AU758240B2 - Enhancing the circulating half-life of antibody-based fusion proteins - Google Patents

Enhancing the circulating half-life of antibody-based fusion proteins Download PDF

Info

Publication number
AU758240B2
AU758240B2 AU27842/99A AU2784299A AU758240B2 AU 758240 B2 AU758240 B2 AU 758240B2 AU 27842/99 A AU27842/99 A AU 27842/99A AU 2784299 A AU2784299 A AU 2784299A AU 758240 B2 AU758240 B2 AU 758240B2
Authority
AU
Australia
Prior art keywords
xaa xaa
antibody
protein
fusion protein
based fusion
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.)
Ceased
Application number
AU27842/99A
Other versions
AU2784299A (en
Inventor
Stephen D. Gillies
Yan Lan
Kin-Ming Lo
John Wesolowski
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.)
Merck Patent GmbH
Original Assignee
EMD Serono Research Center 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 EMD Serono Research Center Inc filed Critical EMD Serono Research Center Inc
Publication of AU2784299A publication Critical patent/AU2784299A/en
Application granted granted Critical
Publication of AU758240B2 publication Critical patent/AU758240B2/en
Assigned to MERCK PATENT GMBH reassignment MERCK PATENT GMBH Alteration of Name(s) in Register under S187 Assignors: LEXIGEN PHARMACEUTICALS CORPORATION
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/55IL-2
    • 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
    • C07K14/70503Immunoglobulin superfamily
    • C07K14/70514CD4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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

Description

WO 99/43713 PCT/US99/03966 ENHANCING THE CIRCULATING HALF-LIFE OF ANTIBODY-BASED FUSION PROTEINS Cross Reference to Related Application This incorporates by reference, and claims priority to and the benefit of, U.S. Provisional Patent Application Serial Number 60/075,887 which was filed on February 25, 1998.
Field of the Invention The present invention relates generally to fusion proteins. More specifically, the present invention relates to methods of enhancing the circulating half-life of antibody-based fusion proteins.
Background of the Invention The use of antibodies for treatment human disease is well established and has become more sophisticated with the introduction of genetic engineering. Several techniques have been developed to improve the utility of antibodies. These include: the generation of monoclonal antibodies by cell fusion to create "hyridomas", or by molecular cloning of antibody heavy (H) and light chains from antibody-producing cells; the conjugation of other molecules to antibodies to deliver them to preferred sites in vivo, radioisotopes, toxic drugs, protein toxins, and cytokines; the manipulation of antibody effector functions to enhance or diminish biological activity; the joining of other protein such as toxins and cytokines with antibodies at the genetic level to produce antibody-based fusion proteins; and the joining of one or more sets of antibody combining regions at the genetic level to produce bi-specific antibodies.
When proteins are joined together through either chemical or genetic manipulation, it is often difficult to predict what properties that the end product will retain from the parent molecules. With chemical conjugation, the joining process may occur at different sites on the molecules, and generally results in molecules with varying degrees of modification that can affect the function of one or both proteins. The use of genetic fusions, on the other hand, makes the joining process more consistent, and results in the production of consistent end products that retain the function of both component proteins. See, for example, Gillies et al., PROC. NATL.
ACAD. SCi. USA 89:1428-1432 (1992); and U.S. Patent No. 5,650,150.
SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 -2- However, the utility of recombinantly-produced antibody-based fusion proteins may be limited by their rapid in vivo clearance from the circulation. Antibody-cytokine fusion proteins, for example, have been shown to have a significantly lower in vivo circulating half-life than the free antibody. When testing a variety of antibody-cytokine fusion proteins, Gillies et al. reported that all of the fusion proteins tested had an ct phase (distribution phase) half-life of less than hour. Indeed, most of the antibody-based fusion protein were cleared to 10% of the serum concentration of the free antibody by two hours. See, Gillies et al., BIOCONJ. CHEM. 4: 230-235 (1993). Therefore, there is a need in the art for methods of enhancing the in vivo circulating halflife of antibody-based fusion proteins.
Summary of the Invention A novel approach to enhancing the in vivo circulating half-life of antibody-based fusion proteins has now been discovered. Specifically, the present invention provides methods for the production of fusion proteins between an immunoglobulin with a reduced binding affinity for an Fc receptor, and a second non-immunoglobulin protein. Antibody-based fusion proteins with reduced binding affinity for Fc receptors have a significantly longer in vivo circulating half-life than the unlinked second non-immunoglobulin protein.
IgG molecules interact with three classes of Fc receptors (FcR) specific for the IgG class of antibody, namely FcyRI, FcyRII and FcyRIII. In preferred embodiments, the immunoglobulin (Ig) component of the fusion protein has at least a portion of the constant region of an IgG that has a reduced binding affinity for at least one of FcyRI, FcyRII or FcyRIII.
In one aspect of the invention, the binding affinity of fusion proteins for Fc receptors is reduced by using heavy chain isotypes as fusion partners that have reduced binding affinity for Fc receptors on cells. For example, both human IgGI and IgG3 have been reported to bind to FcRyI with high affinity, while IgG4 binds 10-fold less well, and IgG2 does not bind at all. The important sequences for the binding of IgG to the Fc receptors have been reported to be located in the CH2 domain. Thus, in a preferred embodiment, an antibody-based fusion protein with enhanced in vivo circulating half-life is obtained by linking at least the CH2 domain of IgG2 or IgG4 to a second non-immunoglobulin protein.
