug/m., e.g., of about 0.6 to 0.7 ug/ml. In another embodiment, higher doses of the fusion protein may be administered. For example, the fusion protein may be administered to achieve an average concentration of between about 5 and 15 ug/ml.
  • a soluble LTBR fusion protein is administered at a dose of between about 70 and about 200 mg/month to an average subject weighing about 75 kg. In another embodiment, a soluble LTBR fusion protein is administered at a dose of about 100, about 150, or about 175 mg/month to an average subject weighing about 75 kg.
  • Embodiments of the invention can include administration of a regimen of LTBR blocking agent, e.g., a soluble LTBR, such as an LTBR fusion protein (e.g., LTBR-Ig), for the treatment of autoimmune arthritides (including rheumatoid arthritis and Sjogren's syndrome), psoriasis, multiple sclerosis, inflammatory bowel disease (IBD) (including ulcerative colitis and Crohn's disease), insulin-dependent diabetes, uveitis, systemic lupus erythematosus (SLE, or lupus), polychondritis, and transplant rejection.
  • a soluble LTBR e.g., an LTBR fusion protein
  • LTBR immunoglobulin fusion Exemplary LTBR-Ig fusion proteins are described herein.
  • the subject has one or more symptoms of RA, e.g., joint swelling, joint pain, joint stiffness, or difficulty moving.
  • the subject has active RA disease (e.g., tender joint count (TJC) or swollen joint count (SJC) > 5 or synovitis).
  • the subject is at risk for developing RA, e.g., a family history, or an autoimmune disease.
  • the soluble LTBR can be administered in an amount and/or for a time sufficient to alleviate the symptoms associated with RA.
  • the soluble LTBR e.g., an LTBR-Fc
  • the soluble LTBR is administered in an amount sufficient to improve symptoms in one or more RA assessment criterion, e.g., a criterion described herein.
  • the soluble LTBR is administered in an amount sufficient to improve symptom scores at least about 10%, 20%, 25%, 30%, 40%, 50% or more.
  • Symptom scores refer, for example, _ to TJC, SJC, patient global assessment of disease activity [PGA visual analog scale (VAS) 0-10 cm], physician global assessment of disease activity (MDGA VAS 0-10 cm) quantities of C-reactive protein (CRP in mg/dl), or Disease Activity Score, 28- joint version (DAS28).
  • AnACR score is an indication of the percent clinical improvement in RA symptoms.
  • An ACR20 is an indication of a 20% clinical improvement in TJC and SJC, as well as a 20% improvement in three of the following five parameters: (i) patient's global assessment, (ii) physician's global assessment, (iii) patient's assessment of pain, (iv) degree of disability, and (v) level of acute-phase reactant.
  • ACR scores of ACR50 and ACR70 can also be used to indicate a 50% improvement, or a 70% improvement, respectively.
  • the soluble LTBR e.g., LTBR-Fc
  • the soluble LTBR is administered more than once, e.g., weekly, biweekly, or monthly.
  • the course of treatment can be maintained for a period of time, such as for 1 or 2 weeks or more; 1, 2, 3, 4, 5, or 6 months or more; 1 year; or longer.
  • the soluble LTBR is administered at a dosage of about 0.03 to 3 mg/kg of body weight per administration, e.g., about 0.6 to about 1.4 mg/kg per administration.
  • the soluble LTBR is administered at a dosage of about 0.3 -3, e.g., 1 mg/kg of body weight per administration, e.g., about 0.6 to about 1.4 mg/kg per administration. In other embodiments, the soluble LTBR is administered at a dosage of about 2.5 to 3.5 mg/kg of body weight per administration, e.g., about 2.8 to 3 mg/kg per administration.
  • the soluble LTBR is administered at a dosage of about 0.01 to 3 mg/kg of body weight per administration, e.g., about 0.01 to about 0.05 mg/kg per administration, about 0.01 to about 0.3 mg/kg per administration, about 0.01 to 0.2 mg/kg per administration, or about 0.01 to about 0.1 mg/kg per administration. In some embodiments, the soluble LTBR is administered at about 0.03 mg/kg per administration, about 0.05 mg/kg per administration, about 0.07 mg/kg per administration, about 0.1 mg/kg per administration, or about 0.2 mg/kg per administration. In some embodiments, the soluble LTBR is administered at a dosage shown in Table 1.
  • Table 1 Exemplary dosages of LTBR-Fc (me per kg of body weight of subject per administration)
  • the lower unit doses disclosed herein can be administered with more frequent dosing (e.g., weekly) while higher unit doses disclosed herein (though still low by relative standards) can be administered with more infrequent dosing (e.g., monthly).
  • the soluble LTBR e.g., LTBR-Fc
  • the soluble LTBR is administered at a dosage of about 0.3 mg/kg per administration to about 3 mg/kg per administration, e.g., about 0.6 mg/kg per day to about 1.4 mg/kg per day.
  • the dose is about 1 mg/kg, or about 3 mg/kg.
  • the dose is administered once every 14-20 days, once or twice monthly, e.g., once or twice every 28-31 days.
  • the dose is about 1 mg/kg or about 3 mg/kg administered every 14 to 20 days, e.g., every 12 to 16 days, or about every two weeks.
  • the dose is about 1 mg/kg or about 3 mg/kg administered monthly, e.g., about every 28 to 31 days.
  • the soluble LTBR e.g., LTBR-Fc
  • the soluble LTBR is administered at a dosage of about 0.6 mg/kg per administration to about 1.4 mg/kg per administration not more than twice every 20 to 40 days, e.g., not more than twice every 25 to 35 days, or not more than twice every 28-31 days.
  • the soluble LTBR e.g., LTBR-Fc
  • the soluble LTBR e.g., LTBR-Fc
  • the soluble LTBR is administered at a dosage of about 0.01 mg/kg per administration to about 0.3 mg/kg per administration, e.g., about 0.01 mg/kg per day to about 0.25 mg/kg per day, e.g., weekly, or every 3 to 10 days, repeated at least twice.
  • the soluble LTBR is administered at a dosage of about 0.02, 0.03, 0.04, 0.05, 0.06, 0.1, or 0.2 mg/kg, administered weekly or every 3-10 days, e.g., every 4, 5, 6, 7, 8, or 9 days.
  • the soluble LTBR is administered at a dosage shown in Table 1.
  • the soluble LTBR is administered at a dosage of about 0.01 mg/kg per day to about 0.3 mg/kg per day, e.g., 0.02 mg/kg per day to about 0.25 mg/kg per day, e.g., biweekly, or every 5 to 20 days, repeated at least twice.
  • the soluble LTBR is administered at a dosage of about 0.03, 0.05, 0.08, 0.1, or 0.2 mg/kg, administered biweekly, or every 5-20 days, e.g., every 6, 8, 10, 12, 14, 16 or 18 days.
  • the soluble LTBR is administered at a dosage shown in Table 1.
  • the soluble LTBR is administered at a dosage of about 0.3 mg/kg per day to about 3 mg/kg per day, e.g., 1 mg/kg per day, e.g., biweekly, or every 5 to 20 days, repeated at least twice.
  • the soluble LTBR is administered at a dosage shown in Table 1.
  • the soluble LTBR is administered at a dosage of about 0.3 mg/kg per day to about 3 mg/kg per day, e.g., 0.4 mg/kg per day to about 3 mg/kg per day, e.g., monthly, or every 15 to 45 days, repeated at least twice.
  • the soluble LTBR is administered at a dosage of about 0.5, 0.8, 1 , 1.5, or 2 mg/kg, administered monthly, or every 15-45 days, e.g., every 16, 18, 20, 25, 30, 35 or 40 days.