In another aspect of the invention, the binding affinity of fusion proteins for Fc receptors is reduced by introducing a genetic modification of one or more amino acid in the constant SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 -3region of the IgG 1 or IgG3 heavy chains that reduces the binding affinity of these isotypes for Fc receptors. Such modifications include alterations of residues necessary for contacting Fc receptors or altering others that affect the contacts between other heavy chain residues and Fc receptors through induced conformational changes. Thus, in a preferred embodiment, an antibody-based fusion protein with enhanced in vivo circulating half-life is obtained by first introducing a mutation, deletion, or insertion in the IgGI constant region at one or more amino acid selected from Leu2 34 Leu 235 Gly 236 Gly 237 Asn2 97 and Pro33, and then linking the resulting immunoglobulin, or portion thereof, to a second non-immunoglobulin protein. In an alternative preferred embodiment, the mutation, deletion, or insertion is introduced in the IgG3 constant region at one or more amino acid selected from Leu 28 1 Leu, 8 2 Gly 28 3 Gly 284 Asn 344 and Pro 37 8 and the resulting immunoglobulin, or portion thereof, is linked to a second non-immunoglobulin protein. The resulting antibody-based fusion proteins have a longer in vivo circulating half-life than the unlinked second non-immunoglobulin protein.
In a preferred embodiment, the second non-immunoglobulin component of the fusion protein is a cytokine. The term "cytokine" is used herein to describe proteins, analogs thereof, and fragments thereof which are produced and excreted by a cell, and which elicit a specific response in a cell which has a receptor for that cytokine. Preferably, cytokines include interleukins such as interleukin-2 hematopoietic factors such as granulocyte-macrophage colony stimulating factor (GM-CSF), tumor necrosis factor (TNF) such as TNFa, and lymphokines such as lymphotoxin. Preferably, the antibody-cytokine fusion protein of the present invention displays cytokine biological activity.
In an alternative preferred embodiment, the second non-immunoglobulin component of the fusion protein is a ligand-binding protein with biological activity. Such ligand-binding proteins may, for example, block receptor-ligand interactions at the cell surface; or neutralize the biological activity of a molecule a cytokine) in the fluid phase of the blood, thereby preventing it from reaching its cellular target. Preferably, ligand-binding proteins include CD4, CTLA-4, TNF receptors, or interleukin receptors such as the IL-1 and IL-4 receptors. Preferably, the antibody-receptor fusion protein of the present invention displays the biological activity of the ligand-binding protein.
SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 -4- In yet another alternative preferred embodiment, the second non-immunoglobulin component of the fusion protein is a protein toxin. Preferably, the antibody-toxin fusion protein of the present invention displays the toxicity activity of the protein toxin.
In a preferred embodiment, the antibody-based fusion protein comprises a variable region specific for a target antigen and a constant region linked through a peptide bond to a second nonimmunoglobulin protein. The constant region may be the constant region normally associated with the variable region, or a different one, variable and constant regions from different species. The heavy chain can include a CH1, CH2, and/or CH3 domains. Also embraced within the term "fusion protein" are constructs having a binding domain comprising framework regions and variable regions complementarity determining regions) from different species, such as are disclosed by Winter, et al., GB 2,188, 638. Antibody-based fusion proteins comprising a variable region preferably display antigen-binding specificity. In yet another preferred embodiment, the antibody-based fusion protein further comprises a light chain. The invention thus provides fusion proteins in which the antigen-binding specificity and activity of an antibody are combined with the potent biological activity of a second non-immunoglobulin protein, such as a cytokine. A fusion protein of the present invention can be used to deliver selectively the second non-immunoglobulin protein to a target cell in vivo so that the second nonimmunoglobulin protein can exert a localized biological effect.
In an alternative preferred embodiment, the antibody-based fusion protein comprises a heavy chain constant region linked through a peptide bond to a second non-immunoglobulin protein, but does not comprise a heavy chain variable region. The invention thus further provides fusion proteins which retain the potent biological activity of a second nonimmunoglobulin protein, but which lack the antigen-binding specificity and activity of an antibody.
In preferred embodiments, the antibody-based fusion proteins of the present invention further comprise sequences necessary for binding to Fc protection receptors (FcRp), such as beta-2 microglobulin-containing neonatal intestinal transport receptor (FcRn).
In preferred embodiments, the fusion protein comprises two chimeric chains comprising at least a portion of a heavy chain and a second, non-Ig protein are linked by a disulfide bond.
SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 The invention also features DNA constructs encoding the above-described fusion proteins, and cell lines, myelomas, transfected with these constructs.
These and other objects, along with advantages and features of the invention disclosed herein, will be made more apparent from the description, drawings, and claims that follow.
Brief Description of the Drawings The foregoing and other objects, features, and advantages of the present invention, as well as the invention itself, may be more fully understood from the following description of preferred embodiments, when read together with the accompanying drawings, in which: FIG. 1 is a homology alignment of the amino acid sequences of the constant region of Cyl and Cy3, aligned to maximize amino acid identity, and wherein non-conserved amino acids are identified by boxes; FIG. 2 is a homology alignment of the amino acid sequences of constant region of Cyl, Cy2, and Cy 4, aligned to maximize amino acid identity, and wherein non-conserved amino acids are identified by boxes; FIG. 3 is a diagrammatic representation of a map of the genetic construct encoding an antibody-based fusion protein showing the relevant restriction sites; FIG. 4 is a bar graph depicting the binding of antibody hu-KS-1/4 and antibody-based fusion proteins, hu-KSyl-IL2 and hu-KSy4-IL2, to Fc receptors on mouse J774 cells in the presence (solid bars) or absence (stippled bars) of an excess of mouse IgG; FIG. 5 is a line graph depicting the in vivo plasma concentration of total antibody (free antibody and fusion protein) of hu-KSyl-IL2 (closed diamond) and hu-KSy4-IL2 (closed triangle) and of intact fusion protein of hu-KSyl-IL2 (open diamond) and hu-KSy4-IL2 (open triangle) as a function of time; FIG. 6 is a diagrammatic representation of protocol for constructing an antibody-based fusion protein with a mutation that reduces the binding affinity to Fc receptors; FIG. 7 is a line graph depicting the in vivo plasma concentration of intact fusion protein of hu-KSyl-IL2 mutated hu-KSy -IL2 and hu-KSy4-IL2 as a function of time.
SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 -6- Detailed Description of the Invention It has now been discovered that fusing a second protein, such as a cytokine, to an immunoglobulin may alter the antibody structure, resulting in an increase in binding affinity for one or more of the cell-bound Fc receptors and leading to a rapid clearance of the antibody-based fusion protein from the circulation. The present invention describes antibody-based fusion proteins with enhanced in vivo circulating half-lives and involves producing, through recombinant DNA technology, antibody-based fusion proteins with reduced binding affinity for one or more Fc receptor.
First, an antibody-based fusion protein with an enhanced in vivo circulating half-life can be obtained by constructing a fusion protein with isotypes having reduced binding affinity for a Fc receptor, and avoiding the use of sequences from antibody isotypes that bind to Fc receptors.
For example, of the four known IgG isotypes, IgGI (Cyl) and IgG3 (Cy3) are known to bind FcRyI with high affinity, whereas IgG4 (Cy4) has a 10-fold lower binding affinity, and IgG2 (Cy2) does not bind to FcRyI. Thus, an antibody-based fusion protein with reduced binding affinity for a Fc receptor could be obtained by constructing a fusion protein with a Cy2 constant region (Fc region) or a Cy4 Fc region, and avoiding constructs with a Cyl Fc region or a Cy3 Fc region.
Second, an antibody-based fusion protein with an enhanced in vivo circulating half-life can be obtained by modifying sequences necessary for binding to Fc receptors in isotypes that have binding affinity for an Fc receptor, in order to reduce or eliminate binding. As mentioned above, IgG molecules interact with three classes of Fc receptors (FcR), namely FcyRI, FcyRII, and FcyRIII. Cyl and Cy3 bind FcRyI with high affinity, whereas Cy4 and Cy2 have reduced or no binding affinity for FcRyI. A comparison of the Cyl and Cy3 indicates that, with the exception of an extended hinge segment in Cy3, the amino acid sequence homology between these two isotypes is very high. This is true even in those regions that have been shown to interact with the Clq fragment of complement and the various FcyR classes. FIG. 1 provides a alignment of the amino acid sequences of Cy and Cy3. The other two isotypes of human IgG (Cy2 and Cy4) have sequence differences which have been associated with FcR binding. FIG. 2 provides a alignment of the amino acid sequences of Cyl, Cy2, and Cy4. The important SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 -7sequences for FcyR binding are Leu-Leu-Gly-Gly (residues 234 through 237 in Cyl), located in the CH2 domain adjacent to the hinge. Canfield and Morrison, J. EXP. MED. 173: 1483-1491 (1991). These sequence motifs are conserved in Cyl and Cy3, in agreement with their similar biological properties, and possibly related to the similarity of pharmacokinetic behavior when used to construct IL-2 fusion proteins. Many mutational analyses have been done to demonstrate the effect of specific mutations on FcR binding, including those in residues 234-237 as well as the hinge-proximal bend residue Pro 33 that is substituted by Ser in IgG4. Another important structural component necessary for effective FcR binding is the presence of an N-linked carbohydrate chain covalently bound to Asn 2 97 Enzymatic removal of this structure or mutation of the Asn residue effectively abolish, or at least dramatically reduce, binding to all classes of FcyR.
Brumbell et al. postulated the existence of a protection receptor (FcRp) that would slow the rate of catabolism of circulating antibodies by binding to the Fc portion of antibodies and, following their pinocytosis into cells, would redirect them back into the circulation. Brumbell et al., NATURE 203: 1352-1355 (1964). The beta-2 microglobulin-containing neonatal intestinal transport receptor (FcRn) has recently been identified as a FcRp. See, Junghans et al., PROC.
NATL. ACAD. SCI. USA 93: 5512-5516 (1996). The sequences necessary for binding to this receptor are conserved in all four classes of human IgG and are located at the interface between the CH2 and CH3 domains. See, Medesan et al., J. IMMUNOL. 158:2211-2217 (1997). These sequences have been reported to be important for the in vivo circulating half-life of antibodies.
See, International PCT publication WO 97/34631. Thus, preferred antibody-based fusion proteins of the present invention will have the sequences necessary for binding to FcRp.
Methods for synthesizing useful embodiments of the invention are described, as well as assays useful for testing their pharmacokinetic activities, both in vitro and in pre-clinical in vivo animal models. The preferred gene construct encoding a chimeric chain includes, in 5' to 3' orientation, a DNA segment which encodes at least a portion of an immunoglobulin and DNA which encodes a second, non-immunoglobulin protein. An alternative preferred gene construct includes, in 5' to 3' orientation, a DNA segment which encodes a second, non-immunoglobulin protein and DNA which encodes at least a portion of an immunoglobulin. The fused gene is assembled in or inserted into an expression vector for transfection of the appropriate recipient cells where it is expressed.
SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 -8- The invention is illustrated further by the following non-limiting examples: Example 1 Improving the in vivo circulating half-life of an antibody-IL2 fusion protein by class switching from Cyl to Cy4 IgG constant regions.
According to the present invention, antibody-based fusion proteins with enhanced in vivo circulating half-lives can be obtained by constructing antibody-based fusion proteins using sequences from antibody isotypes that have reduced or no binding affinity for Fc receptors.
In order to assess whether the in vivo circulating half-life of the antibody-based fusion protein can be enhanced by using sequences from antibody isotypes with reduced or no binding affinity for Fc receptors, an antibody-IL2 fusion protein with a human Cyl constant region (Fc region) was compared to an antibody-IL2 fusion protein with a human Cy4 Fc region.
1.1 Construction of antibody-IL2 fusion proteins with a Cy4 IgG constant region.
The construction of antibody-IL2 fusion proteins with a Cyl constant region has been described in the prior art. See, for example, Gillies et al., PROC. NATL. ACAD. SCL. USA 89: 1428-1432 (1992); and U.S. Patent No 5,650,150, the disclosure of which is incorporated herein by reference.