  • the soluble LTBR is administered at a dosage shown in Table 1.
  • the dose of soluble LTBR is about 0.4 mg to about 375 mg, about 0.4 mg to about 6.25 mg, about 2 mg to about 6.25 mg, or about 4 mg to about 12.5 mg.
  • the invention provides for the administration of a particularly low dose of a soluble LTBR, e.g., an LTBR-Fc, for treatment of an autoimmune disorder, e.g., RA.
  • the soluble LTBR is administered intravenously or parenterally, e.g., subcutaneously or intramuscularly.
  • the soluble LTBR is administered as a monotherapy.
  • the invention provides methods of treating an autoimmune condition, e.g., RA, in a subject, such as a human.
  • the method includes administering to the human a dose of a soluble LTBR, e.g., LTBR-Fc, wherein the dose is between about 0.01 mg and about 3 mg LTBR-Fc per kg body weight of the human (mg/kg), e.g., between about 0.01 mg/kg and about 0.05 mg/kg.
  • the human is administered a weekly dose of a soluble LTBR, e.g., LTBR-Fc, over the course of at least two weeks, where the dose is about 0.01 mg/kg per day to about 0.3 mg/kg per day, e.g., about 0.01 mg/kg per day to about 0.25 mg/kg per day, administered weekly, e.g., every 3 to 10 days.
  • the soluble LTBR is administered at a dosage of about 0.02, 0.03, 0.04, 0.05, 0.06, 0.1, or 0.2 mg/kg per day, e.g., every 4, 5, 6, 7, 8, or 9 days.
  • the soluble LTBR is administered at a dosage shown in Table 1.
  • the human is administered a biweekly dose of a soluble LTBR, e.g., LTBR-Fc, over the course of at least four weeks, where the dose is about 0.01 mg/kg per day to about 0.5 mg/kg per day, e.g., about 0.01 mg/kg per day to about 0.3 mg/kg per day repeated every 5 to 20 days.
  • the soluble LTBR is administered at a dosage of about 0.03, 0.05, 0.08, 0.1, 0.2, 0.3 or 0.4 mg/kg, e.g., every 6, 8, 10, 12, 14, 16 or 18 days.
  • the soluble LTBR is administered at a dosage shown in Table 1.
  • the human is administered a monthly dose of a soluble LTBR, e.g., LTBR-Fc, over the course of at least two months, where the dose is about 0.1 mg to about 3 mg soluble LTBR per kg body weight of the human per day.
  • the dose is about 0.3 mg/kg per day to about 3 mg/kg administered monthly, e.g., every 15 to 45 days.
  • the soluble LTBR is administered at a dosage of about 0.5, 0.8, 1, 1.5, or 2 mg/kg per day, e.g., every 16, 18, 20, 25, 30, 35 or 40 days.
  • the soluble LTBR is administered at a dosage shown in Table 1.
  • a patient exhibiting an inadequate response to a therapy has not shown a clinically acceptable or significant improvement in response to the therapy.
  • the patient initially showed an improvement in response to a therapy but no longer demonstrates an improvement, as assessed by a standard clinical measure for the specific disorder.
  • a DMARD-IR subject is a subject who has had an inadequate response to a disease modifying antirheumatic drug (DMARD), such as methotrexate, leflunomide (Arava®), anakinra (Kineret®), hydroxycholoquine sulfate (Plaquenil®) antimalarials, gold salts, sulfasalazine (Azulfidine®), minocycline (Minocin®), d-penicillamine, cyclosporin A, cyclosporine (Neoral®), cyclophosphamide and azathioprine (Imuran®).
  • DMARD disease modifying antirheumatic drug
  • the invention features a delivery device, e.g., a transcutaneous delivery device, e.g., a syringe, designed for subcutaneous or intramuscular administration, where the device is packaged with or contains at least one unit dose of a soluble LTBR, e.g., LTBR-Fc, such that an appropriately low quantity of the agent will be administered to a human.
  • a transcutaneous delivery device e.g., a syringe
  • LTBR-Fc soluble LTBR
  • the device contains or is packaged with a unit dose of LTBR-Fc such that administration to a human will deliver between about 0.01 mg/kg and 3 mg/kg LTBR-Fc, e.g., about 0.01 to about 0.05 mg/kg LTBR-Fc, or about 0.6 mg/kg and 1.4 mg/kg LTBR-Fc, to the human.
  • the device contains or is packaged with a unit dose of LTBR-Fc such that administration to a human will deliver between about 2.5 mg/kg and 3.5 mg/kg LTBR-Fc to the human.
  • the delivery device will deliver about 1 mg/kg or about 3 mg/kg LTBR-Fc to the human.
  • Exemplary unit dose amounts appropriate for humans of various weights is provided in Table 2.
  • the liquid in the second compartment is water or a buffer.
  • the liquid can include, e.g., a pharmaceutically acceptable carrier, such as a solvent, dispersion media, antibacterial or antifungal agent, or isotonic or absorption- delaying agent.
  • a pharmaceutically acceptable carrier such as a solvent, dispersion media, antibacterial or antifungal agent, or isotonic or absorption- delaying agent.
  • the liquid may also include a pharmaceutically acceptable salt.
  • the unit dose of the soluble LTBR e.g., a soluble LTBR
  • LTBR-Fc included in a delivery device is about 0.4 mg to about 375 mg, about 0.4 mg to about 6.25 mg, about 2 mg to about 6.25 mg, or about 4 mg to about 12.5 mg.
  • the device contains lyophilized LTBR-Fc.
  • the invention features a kit including two or more unit doses of between about 0.4 mg to about 375 mg LTBR-Fc.
  • the unit doses are such that an appropriately low quantity will be administered to a human, as determined by the weight of the human.
  • Exemplary unit dose amounts appropriate for humans of various weights is provided in Table 2.
  • the invention features a device, e.g., a transcutaneous delivery device, e.g., a syringe having at least two compartments, where a first compartment contains a unit dose of lyophilized LTBR-Fc and a second compartment contains a liquid for reconstituting the LTBR-Fc prior to administration to a subject.
  • a transcutaneous delivery device e.g., a syringe having at least two compartments
  • a first compartment contains a unit dose of lyophilized LTBR-Fc
  • a second compartment contains a liquid for reconstituting the LTBR-Fc prior to administration to a subject.
  • he unit dose is such that administration of the LTBR-Fc will deliver between about 0.01 to 3 mg/kg, e.g., 1.0 mg/kg LTBR-Fc to the human.
  • the unit dose of LTBR-Fc is between about 0.4 mg and about 375 mg LTBR-Fc, e.g., between about 0.4 mg and about 6.25 mg LTBR-Fc.
  • the unit doses are such that an appropriately low quantity will be administered to a human, as determined by the weight of the human.
  • Exemplary unit dose amounts appropriate for humans of various weights is provided in Table 2.
  • the invention features a method of instructing a patient having rheumatoid arthritis to treat the RA by (i) providing the patient with at least two unit doses of LTBR-Fc; and (ii) instructing the patient to self-administer the unit doses, e.g., subcutaneously, one dose at a time.
  • the dose delivered will be between about 0.01 mg and 3 mg LTBR-Fc per kg of body weight of the patient (mg/kg), e.g., between about 0.6 mg/kg and 1.4 mg/kg, or about 0.01 mg/kg and 0.05 mg/kg.
  • the dose delivered will be between about 2.5 to 3.5 mg/kg LTBR-Fc per kg of body weight of the patient (mg/kg).