To construct antibody-IL2 fusion proteins with a Cy4 constant region, a plasmid vector, capable of expressing a humanized antibody-IL2 fusion protein with variable regions specific for a human pancarcinoma antigen (KSA) and the human Cyl heavy chain fused to human IL-2, was modified by removing the Cyl gene fragment and replacing it with the corresponding sequence from the human Cy4 gene. A map of some of the relevant restriction sites and the site of insertion of the Cy4 gene fragment is provided in FIG. 3. These plasmid constructs contain the cytomegalovirus (CMV) early promoter for transcription of the mRNA encoding the light (L) and heavy chain variable regions derived from the mouse antibody KS-1/4. The mouse V regions were humanized by standard methods and their encoding DNA sequences were chemically synthesized. A functional splice donor site was added at the end of each V region so that it could be used in vectors containing H and L chain constant region genes. The human CK light chain gene was inserted downstream of the cloning site for the VL gene and was followed by its endogenous 3' untranslated region and poly adenylation site. This transcription unit was followed by a second independent transcription unit for the heavy chain-IL2 fusion protein. It is SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 -9also driven by a CMV promoter. The VH encoding sequence was inserted upstream of the DNA encoding the Cy heavy chain gene of choice, fused to human IL-2 encoding sequences. Such Cy genes contain splice acceptor sites for the first heavy chain exon (CHI), just downstream from a unique Hind III common to all human Cy genes. A 3' untranslated and polyadenylation site from SV40 virus was inserted at the end of the IL-2 encoding sequence. The remainder of the vector contained bacterial plasmid DNA necessary for propagation in E. coli and a selectable marker gene (dihydrofolate reductase dhfr) for selection of transfectants of mammalian cells.
The swapping of the Cyl and Cy4 fragments was accomplished by digesting the original Cyl-containing plasmid DNA with Hind III and Xho I and purifying the large 7.8 kb fragment by agarose gel electrophoresis. A second plasmid DNA containing the Cy4 gene was digested with Hind III and Nsi I and the 1.75 kb fragment was purified. A third plasmid containing the human IL-2 cDNA and SV40 poly A site, fused to the carboxyl terminus of the human Cy1 gene, was digested with Xho I and Nsi I and the small 470 bp fragment was purified. All three fragments were ligated together in roughly equal molar amounts and the ligation product was used to transform competent E. coli. The ligation product was used to transform competent E. coli and colonies were selected by growth on plates containing ampicillin. Correctly assembled recombinant plasmids were identified by restriction analyses of plasmid DNA preparations from isolated transformants and digestion with Fsp I was used to discriminate between the Cyl (no Fsp I) and Cy4 (one site) gene inserts. The final vector, containing the Cy4-IL2 heavy chain replacement, was introduced into mouse myeloma cells and transfectants were selected by growth in medium containing methotrexate (0.1 gM). Cell clones expressing high levels of the antibody-IL2 fusion protein were expanded and the fusion protein was purified from culture supernatants using protein A Sepharose chromatography. The purity and integrity of the Cy4 fusion protein was determined by SDS-polyacrylamide gel electrophoresis. IL-2 activity was measured in a T-cell proliferation assay and found to be identical to that of the Cy construct.
1.2 Binding to Fc receptors by antibody and antibody-IL2 fusion proteins with Cyl and Cy4 IgG constant region.
Various mouse and human cell lines express one or more Fc receptor. For example, the mouse J774 macrophage-like cell line expresses FcRyI that is capable of binding mouse or human IgG of the appropriate subclasses. Likewise, the human K562 erythroleukemic cell line SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 expresses FcRyII but not FcRyI. In order to assess the potential contribution of Fc receptor binding to clearance of antibody-based fusion proteins from the circulation, the binding affinities of an antibody, a Cyl-IL2 fusion protein, and a Cy4-IL2 fusion protein for FcRyI were compared in the mouse J774 cell line.
The two antibody-IL-2 fusion proteins described in Example 1, hu-KSyl-IL2 and hu-KSy4-IL2, were diluted to 2 pg/ml in PBS containing 0.1% bovine serum albumin (BSA), together with 2x10 5 J774 cells in a final volume of 0.2 ml. After incubation on ice for 20 min, a FITC-conjugated anti-human IgG Fc antibody (Fab2) was added and incubation was continued for an additional 30 min. Unbound antibodies were removed by two washes with PBS-BSA, and the cells were analyzed in a fluorescence-activated cell sorter (FACS). Control reactions contained the same cells mixed with just the FITC-labeled secondary antibody or with the humanized KSyl antibody (without IL-2).
As expected, the binding of the Cy4-IL2 fusion protein to J774 cells was significantly lower than the binding of the Cyl-IL2 fusion protein. See FIG. 4. Unexpectedly, however, both the Cl1-IL2 and Cy4-IL2 fusion proteins had significantly higher binding to J774 cells than the KSyl antibody (without IL-2). This suggests that fusing a second protein, such as a cytokine, to an immunoglobulin may alter the antibody structure, resulting in an increase in binding affinity for one or more of the cell-bound Fc receptors, thereby leading to a rapid clearance from the circulation.
In order to determine whether the greater binding observed with IL-2 fusion proteins was due to the presence of IL-2 receptors or FcRyI receptors on the cells, excess mouse IgG (mIgG) was used to compete the binding at the Fc receptors. As illustrated in FIG. 4, background levels of binding were observed with the antibody and both antibody-IL2 fusion proteins in the presence of a 50-fold molar excess of mIgG. This suggests that the increased signal binding of antibody-IL2 fusion proteins was due to increased binding to the Fc receptor.