  • the patient is instructed to self-administer the doses weekly, over the course of at least 2 weeks; biweekly, over the course of at least 4 weeks; or monthly, over the course of at least 2 months.
  • the unit dose is about 0.01 to about 3.0 mg/kg LTBR-Fc per administration.
  • the unit dose is about 0.01 to about 2.5 mg/kg per administration, about 0.02 to about 0.5 mg/kg per administration, about 0.01 to about 0.3 mg/kg per administration, about 0.01 to about 0.2 mg/kg per administration, about 0.01 to about 0.1 mg/kg per administration, or about 0.01 to about 0.05 mg/kg per administration. In certain embodiments, the unit dose is about 0.03 mg/kg, about 0.05 mg/kg, about 0.07 mg/kg, about 0.1 mg/kg, or about 0.2 mg/kg per administration.
  • the patient is instructed to self-administer the doses weekly, over the course of at least 2 weeks, where the unit dose is about 0.01 mg/kg to about 3.0 mg/kg LTBR-Fc per administration, e.g., about 0.01 mg/kg to about 0.25 mg/kg, about 0.01 mg/kg to about 0.05 mg/kg LTBR-Fc per administration.
  • the unit dose is about 0.02, 0.03, 0.04, 0.05, 0.06, 0.1, or 0.2 mg/kg per administration.
  • the patient is instructed to self-administer the doses biweekly, over the course of at least 4 weeks, where the unit dose is between about 0.01 andl.5 mg/kg, e.g., about 0.01 to about 0.3, or about 0.02 mg/kg to about 0.3 mg/kg or about 0.5-1.25 mg/kg LTBR-Fc.
  • the unit dose is about 0.03, 0.05, 0.08, 0.1, 0.2, 0.3, 0.4, 0.5, 0.75, or 1.0 mg/kg per administration.
  • the patient is instructed to self-administer the doses monthly, over the course of at least 2 months, where the unit dose is between about 0.03 to about 3 mg LTBR-Fc per kg of body weight of the patient (mg/kg).
  • the unit dose is about 0.2, 0.5, 0.8, 1, 1.5, or 2 mg/kg.
  • the invention features a method of managing RA in a patient including (i) instructing a patient to stop taking a therapy to treat RA, and (ii) administering to the patient a unit dose of LTBR-Fc.
  • the unit dose is between about 0.03 to about 3 mg/kg LTBR-Fc, e.g., about 0.6 to about 1.4 mg/kg LTBR-Fc. In another embodiment, the unit dose is about 2.5 to about 3.5 mg/kg LTBR-Fc. In other embodiments, the unit dose is between about 0.01 to about 0.05 mg/kg LTBR-Fc. [00196] In another embodiment, the unit dose is between about 0.3 mg/kg and
  • the unit dose is administered not more than twice every 20-40 days, e.g., every 25-35 days, or 28-31 days.
  • the unit dose is between about 0.6 mg/kg and about 1.4 mg/kg LTBR-Fc, administered not more than twice every 20-40 days, e.g., every 25-25 days, or every 28-31 days.
  • the unit dose is between about 0.01 mg/kg and
  • the unit dose is administered not more than once every 3-10 days, e.g., weekly.
  • the unit dose is between about 0.01 mg/kg and about 0.3 mg/kg LTBR-Fc administered not more than once every 5-20 days, e.g., biweekly.
  • the unit dose is between about 0.3 mg/kg and about 3 mg/kg LTBR-Fc administered not more than once every 28-31 days, e.g., monthly.
  • the unit dose is about 0.01 mg/kg to about 0.25 mg/kg LTBR-Fc, and the unit dose is administered weekly over the course of at least 2 weeks.
  • the unit dose is about 0.02, 0.03, 0.04, 0.05, 0.06, 0.1, or 0.2 mg/kg per administration.
  • the unit dose is about 0.02 mg/kg to about 0.5 mg/kg LTBR-Fc, and the unit dose is administered biweekly over the course of at least 4 weeks.
  • the unit dose is about 0.03, 0.05, 0.08, 0.1, 0.2, 0.3 or 0.4 mg/kg per administration.
  • the unit dose is about 0.03 to about 3 mg
  • the unit dose is administered monthly over the course of at least 2 months.
  • the unit dose is about 0.2, 0.5, 0.8, 1, 1.5, or 2 mg/kg per administration.
  • the patient is instructed to stop an NSAID, corticosteroid, or DMARD therapy.
  • the invention features a method of managing RA in a patient that includes administering to the patient a unit dose of a soluble LTBR, e.g., LTBR-Fc, where (i) the unit dose is between about 0.01 mg and about 0.05 mg of the soluble LTBR per kg of body weight of the patient (mg/kg); (ii) the unit dose is between about 0.01 mg/kg and 0.3 mg/kg LTBR-Fc administered not more than once every 3-10 days, e.g., weekly; (iii) the unit dose is between about 0.01 mg/kg and 1 mg/kg LTBR-Fc administered not more than once every 5-20 days, e.g., biweekly; (iv) the unit dose is between about 0.3 mg/kg and 3 mg/kg LTBR-Fc (e.g., between about 0.6 and 1.4 mg/kg LTBR-Fc) administered not more than twice every 20-40 days, e.g., every 25-35
  • the patient receives a first therapy, e.g., an NSAID, corticosteroid, or DMARD therapy, to treat RA prior to the administration of the soluble LTBR, and the levels of the first therapy are maintained in the patient at least when the first unit dose of the soluble LTBR is administered to the patient.
  • a first therapy e.g., an NSAID, corticosteroid, or DMARD therapy
  • the levels of the first therapy are maintained in the patient at least when the first unit dose of the soluble LTBR is administered to the patient.
  • the levels of the first therapy are not maintained in the patient after the first or second unit dose of the soluble LTBR.
  • the levels of the first therapeutic agent are diminished, e.g., by administration of lower doses of the first therapeutic agent, or by ceasing administration and allowing the levels of the first therapeutic agent to be cleared from the body.
  • the patient continues to receive the first therapy during the administration of the soluble LTBR.
  • NSAIDs typically relieve pain by reducing inflammation. Suitable
  • NSAIDs include, for example, aspirin, ibuprofen, naproxen, and ketoprofen.
  • COX-2 inhibitors are a similar class of drugs that can be used in combination with a soluble salt.
  • LTBR for treatment of an autoimmune disorder.
  • DMARDs typically slow the progression of rheumatoid arthritis.
  • Suitable DMARDS include methotrexate, leflunomide (Arava®), anakinra
  • TNF inhibitors also include TNF inhibitors.
  • Tumor necrosis factor alpha (TNF- ⁇ ) is a pro- inflammatory cytokine produced by macrophages and lymphocytes. TNF inhibitors help to relieve the proinflammatory effects of this molecule. TNF inhibitors include etanercept (EnbrelTM), infliximab (RemicadeTM), and adalimumab (HumiraTM).
  • Corticosteroids typically function as anti-inflammatory agents and can also be used in combination therapy with a soluble LTBR.
  • a soluble LTBR is use as a second line therapy.
  • a patient who is determined to be a DMARD-IR will stop receiving treatment with DMARD and will begin treatment with a soluble LTBR, e.g., LTBR-Fc.
  • a patient receiving a combination therapy of NSAID and DMARD will stop receiving the DMARD and will begin receiving a soluble LTBR in combination with the NSAID.
  • the soluble LTBR may be administered in combination with NSAID, or separately.
  • a patient receiving a daily dose of NSAID may only receive a weekly, or biweekly, or monthly dose of a soluble LTBR.
  • the patient will continue to receive the DMARD and/or NSAID therapies while receiving treatment with the soluble LTBR.
  • the soluble LTBR is administered in combination with a second treatment for RA.
  • the combination therapy includes administering a second agent that provides a therapeutic benefit to a patient who has or is at risk for RA.
  • Exemplary second agents include, e.g., non-steroidal anti-inflammatory agents (NSAIDs), corticosteroids, or disease modifying antirheumatic drugs (DMARDs).
  • the subject is treated with an RA drug after being diagnosed with RA and prior to administration of a soluble LTBR.
  • the RA drug can be, for example, an NSAID, corticosteroid, or DMARD.
  • the subject is evaluated to determine if the response to the RA drug is inadequate prior to administration of the soluble LTBR. In certain embodiments, if the subject is determined to have an inadequate response to the RA drug, then the subject is administered a soluble LTBR. In one embodiment, the subject is determined to be asymptomatic, or an adequate responder, for a first manifestation of RA, such as TJC or SJC.
  • the subject is determined to be asymptomatic, or an adequate responder, for a first manifestation of RA, such as TJC or SJC, and symptomatic, or an inadequate responder for a second manifestation of rheumatoid arthritis, such as synovitis.
  • Synovitis can be detected by any method known in the art, including, for example, by magnetic resonance imaging (MRI).
  • MRI magnetic resonance imaging
  • the soluble LTBR and the second agent are administered at the same time.
  • the soluble LTBR is administered first in time and the second agent is administered second in time.
  • the second agent is administered first in time and the soluble LTBR is administered second in time.
  • the soluble LTBR can replace or augment a previously or currently administered therapy.
  • administration of the second agent can cease or diminish, e.g., be administered at lower levels.
  • administration of the previous therapy is maintained.
  • a previous therapy will be maintained until the level of LTBR reaches a level sufficient to provide a therapeutic effect.
  • the two drugs can be administered in combination.
  • a human receiving a first therapy for RA e.g., an
  • NSAID corticosteroid, or DMARD
  • a soluble LTBR e.g., LTBR-Fc.
  • the first therapy is halted.
  • the human is monitored for a first preselected result, e.g., an improvement in RA symptoms, e.g., a. decrease in TJC or SJC by 10%, 20%, 30%, or more.
  • treatment with the soluble LTBR is decreased or halted.
  • the human is then monitored for a second preselected result after treatment with the soluble LTBR is halted, e.g., a worsening of an RA symptom, e.g., an increase in TJC or SJC by 10%, 20%, 30% or more.
  • a second preselected result e.g., a worsening of an RA symptom, e.g., an increase in TJC or SJC by 10%, 20%, 30% or more.
  • administration of the soluble LTBR to the human is reinstated or increased, or administration of the first therapy is reinstated, or the human is administered both a soluble LTBR, or an increased amount of soluble LTBR, and the first therapeutic regimen.
  • a human receiving a first therapy for RA who is then treated with a soluble LTBR, e.g., an LTBR-Fc, continues to receive the first therapy at the same or a reduced amount.
  • a soluble LTBR e.g., an LTBR-Fc
  • treatment with the first therapy overlaps for a time with treatment with the soluble LTBR, but treatment with the first therapy is subsequently halted.
  • the subject is a DMARD inadequate responder
  • the method includes evaluating the subject for an improvement in RA symptoms.
  • the evaluation is performed at least 1 hour, e.g., at least 2, 4, 6, 8, 12, 24, or 48 hours, or at least 1 day, 2 days, 4 days, 10 days, 13 days, 20 days or more, or at least 1 week, 2 weeks, 4 weeks, 10 weeks, 13 weeks, 20 weeks or more, after the administration of the soluble LTBR.
  • the subject can be evaluated in one or more of the following periods: prior to beginning of treatment; during treatment; or after one or more elements of the treatment have been administered.
  • Evaluating can include evaluating the need for further treatment with the same soluble LTBR or for additional treatment with additional agents.
  • an additional step is taken, e.g., the subject is administered another treatment or another evaluation or test is performed.
  • a soluble LTBR e.g., LTBR-Fc
  • a pharmaceutical composition can be formulated as a pharmaceutical composition, e.g., for administration to a subject to treat an autoimmune disorder, such as RA.
  • a pharmaceutical composition includes a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the composition can include a pharmaceutically acceptable salt, e.g., an acid addition salt or a base addition salt (see e.g., Berge et al., J. Pharm. Sci. 66:1-19, 1977).
  • the soluble LTBR can be formulated according to standard methods.
  • a soluble LTBR e.g., LTBR-Fc
  • excipient materials such as sodium chloride, sodium dibasic phosphate heptahydrate, sodium monobasic phosphate, and a stabilizer. It can be provided, for example, in a buffered solution at a suitable concentration and can be stored at 2-8°C.
  • compositions may be in a variety of forms. These include, for example, liquid, semi-solid and solid dosage forms, such as liquid solutions (e.g., injectable and infusible solutions), dispersions or suspensions, tablets, pills, powders, liposomes and suppositories.
  • liquid solutions e.g., injectable and infusible solutions
  • dispersions or suspensions tablets, pills, powders, liposomes and suppositories.
  • the preferred form can depend on the intended mode of administration and therapeutic application.
  • compositions for the agents described herein are in the form of injectable or infusible solutions.
  • Such compositions can be administered by a parenteral mode (e.g., intravenous, subcutaneous, intraperitoneal, or intramuscular injection).
  • parenteral administration and “administered parenterally” as used herein mean modes of administration other than enteral and topical administration, usually by injection, and include, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural, intracerebral, intracranial, intracarotid and intrasternal injection and infusion.
  • the composition can be formulated as a solution, microemulsion, dispersion, liposome, or other ordered structure suitable for stable storage at high concentration.
  • Sterile injectable solutions can be prepared by incorporating an agent described herein in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating an agent described herein into a sterile vehicle that contains a basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying that yields a powder of an agent described herein plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the proper fluidity of a solution can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prolonged absorption of injectable compositions can be brought about by including in the composition an agent that delays absorption, for example, monostearate salts and gelatin.