Cell lines expressing Fc receptors are useful for testing the binding affinities of candidate fusion proteins to Fc receptors in order to identify antibody-based fusion proteins with enhanced in vivo half lives. Candidate antibody-based fusion proteins can be tested by the above-described methods. Candidate antibody-based fusion proteins with substantially reduced binding affinity SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 11 for an Fc receptor will be identified as antibody-based fusion proteins with enhanced in vivo half lives.
1.3 Measuring the circulating half-life of antibody-IL2 fusion proteins with Cyl and Cy4 IgG constant region.
In order to assess whether using the Fc region of an IgG isotype having reduced affinity for Fc receptors will enhance the in vivo circulating half-life, fusion proteins containing the Cy1 isotype heavy chain hu-KSy 1-IL2) were compared to fusion proteins containing the Cy4 isotype heavy chain hu-KSy4-IL2).
Purified humanized KS-1/4-IL2 fusion proteins containing either the Cyl or Cy4 isotype heavy chain were buffer-exchanged by diafiltration into phosphate buffered saline (PBS) and diluted further to a concentration of-100 Lg/ml. Approximately 20 .tg of the antibody-based fusion protein (0.2 ml) was injected into 6-8 week old Balb/c mice in the tail vein using a slow push. Four mice were injected per group. At various time points, small blood samples were taken by retro-orbital bleeding from anaesthetized animals and collected in tubes containing citrate buffer to prevent clotting. Cells were removed by centrifugation in an Eppendorf high-speed tabletop centrifuge for 5 min. The plasma was removed with a micropipettor and frozen at -70 0 C. The concentration of human antibody determinants in the mouse blood was measured by ELISA. A capture antibody specific for human H and L antibody chains was used for capture of the fusion proteins from the diluted plasma samples. After a two hour incubation in antibody-coated 96-well plates, the unbound material was removed by three washes with ELISA buffer (0.01% Tween 80 in PBS). A second incubation step used either an anti-human Fc antibody (for detection of both antibody and intact fusion protein), or an anti-human IL-2 antibody (for detection of only the intact fusion protein). Both antibodies were conjugated to horse radish peroxidase (HRP). After a one hour incubation, the unbound detecting antibody was removed by washing with ELISA buffer and the amount of bound HPR was determined by incubation with substrate and measuring in a spectrophotometer.
As depicted in FIG. 5, the a phase half-life of the hu-KSy4-IL2 fusion protein was significantly longer than the a phase half-life of the hu-KSyl-IL2 fusion protein. The increased half-life is best exemplified by the significantly higher concentrations of the hu-KSy4-IL2 fusion SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 -12protein (3.3 ig/ml) compared to the hu-KSyl-IL2 fusion protein (60 ng/ml) found in mice after 24 hours.
The hu-KSyl-IL2 protein had a rapid distribution phase followed by a slower catabolic phase, as reported earlier for the chimeric 14.18-IL2 fusion protein. See, Gillies et al., BIOCONJ. CHEM. 4: 230-235 (1993). In the Gillies et al. study, only antibody determinants were measured, so it was not clear if the clearance represented the clearance of the intact fusion protein or the clearance of the antibody component of the fusion protein. In the present Example, samples were assayed using both an antibody-specific ELISA, and a fusion proteinspecific ELISA an ELISA that requires that both the antibody and IL-2 components be physically linked). As illustrated in FIG. 5, in animals injected with the hu-KSyl-IL2 fusion protein, the amount of circulating fusion protein was lower than the total amount of circulating antibody, especially at the 24 hr time point. This suggests that the fusion protein is being proteolytically cleaved in vivo and that the released antibody continues to circulate. Surprisingly, in animals injected with the hu-KSy4-IL2 fusion protein, there was no significant differences between the amount of circulating fusion protein and the total amount of circulating antibody.
This suggests the hu-KSy4-IL2 fusion protein was not being proteolytically cleaved in these animals during the 24 hour period measured.
As discussed above, Cyl and Cy3 have binding affinity for Fc receptors, whereas while Cy4 has reduced binding affinity and Cy2 has no binding affinity for Fc receptors. The present Example described methods for producing antibody-based fusion proteins using the Cy4 Fc region, an IgG isotype having reduced affinity for Fc receptors, and established that such antibody-based fusion proteins have enhanced in vivo circulating half-life. Accordingly, a skilled artisan can use these methods to produce antibody-based fusion proteins with the Cy2 Fc region, instead of the Cy4 Fc region, in order to enhance the circulating half-life of fusion proteins. A Hu-KS-IL2 fusion protein utilizing the human Cy2 region can be constructed using the same restriction fragment replacement and the above-described methods for Cy4-IL2 fusion protein.
and tested using the methods described herein to demonstrate increased circulating half-life.
Antibody-based fusion proteins with the Cy2 Fc region, or any other Fc region having reduced binding affinity or lacking binding affinity for a Fc receptor will have enhanced in vivo SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCTIUS99/03966 -13circulating half-life compared to antibody-based fusion proteins having binding affinity for a Fc receptor.
Example 2 Mutating the human Cyl or C 7 3 gene in antibody-based fusion protein constructs to improve their in vivo circulating half-life.
IgG molecules interact with several molecules in the circulation, including members of the complement system of proteins C q fragment), as well as the three classes of FcR. The important residues for Clq binding are residues Glu 3 1 8 Lys 320 and Lys 322 which are located in the CH2 domains of human heavy chains. Tao et al., J. Exp. MED. 178: 661-667 (1993). In order to discriminate between FcR and C q binding as mechanisms for rapid clearance, we substituted the more drastically altered Cy2 hinge-proximal segment into the Cyl heavy chain. This mutation is expected to affect FcR binding but not complement fixation.