  • the soluble LTBR may be prepared with a carrier that will protect the compound against rapid release, such as a controlled release formulation, including implants, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J.R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
  • a soluble LTBR (e.g., LTBR-Fc) can be modified, e.g., with a moiety that improves its stabilization and/or retention in circulation, e.g., in blood, serum, or other tissues, e.g., by at least 1.5, 2, 5, 10, or 50 fold.
  • the modified agent can be evaluated to assess whether it can reach sites of inflammation such as may occur in an autoimmune disorder, such as RA (e.g., by using a labeled form of the agent).
  • the soluble LTBR can be associated with a polymer, e.g., a substantially non-antigenic polymer, such as a polyalkylene oxide or a polyethylene oxide.
  • Suitable polymers will vary substantially by weight. Polymers having molecular number average weights ranging from about 200 to about 35,000 Daltons (or about 1,000 to about 15,000, and 2,000 to about 12,500) can be used.
  • a soluble LTBR can be conjugated to a water soluble polymer, e.g., a hydrophilic polyvinyl polymer, e.g., polyvinylalcohol or polyvinylpyrrolidone.
  • polymers include polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof, provided that the water solubility of the block copolymers is maintained.
  • Additional useful polymers include polyoxyalkylenes such as polyoxyethylene, polyoxypropylene, and block copolymers of polyoxyethylene and polyoxypropylene (Pluronics); polymethacrylates; carbomers; and branched or unbranched polysaccharides.
  • the two agents can be formulated separately or together.
  • the respective pharmaceutical compositions can be mixed, e.g., just prior to administration, and administered together or can be administered separately, e.g., at the same or different times.
  • a pharmaceutical composition comprises a population of lymphotoxin- ⁇ receptor (LT- ⁇ -R)-Ig-fusion proteins which comprise a variant LT- ⁇ -R extracellular domain of 193 or 194 amino acids in length and a variant Ig portion of 227 amino acids in length, wherein at least 90% of the LT- ⁇ -R-Ig-fusion proteins are missing no more than 5 amino acids from the N-terminus of the mature form of the wild type LT- ⁇ -R extracellular domain set forth in SEQ ID NO:21and wherein the LT- ⁇ -R-Ig-fusion proteins lack N-terminal pyroglutamic acid and a pharmaceutically acceptable carrier.
  • LT- ⁇ -R lymphotoxin- ⁇ receptor
  • a pharmaceutical composition comprises a population of lymphotoxin- ⁇ receptor-immunoglobulin (LT- ⁇ -R-Ig)-fusion proteins, the fusion proteins comprising a variant LT- ⁇ -R extracellular domain of 193 or 194 amino acids in length and a variant Ig portion, wherein the population has reduced N- terminal pyroglutamic acid formation and reduced C-terminal heterogeneity compared to wild-type LT- ⁇ -R-Ig fusion proteins and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition of the invention comprises the amino acid sequence set forth in SEQ ID NO:5.
  • a soluble LTBR (e.g., LTBR-Fc) can be administered to a subject, e.g., a human subject, by a variety of methods.
  • the route of administration is one of: intravenous injection or infusion (IV), subcutaneous injection (SC), intraperitoneally (IP), or intramuscular injection.
  • IV intravenous injection or infusion
  • SC subcutaneous injection
  • IP intraperitoneally
  • administration may be directly into the CNS, e.g., intrathecal, intracerebroventricular (ICV), intracerebral or intracranial.
  • the agent can be administered as a fixed dose, or in a mg/kg dose.
  • the dose can also be chosen to reduce or avoid production of antibodies against the agent.
  • the route and/or mode of administration of the soluble LTBR can also be tailored for the individual case, e.g., by monitoring the subject, e.g., using TJC, SJC, CRP levels and standard parameters associated with RA or other autoimmune diseases, e.g., the assessment criteria described herein.
  • Dosage regimens are adjusted to provide the desired response, e.g., a therapeutic response or a combinatorial therapeutic effect.
  • a low dose of a soluble LTBR e.g., LTBR-Fc
  • an appropriate dose of a second therapeutic agent can be used to provide a subject with the soluble LTBR.
  • Exemplary doses of the soluble LTBR are described herein.
  • Dosage unit form or "fixed dose" as used herein refers to physically discrete units suited as unitary dosages for the subjects to be treated; each unit contains a predetermined quantity of active compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier and optionally in association with the other agent.
  • a pharmaceutical composition may include a therapeutically effective amount of a soluble LTBR described herein. Such effective amounts can be determined based on the effect of the administered agent, or the combinatorial effect of an agent and secondary agent if more than one agent is used.
  • a therapeutically effective amount of an agent may also vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of the compound to elicit a desired response in the individual, e.g., amelioration of at least one disorder parameter, e.g., RA parameter, or amelioration of at least one symptom of the disorder, e.g., RA.
  • a therapeutically effective amount is also one in which any toxic or detrimental effects of the composition is outweighed by the therapeutically beneficial effects.
  • a therapeutically effective amount is a low dose as described elsewhere herein.
  • compositions that include a soluble LTBR can be administered with a medical device.
  • the device can be designed with features such as portability, room temperature storage, and ease of use so that it can be used in emergency situations, e.g., by an untrained subject or by emergency personnel in the field, removed to medical facilities and other medical equipment.
  • the device can include, e.g., one or more housings for storing pharmaceutical preparations that include a soluble LTBR, and can be configured to deliver one or more unit doses of the agent.
  • the pharmaceutical composition can be administered with a transcutaneous delivery device, such as a syringe, including a hypodermic or multichamber syringe.
  • a transcutaneous delivery device such as a syringe, including a hypodermic or multichamber syringe.
  • Other suitable deliver devices include stents, catheters, transcutaneous patches, microneedles, and implantable controlled release devices.
  • the pharmaceutical composition can be administered with a needleless hypodermic injection device, such as the devices disclosed in US 5,399,163; 5,383,851; 5,312,335; 5,064,413; 4,941,880; 4,790,824; or 4,596,556.