The mutation was achieved by cloning and adapting the small region between the hinge and the beginning of the CH2 exon of the germ line Cyl gene using overlapping polymerase chain reactions (PCR). The PCR primers were designed to substitute the new sequence at the junction of two adjacent PCR fragments spanning a Pst I to Drd I fragment (see FIG. In the first step, two separate PCR reactions with primers 1 and 2 (SEQ ID NOS: 5 and 6, respectively), or primers 3 and 4 (SEQ ID NOS: 7 and 8, respectively), were prepared using the Cyl gene as the template. The cycle conditions for the primary PCR were 35 cycles of: 94°C for 45 sec, annealing at 480C for 45 seconds, and primer extension at 72°C for 45 sec. The products of each PCR reaction were used as template for the second, joining reaction step. One tenth of each primary reaction was mixed together and combined with primers 1 and 4 to amplify only the combined product of the two initial PCR products. The conditions for the secondary PCR were: 94°C for 1 min, annealing at 51 C for 1 min, and primer extension at 720C for 1 min. Joining occurs as a result of the overlapping between the two individual fragments which pairs with the end of the other, following denaturation and annealing. The fragments that form hybrids get extended by the Taq polymerase, and the complete, mutated product was selectively amplified by the priming of the outer primers, as shown in FIG. 6. The final PCR product was cloned in a plasmid vector and its sequence verified by DNA sequence analysis.
The assembly of the mutated gene was done in multiple steps. In the first step, a cloning vector containing the human Cyl gene was digested with Pst I and Xho I to remove the SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 -14non-mutated hinge-CH2-CH3 coding sequences. A Drd I to Xho I fragment encoding part of CH2, all of CH3 and the fused human IL-2 coding sequences was prepared from the Cy1-IL2 vector, described above. A third fragment was prepared from the subcloned PCR product by digestion with Pst I and Drd I. All three fragments were purified by agarose gel electrophoresis and ligated together in a single reaction mixture. The ligation product was used to transform competent E. coli and colonies were selected by growth on plates containing ampicillin.
Correctly assembled recombinant plasmids were identified by restriction analyses of plasmid DNA preparations from isolated transformants and mutated genes were confirmed by DNA sequence analysis. The Hind III to Xho I fragment from the mutated Cyl-IL2 gene was used to reassemble the complete hu-KS antibody-IL2 fusion protein expression vector.
In order to assess the enhancement of the in vivo circulating half-life induced by a mutation of an important amino acid for FcR binding, and to discriminate between FcR and C 1 q binding as mechanisms for rapid clearance, the in vivo plasma concentration of the mutated hu-KSyl-IL2 was compared to the plasma concentration ofhu-KSyl-IL2 at various specified times. As illustrated in FIG. 7, the in vivo clearance rates of the mutated hu-KSyl-IL2 and hu-KSy4-IL2 were significantly lower than the clearance rate of hu-KSyl-IL2. These results suggests that an antibody-based fusion protein with enhanced in vivo circulating half-life can be obtained by modifying sequences necessary for binding to Fc receptors in isotypes that have binding affinity for an Fc receptor. Further, the results suggests that the mechanisms for rapid clearance involve FcR binding rather than Clq binding.
The skilled artisan will understand, from the teachings of the present invention, that several other mutations to the Cyl or Cy3 genes can be introduced in order to reduce binding to FcR and enhance the in vivo circulating half-life of an antibody-based fusion protein. Moreover, mutations can also be introduced into the Cy4 gene in order to further reduce the binding of Cy4 fusion proteins to FcR. For example, additional possible mutations include mutations in the hinge proximal amino acid residues, mutating Pro 3 3 or by mutating the single N-linked glycosylation site in all IgG Fc regions. The latter is located at Asn 29 7 as part of the canonical sequence: Asn-X-Thr/Ser, where the second position can be any amino acid (with the possible exception of Pro), and the third position is either Thr or Ser. A conservative mutation to the amino acid Gin, for example, would have little effect on the protein but would prevent the SUBSTITUTE SHEET (RULE 26) WO 99/43713 PCT/US99/03966 attachment of any carbohydrate side chain. A strategy for mutating this residue might follow the general procedure, just described, for the hinge proximal region. Methods for generating point mutations in cloned DNA sequences are well established in the art and commercial kits are available from several vendors for this purpose.
Example 3 Increasing the circulating half-life of receptor-antibody-based fusion proteins.
Several references have reported that the Fc portion of human IgG can serve as a useful carrier for many ligand-binding proteins, or receptors, with biological activity. Some of these ligand-binding proteins have been fused to the N-terminal of the Fc portion of an Ig, such as CD4, CTLA-4, and TNF receptors. See, for example, Capon et al., NATURE 337: 525-531 (1989); Linsley et al., J. EXP. MED. 174: 561-569 (1991); Wooley et al., J. IMMUNOL. 151: 6602- 6607 (1993). Increasing the circulating half-life of receptor-antibody-based fusion proteins may permit the ligand-binding protein partner the second non-Ig protein) to more effectively block receptor-ligand interactions at the cell surface; or neutralize the biological activity of a molecule a cytokine) in the fluid phase of the blood, thereby preventing it from reaching its cellular target. In order to assess whether reducing the ability of receptor-antibodybased fusion proteins to bind to IgG receptors will enhance their in vivo circulating half-life, receptor-antibody-based fusion proteins with human Cyl Fc regions are compared to antibodybased fusion proteins with human Cy4 Fc regions.
To construct CD4-antibody-based fusion proteins, the ectodomain of the human CD4 cell surface receptor is cloned using PCR from human peripheral blood monocytic cells (PBMC).
The cloned CD4 receptor includes compatible restriction sites and splice donor sites described in Example 1. The expression vector contains a unique Xba I cloning site downstream of the CMV early promoter, and the human Cyl or Cy4 gene downstream of their endogenous Hind III site.
The remainder of the plasmid contains bacterial genetic information for propagation in E. coli, as well as a dhfr selectable marker gene. Ligated DNAs are used to transform competent bacteria and recombinant plasmids are identified from restriction analyses from individual bacterial colonies. Two plasmid DNA constructs are obtained: CD4-Cyl and CD4-Cy4.