  • implants and modules examples include: US 4,487,603, which discloses an implantable micro-infusion pump for dispensing medication at a controlled rate; US 4,486,194, which discloses a therapeutic device for administering medicants through the skin; US 4,447,233, which discloses a medication infusion pump for delivering medication at a precise infusion rate; US 4,447,224, which discloses a variable flow implantable infusion apparatus for continuous drug delivery; US 4,439,196, which discloses an osmotic drug delivery system having multi- chamber compartments; and US 4,475,196, which discloses an osmotic drug delivery system. Many other devices, implants, delivery systems, and modules are also known.
  • a soluble LTBR (e.g., an LTBR-Fc) can be provided in a kit.
  • the kit includes (a) a container that contains a composition that includes a soluble LTBR, and optionally (b) informational material.
  • the informational material can be descriptive, instructional, marketing or other material that relates to the methods described herein and/or the use of the agents for therapeutic benefit.
  • the kit also includes a second agent for treating an autoimmune disorder, such as a DMARD for treatment of RA.
  • the kit includes a first container that contains a composition that includes the soluble LTBR, and a second container that includes the second agent.
  • the informational material of the kits is not limited in its form.
  • the informational material can include information about production of the compound, molecular weight of the compound, concentration, date of expiration, batch or production site information, and so forth.
  • the informational material relates to methods of administering the soluble LTBR (e.g., LTBR-Fc), e.g., in a suitable dose, dosage form, or mode of administration (e.g., a dose, dosage form, or mode of administration described herein), to treat a subject who has an autoimmune disorder, or who is at risk for experiencing an episode associated with an autoimmune disorder.
  • the information can be provided in a variety of formats, including printed text, computer readable material, video recording, or audio recording, or information that provides a link or address to substantive material.
  • the composition in the kit can include other ingredients, such as a solvent or buffer, a stabilizer, or a preservative.
  • the agent can be provided in any form, e.g., liquid, dried or lyophilized form, preferably substantially pure and/or sterile.
  • the liquid solution preferably is an aqueous solution.
  • the agents are provided as a dried form, reconstitution generally is by the addition of a suitable solvent.
  • the solvent e.g., sterile water or buffer, can optionally be provided in the kit.
  • the kit can include one or more containers for the composition or compositions containing the agents.
  • the kit contains separate containers, dividers or compartments for the composition and informational material.
  • the composition can be contained in a bottle, vial, or syringe, and the informational material can be contained in a plastic sleeve or packet.
  • the separate elements of the kit are contained within a single, undivided container.
  • the composition is contained in a bottle, vial or syringe that has attached thereto the informational material in the form of a label.
  • the kit includes a plurality (e.g., a pack) of individual containers, each containing one or more unit dosage forms (e.g., a dosage form described herein) of the agents.
  • the containers can include a combination unit dosage, e.g., a unit that includes both the soluble LTBR and the second agent, e.g., in a desired ratio.
  • the kit includes a plurality of syringes, ampules, foil packets, blister packs, or medical devices, e.g., each containing a single combination unit dose.
  • the containers of the kits can be air tight, waterproof (e.g., impermeable to changes in moisture or evaporation), and/or light-tight.
  • the kit optionally includes a device suitable for administration of the composition, e.g., a syringe or other suitable delivery device.
  • the device can be provided pre-loaded with one or both of the agents or can be empty, but suitable for loading.
  • Example 1 Construction and characterization of LTBR-IgG constructs to reduce heterogeneity
  • LTBR LTBR
  • NN neural networks
  • HMM hidden Markov models
  • LTBRO 1 (LTBR-IgG fusion protein version 01 ) was the initial molecule that was cloned.
  • the N-terminus of LTBROl started at S28 based on intact LTBR numbering, i.e., including signal sequence.
  • To the LTBR C-termini at M225 was fused the hinge Fc of an IgGl starting at C220 (Kabat numbering) of an IgG heavy chain.
  • a mutation of the antibody derived cysteine (C220) of the IgGl hinge to the structurally similar amino acid valine (V 199 in LTBROl) was included LTBRIgG.
  • LTBR05 was constructed with a C-terminal deletion mutant to avoid having a C-terminal lysine that might undergo proteolysis.
  • MS mass spectrophotometry
  • LTBR06 was constructed. LTBR06 was designed to have four amino acids deleted from the N- terminus. This deletion resulted in dramatically decreased N-terminal sequence variability, as described in Table 1. LTBR06 had more than 90% of the desired N- termini (N-4 and N-5 relative to wild type) and N-I N-termini. The deletions of LTB R06 are also described in Figures 1 and 3.
  • Table 3 provides an overview of the different constructs which were created by deleting portions of the N and C termini of LTBR, as well as the results from such deletions.
  • LTBR05 had a single amino acid removed from the carboxy terminus of LTBROl (C-I)
  • LTBR06 had the single C-I amino acid deletion as well as 4 amino acids deleted from the amino terminus (following the signal sequence - termed N-4).
  • N-4 the signal sequence - termed N-4
  • LTBR05 was biochemically characterized, including the N-terminal heterogeneity (results summarized in Table 4). The amino-terminus of LTBR05 was found to exist as N, N-I, and N-3 with the N-I and N-3 forms converted predominantly to pyroglutamate ( ⁇ Q).
  • LTBR06 was biochemically characterized to determine whether heterogeneity was improved, including whether the N- and C- terminal heterogeneity was improved based on the amino acid substitutions (results summarized in Table 5).
  • LTBR09 was a construct based on LTB R06, with an additional N275Q mutation (amino acid position refers to mature protein - for comparison of sequence and changes between various constructs see Figure 2).
  • Table 6 describes the glycosylation occupancy for each of three LTBR constructs at Asnl3 and Asnl50.
  • glycosylation occupancy was relatively invariant within the different constructs for Asnl3 and Asnl50.
  • glycosylation mutants were explored in which the N-terminal glycosylation of LTBRIgG was eliminated in either the LTBR extracellular domain or Fc portion of the protein. Elimination of glycosylation sites in the LTBR domain successfully reduced glycosylation and also decreased expression of this protein about ten fold.
  • Table 7 provides an overview of the various mutants and the results from studies which characterized the effect(s) of the deletion of glycosylation sites.
  • LTBR03 is shown in Figure 5 and SEQ ID NO: 10. Aglycosylated
  • LTBR03 showed binding equivalency to wildtype LTBRIgG (unmodified LTBR extracellular domain and unmodified Fc region) in a competitive FACS assay.
  • Soluble aglycosylated LTBRIgG was also generated by first capturing the protein using Protein A (primary capture). The captured protein was then run over a phenyl column to remove misfolded and aggregated proteins. Following phenyl chromatography, agly LTBRIgG was purified into a homogenous population using sephacryl S-200. Soluble LTBR was then generated using Asp N digestion, where the digest was subsequently re-exposed to Protein A to remove the Fc portion of the fusion protein. The sLTBR was then purified using DEAE (which removed the Asp N) and gel filtration / characterization, the latter of which is an optional polishing step. Finally, a primary crystal screen was performed.
  • Example 3 Sialylation effect on LTBRIgG pharmacokinetics [00260]
  • sialyation of LTBRIgG was forced using ⁇ -2,6-Sialyltransferase from rat liver (Boehringer Mannheim).
  • ⁇ -2,6-Sialytransferase is specific for rat liver (Boehringer Mannheim).
  • Figure 13 describes the difference in pharmacokinetics between huLTBRIgG and LFA3TIP (dimeric fusion protein that consists of the extracellular CD2-binding portion of the human leukocyte function antigen-3 (LF A-3) linked to the Fc (hinge, CH2 and CH3 domains) portion of human IgGl).
  • LTBRIgG had increased serum levels regardless of sialylation in comparison to LFA3TIP.
  • the TNFRIgG product entanercept has significant misfolding heterogenities within the protein, a situation similar to LTBRIgG.
  • a truncated version of TNFRFc was created using 2.6 domains of TNFR.
  • the truncated TNF receptor molecule retained full binding activity to the TNFa ligand.
  • Binding affinities of the various truncations (including D 3-1, D 2-1
  • LTBR06 hinge VDKTHTCPPCPAP (SEQ ID NO: 13)
  • Hinge 2211 (D 169N): VNKTHTCPPCPAP (SEQ ID NO: 14) Hinge 2212 (Tl 98N): VDKNHTCPPCPAP (SEQ ID NO: 15) Hinge 2221 (valine deletion): DKTHTCPPCPAP (SEQ ID NO: 16) Hinge 2217 (full length): EPKSCDKTHTCPPCPAP (SEQ ID NO: 17) Hinge 2219 (G4-hinge-G4Fc): ESCDKYGPPCPPCPAP (SEQ ID NO: 18) Hinge 2218 (G2-hinge-G2Fc): CCVECPPCP APPV AGP (SEQ ID NO: 19) Hinge 2220 (short hinge - GlFc): CPPCPAP (SEQ ID NO: 20)
  • LTBRIgG (more specifically LTBR06) for the treatment of an autoimmune disorder.
  • Example 6 Low doses of LTBR-Fc are effective to treat rheumatoid arthritis.
  • a dose of 0.05 mg/kg LTBR-Fc (version LTBR06) was chosen as the lowest of five dose cohorts in randomized, blinded, placebo-controlled, drug escalating clinical study to evaluate the safety, tolerability and pharmacokinetics of multiple doses of LTBR-Fc in RA patients.
  • 0.05 mg/kg was chosen as the lowest dose cohort because it was expected to be a no-effect dose with regard to safety and pharmacokinetic findings. Indeed this was a no-effect dose with regard to acute phase response in earlier studies. Assessing efficacy was not a primary or secondary objective of the study. Dosing was performed once weekly for 4 weeks. All members of the cohort were DMARD-IR. The patients received methotrexate therapy while receiving the LTBR-Fc of the study.
  • TJC Tender joint counts
  • SJC swollen joint counts
  • the study was a blinded, randomized, placebo-controlled, dose-ranging study in 47 DMARD-IR RA patients. Screening for the study included identifying patients with active RA who were methotrexate (mtx)-IR. Cohort doses included the following groups: 0.01 mg/kg, 0.05 mg/kg, 0.1 mg/kg, 0.3 mg/kg, 1.0 mg/kg or 3.0mg/kg.
  • LTBR-Fc was administered in combination with mtx. The randomized to LTBR-Fc or placebo ratio was 3:1. Subcutaneous injection once weekly for 4 weeks followed by 8 weeks of observation. Baseline demographics and disease characteristics were similar throughout the placebo and experimental groups (11 placebo, 36 LTB R06 patients).
  • the average duration of RA including all of the patients was 3.1 years. 39 of the patients (83%) had had prior DMARD treatment, and 3 of 6% had had prior anti-TNF treatment.
  • the median dose of methotrexate (mtx) at baseline was 15 mg/week.
  • the median of ACR core set of measurements was 16 SJC, 21 TJC, and 1.33 HAQ.
  • LTBR-Fc proved effective at treating RA in patients. The results showed substantial improvement in the majority of ACR core set measurements in a dose dependent manner.
  • Table 10 ACR20/50/70 response rates (% of patients) at Day 77
  • ACR20 (ACR50, ACR70) response is defined as a 20% (50%, 70%) improvement in SJC and TJC, with a 20% (50%, 70%) improvement in at least 3 of the following indices: IGA, PGA, pain- VAS, HAQ, CRP (ESR if CRP is missing).
  • LTBR-Fc proved safe in patients who received treatment. 55% of patients (6/11) receiving placebo and 67% of patients (24/36) receiving LTBR-Fc (LTB R06) experienced adverse events (AEs) (e.g., headaches, chills, fatigue) although none were severe.
  • AEs adverse events
  • LTBR-Fc LTBR06
  • LTBR06 LTBR-Fc
  • No drug-related serious infections or drug-related serious AEs were reported during the study period.
  • One serious infection was reported in the 3.0 mg/kg group on Day 91 (acute bronchitis, unlikely related to treatment).
  • Transient mild-to- moderate flu-like symptoms were observed in 25% of patients within 24 h after the first dose of LTBR-Fc. These symptoms responded well to acetaminophen and did not usually recur after subsequent doses (decreased on subsequent doses - reported in 8%, 9% and 6% of patients after the second, third, and fourth doses, respectively).
  • LTBR-Fc was effective at treating RA in doses ranging from 0.01 mg/kg to 3 mg/kg.
  • a long term extension study is used to further evaluate the safety, tolerability, and efficacy of LTBR-Fc (LTBR06) in subjects with RA.
  • Patients from previous studies, such as the study described in Example 7 may be included in the study.
  • the study design includes administering LTBR-Fc (LTBR06) to RA patients in one of the following doses subcutaneously: 70 mg every other week; 200 mg every other week; 70 mg monthly; 200 mg monthly; or a placebo dose.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Zoology (AREA)
  • Biomedical Technology (AREA)
  • Molecular Biology (AREA)
  • Wood Science & Technology (AREA)
  • Rheumatology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Plant Pathology (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Physics & Mathematics (AREA)
  • Pain & Pain Management (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Microbiology (AREA)
  • Toxicology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention concerne des méthodes de traitement d'une maladie à l'aide d'inhibiteurs solubles de la voie de la lymphotoxine présentant des propriétés améliorées. L'invention concerne également des protéines de fusion LTBR-Ig améliorées et des compositions pharmaceutiques associées.
EP08742126A 2007-03-15 2008-03-17 Traitement de troubles auto-immuns Withdrawn EP2139509A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US91851807P 2007-03-15 2007-03-15
PCT/US2008/003548 WO2008112325A2 (fr) 2007-03-15 2008-03-17 Traitement de troubles auto-immuns