The expression plasmids are used to transfect mouse myeloma cells by electroporation and transfectants are selected by growth in culture medium containing methotrexate (0.1 tM).
SUBSTITUTE SHEET (RULE 26) -16- Transfectants expressing the fusion proteins are identified by ELISA analyses and are expanded in culture in order to generate fusion protein for purification by binding to and elution from protein A Sepharose. Purified proteins in chromatography elution buffer are diafiltered into PBS and diluted to a final concentration of 100 pg/ml. Balb/c mice are injected with 0.2 ml (20 Pg) of either the CD4-Cyl or CD4-Cy4 fusion protein and the pharnnacokinetics are tested as described in Example 1.3. The CD4-Cy4 fusion protein has a significantly greater half-life than the CD4-Cyl fusion protein.
Equivalents The invention may be embodied in other specific forms without departing from the spirit 10 or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting on the invention described herein. Scope of the :invention is thus indicated by the appended claims rather than by the foregoing.description, and all changes which come within the meaning and range of equivalency of the claims are intended to be embraced therein.
Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or step or group f integers or steps but not the exclusion of any other integer or step or group of integers or steps.
The reference to any prior art in this specification is not, and should not be taken as, an acknowledgment or any form of suggestion that that prior art forms part of the common general knowledge in Australia.
EDITORIAL NOTE APPLICATION NUMBER 27842/99 The following Sequence Listing pages numbered 1 to 6 are part of the Description.
WO 99/43713 <110> GILLIES, Stephen D LO, Kin-Ming LAN, Yan WESOLOWSKI, John PCTIUS99/03966 Antibody-based Fusion <130> LEX-003PC <140> <141> <150> US 60/075, 887 <151> 1998-02-25 <160> 8 <170> Patentln Ver. <210> 1 <211> 447 <212> PRT <213> Homno sapiens <220> <223> IGG-1 CHAIN C REGION <400> 1 Xaa Xaa Xaa Xaa Xaa Xaa Xa~ 1 5 Xaa Xaa Xaa Xaa Xaa Xaa Xa.
Xaa Xaa Xaa Xaa Xaa Xaa Xa Xaa Xaa Xaa Xaa Xaa Xaa Xa 5 Xaa Xaa Xaa Xaa Xaa Xaa Xa.
70 Xaa Xaa Xaa Xaa Xaa Xaa Xa Xaa Xaa Xaa Xaa Xaa Xaa Xa 100 Xaa Xaa Xaa Xaa Xaa Ala Se 115 Ala Pro Ser Ser Lys Ser Th 130 13 Leu Val Lys Asp Tyr Phe Pr 145 150 Gly Ala Leu Thr Ser Gly Va 165 Ser Gly Les Tyr Ser Leu Se 180 Leu Gly Thr Gin Thr Tyr Il 195 Thr Lys Val Asp Lys Lys ye 210 21 Thr Cys Pro Pro Cys Pro A] 225 230 Phe Leu Phe Pro Pro Lys P~ 245 Pro Glu Val Thr Cys Val V~ ting Half-life of Proteins aXaa Xaa a a a a a a r r 5 0 .1 .5 Xaa Xaa 40 Xaa Xaa Xaa Xaa Thr 120 Ser Glu His Ser Cys 200 Glu Pro Lys Val Xaa 25 Xaa Xaa Xaa Xaa Xaa 105 Lys Gly Pro Thr Val 185 As n Pro Giu Asp Asp Xaa 10 Xaa Xaa Xaa Xaa Xaa 90 Xaa Gly Gly Val Phe 170 Val Val Lys Leu Thr 250 Val aa Xaa Xaa Xaa Xaa Xaa Xaa X~aa Xaa X(aa 75 Xaa Xaa Pro Thr Thr 155 Pro Thr Asn Ser Leu 235 Leu Ser Xaa Xaa Xaa Xaa Xaa Xaa Ser Ala 140 Val Ala Val His Cys 220 Gly Met His Xaa Xaa Xaa Xaa Xaa Xaa Val 125 Ala Ser Val Pro Lys 205 Asp Gly Ile Gli.