Publications (1)

Publication Number Publication Date
EP2139509A2 true EP2139509A2 (fr) 2010-01-06

Family

ID=39630536

Family Applications (1)

Application Number Title Priority Date Filing Date
EP08742126A Withdrawn EP2139509A2 (fr) 2007-03-15 2008-03-17 Traitement de troubles auto-immuns

Country Status (5)

Country Link
EP (1) EP2139509A2 (fr)
JP (1) JP5431171B2 (fr)
CN (1) CN101951940A (fr)
CA (1) CA2680792A1 (fr)
WO (1) WO2008112325A2 (fr)

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2007311052B2 (en) * 2006-10-20 2014-01-16 Biogen Idec Ma Inc. Treatment of demyelinating disorders with soluble lymphotoxin-beta-receptor
MX2010009398A (es) * 2008-02-29 2010-11-12 Biogen Idec Inc Proteinas de fusion de inmunoglobulinas purificadas y los metodos para su purificacion.
AU2009298708A1 (en) * 2008-09-30 2010-04-08 Genentech, Inc. Biological markers predictive of rheumatoid arthritis response to lymphotoxin antagonists
EP2382228B1 (fr) 2009-01-21 2020-08-26 Amgen Inc. Compositions et procédés de traitement de maladies inflammatoires et auto-immunes
EP2638073A4 (fr) * 2010-11-09 2014-05-07 Altimab Therapeutics Inc Complexes protéiques pour la liaison à un antigène, et leurs procédés d'utilisation
US9580486B2 (en) 2013-03-14 2017-02-28 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
MY188430A (en) 2015-04-10 2021-12-08 Amgen Inc Interleukin-2 muteins for the expansion of t-regulatory cells
EP3900716A1 (fr) * 2020-04-21 2021-10-27 Dompe' Farmaceutici S.P.A. Co-cristal de gabapentine, de kétoprofène et de lysine, compositions pharmaceutiques et leur utilisation médicale

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008049053A2 (fr) * 2006-10-20 2008-04-24 Biogen Idec Ma Inc. Traitement de troubles de la démyélinisation
WO2008143954A2 (fr) * 2007-05-14 2008-11-27 Biogen Idec Ma Inc. Régions fc simple chaîne, polypeptides de liaison comportant de telles régions, et procédés correspondants
WO2009111347A1 (fr) * 2008-02-29 2009-09-11 Biogen Idec Ma Inc. Protéines hybrides purifiées d’immunoglobuline et leurs procédés de purification

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5925351A (en) * 1995-07-21 1999-07-20 Biogen, Inc. Soluble lymphotoxin-β receptors and anti-lymphotoxin receptor and ligand antibodies as therapeutic agents for the treatment of immunological disease
US6013631A (en) * 1998-06-19 2000-01-11 Xoma Corporation Bactericidal/permeability-increasing protein (BPI) deletion analogs
TR200504220T2 (tr) * 1998-12-17 2007-04-24 Biogen Idec Ma Inc. Aktif limfotoksin-beta reseptör imunoglobülin şimeAktif limfotoksin-beta reseptör imunoglobülin şimerik proteinlerinin yüksek düzey ifadesi ve saflaştrik proteinlerinin yüksek düzey ifadesi ve saflaştırılması için bir yöntem.ırılması için bir yöntem.
US7700317B2 (en) * 2003-03-28 2010-04-20 Biogen Idec Ma Inc. Truncated baff receptors
EA008831B1 (ru) * 2003-06-12 2007-08-31 Эли Лилли Энд Компани Слитые белки аналогов glp-1
CA2528551A1 (fr) * 2003-06-13 2005-01-13 Biogen Idec Ma Inc. Anticorps d'aglycosyle anti-cd154 (ligand cd40) et utilisations correspondantes
CN1871259A (zh) * 2003-08-22 2006-11-29 比奥根艾迪克Ma公司 具有改变的效应物功能的经改进的抗体和制备它的方法
NZ552264A (en) * 2004-07-20 2010-03-26 Symphogen As A procedure for structural characterization of a recombinant polyclonal protein or a polyclonal cell line

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008049053A2 (fr) * 2006-10-20 2008-04-24 Biogen Idec Ma Inc. Traitement de troubles de la démyélinisation
WO2008143954A2 (fr) * 2007-05-14 2008-11-27 Biogen Idec Ma Inc. Régions fc simple chaîne, polypeptides de liaison comportant de telles régions, et procédés correspondants
WO2009111347A1 (fr) * 2008-02-29 2009-09-11 Biogen Idec Ma Inc. Protéines hybrides purifiées d’immunoglobuline et leurs procédés de purification

Also Published As

Publication number Publication date
CN101951940A (zh) 2011-01-19
CA2680792A1 (fr) 2008-09-18
WO2008112325A3 (fr) 2009-12-23
WO2008112325A2 (fr) 2008-09-18
JP2010521472A (ja) 2010-06-24
JP5431171B2 (ja) 2014-03-05

Similar Documents

Publication Publication Date Title
KR101378194B1 (ko) 안정한 단백질 제제
EP2260054B1 (fr) Protéines hybrides purifiées d immunoglobuline et leurs procédés de purification
RU2614257C2 (ru) СТАБИЛЬНЫЕ ЖИДКИЕ ФАРМАЦЕВТИЧЕСКИЕ ПРЕПАРАТЫ СЛИТОГО БЕЛКА TNFR:Fc
EP2139509A2 (fr) Traitement de troubles auto-immuns
AU2001273174B2 (en) Methods for treating rheumatic diseases using a soluble CTLA4 molecule
US9649383B2 (en) Liquid formulations for TNFR:Fc fusion proteins
US8722615B2 (en) Compositions and methods for increasing serum half-life
US20130259861A1 (en) Treatment of autoimmune disorders
US11717558B2 (en) Nucleic acid molecules encoding activin receptor type IIA variants
US20230277672A1 (en) METHODS OF USING CTLA4Ig SUBCUTANEOUS FORMULATIONS
US20180280474A1 (en) Treatment of bile acid disorders
KR20240053675A (ko) sBCMA 변이체 및 이의 FC 융합 단백질을 이용한 IgA, IgM 및/또는 IgG의 생산 감소 방법
CN117915937A (zh) 使用TACI-Fc融合免疫调节蛋白的给药和治疗方法
AU2013204512A1 (en) Purified Immunoglobulin Fusion Proteins and Methods of Their Purification

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20091015

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA MK RS

R17D Deferred search report published (corrected)

Effective date: 20091223

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 14/715 20060101AFI20091230BHEP

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1139959

Country of ref document: HK

17Q First examination report despatched

Effective date: 20101220

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: BIOGEN MA INC.

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20150825

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

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

18D Application deemed to be withdrawn

Effective date: 20160105

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1139959

Country of ref document: HK