Xaa Xaa Xaa Xaa Xaa Xaa 110 Phe Leu Trp Leu Ser 190 Pro Lys Pro Ser 1Asp Xaa X(aa Xa a Xaa Xaa Xaa Pro Gly Asn Gin 175 Ser Ser Thr Ser Arc 255 Pro Xaa Xaa Xaa Xaa Xaa Xaa Leu Cys Ser 160 Ser Ser Asn His Val 240 IThr Glu SUBSTITUTE SHEET (RULE 26) WO 99/43713 WO 9943713PCT/US99/03966 Val1 Thr Val 305 Cys Ser Pro Val1 Gly 385 Asp Trp L ys Lys 290 Leu Lys Lys Ser Lys 370 Gin Gly Gin Phe 275 Pro Thr Val1 Ala Arg 355 Gi y Pro Ser Gin 260 Asn Arg Val1 Ser Lys 340 Asp Phe Glu Phe Gly 420 Trp Tyr Val Glu Leu Asn 325 Gly Glu Tyr Asn Phe 405 Asn Glu His 310 Lys Gin Leu Pro As n 390 Leu Val Gin 295 Gin Al a Pro Thr Ser 375 Tyr Tyr Phe 265 Asp Gly 280 Tyr Asn Asp Trp Leu Pro Arg Glu 345 Lys Asn 360 Asp Ile Lys Thr Ser Lys Ser Cys 425 Val Ser Leu Al a 330 Pro Gin Ala Thr Leu 410 Ser Giu Thr Asn 315 Pro Gin Vai Val Pro 395 Thr Val1 Val1 Tyr 300 Gly Ile Val Ser Glu 380 Pro Val1 Met His 285 Arg Lys Glu Tyr Leu 365 T rp Vai Asp His 270 Asn Val1 Glu Lys Thr 350 Thr Glu Leu Lys Glu 430 Al a Val Tyr Thr 335 Leu Cys Ser Asp Ser 415 Ala Lys Ser Lys 320 Ile Pro Leu Asn Ser 400 Ar g Leu His Asn His 435 Tyr Thr Gin Lys Ser 440 Leu Ser Leu Ser Pro Gly Lys 445 <210> 2 <211> 443 <212> PRT <213> Homo sapiens <220> <223> IGG-2 CHAIN C REGION <400> 2 Xaa Xaa 1 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Aia Pro 130 Leu Val 145 Xaa Xaa Xaa Xaa Xaa Xaa Xa a Xaa 115 Cys Lys Xaa Xaa Xaa Xaa Xaa Xaa Xaa 100 Xaa Ser Asp Xaa Xaa Xaa Xaa Xaa Xaa Xaa Ar g Tyr Xaa Xaa Xaa Xaa Xaa 70 Xaa Xaa Ala Ser Phe 150 Xaa Xaa Xaa Xaa 55 Xaa Xaa Xaa Ser Thr 135 Pro Val1 Xaa Xaa Xaa 40 Xaa Xaa Xaa Xaa Thr 120 Ser Glu His Xaa Xaa 25 Xaa Xaa Xaa Xaa Xaa 105 Lys Giu Pro Thr Xa a Xaa Xaa Xaa Xaa Xaa 90 Xaa Gly Ser Vai Phe 170 Xaa Xaa Xaa Xaa Xaa 75 Xaa Xaa Pro Thr Thr 155 Pro Xaa Xaa Xaa Xaa 60 Xaa Xaa Xaa Ser Ala 140 Val Ala Xaa Xaa Xaa 45 Xaa Xaa Xaa Xaa Val 125 Al a Ser Val Xaa Xaa 30 Xaa Xaa Xaa Xaa Xaa 110 Phe Leu Trp Leu Xaa Xaa Xaa Xaa Xaa Xaa Pro Gly As n Gin 175 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Leu C ys Ser 160 Ser Gly Ala Leu Thr Ser Giy 165 SUBSTITUTE SHEET (RULE 26) WO 99/43713 WO 9/4713PCT/US99/03966' Ser Phe Thr Pro 225 Pro Cys Trp Giu Val 305 Asn Gly Giu Tyr Asn 385 Phe Asn Gly Gly Lys 210 Cys Lys Val Tyr Giu 290 His Lys Gin Met Pro 370 Asn Leu Val1 Leu Thr 195 Val1 Pro Pro Val Val1 275 Gin Gin Gly Pro Thr 355 Ser Tyr Tyr Phe Tyr 180 Gin Asp Ala Lys Val 260 Asp Phe Asp Leu Arq 340 Lys Asp Lys Ser Ser 420 Ser Leu Thr Tyr Lys Thr Pro Pro 230 Asp Thr 245 Asp Vai Gly Val Asn Ser Trp Leu 310 Pro Ala 325 Glu Pro Asn Gin Ile Ala Thr Thr 390 Lys Leu 405 Cys Ser Ser Thr Val1 215 Val1 Leu Ser Glu Thr 295 Asn Pro Gin Val Vali 375 Pro Thr Val1 Ser Cys 200 Giu Ala Met His Val 280 Phe Gly Ile Val Se r 360 Giu Pro Val1 Met Val 185 Asn Arg Gly Ile Giu 265 His Arg Lys Giu Tyr 345 Leu T rp Met Asp His 425 Val1 Vai Lys Pro Ser 250 Asp Asn Vai Giu Lys 330 Thr Thr Giu Le u Lys 410 Thr Asp Cys Ser 235 Arg Pro Ala Val1 Tyr 315 Thr Leu Cys Ser Asp 395 Ser Val His Cys 220 Val Thr Giu Lys Ser 300 Lys Ile Pro Leu As n 380 Ser Arg Pro Lys 205 Val Phe Pro Val1 Thr 285 Vai Cys Ser Pro Val 365 Gly Asp T rp Ser 190 Pro Giu Leu Glu Gin 270 Lys Leu Lys Lys Ser 350 Lys Gin Gly Gin Ser Asn Ser Asn Cys Pro Phe Pro 240 Val Thr 255 Phe Asn Pro Arq Thr Vai Val Ser 320 Thr Lys 335 Arg Giu Giy Phe Pro Giu Ser Phe 400 Gin Gly 415 Giu Ala Leu His Asn His Tyr 430 Thr Gin Lys 435 Ser Leu Ser Leu Pro Gly Lys <210> 3 <211> 494 <212> PRT <213> Homo sapiens <220> <223> TGG-3 CHAIN C REGION <400> 3 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa 1 5-1 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa 25 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa 40 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa 55 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa 70 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa 75 SUBSTITUTE SHEET (RULE 26) \WOf 00/41711 PCT/II S9/03966 vJ 7/J 4 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa 90 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa 100 105 110 Xaa Xaa Xaa Xaa Xaa Ala Ser Thr Lys Gly Pro Ser Val Phe Pro Leu 115 120 125 Ala Pro Cys Ser Arg Ser Thr Ser Gly Gly Thr Ala Ala Leu Gly Cys 130 135 140 Leu Val Lys Asp Tyr Phe Pro Glu Pro Val Thr Val Ser Trp Asn Ser 145 150 155 160 Gly Ala Leu Thr Ser Gly Val His Thr Phe Pro Ala Val Leu Gin Ser 165 170 175 Ser Gly Leu Tyr Ser Leu Ser Ser Val Val Thr Val Pro Ser Ser Ser 180 185 190 Leu Gly Thr Gin Thr Tyr Thr Cys Asn Val Asn His Lys Pro Ser Asn 195 200 205 Thr Lys Val Asp Lys Arg Val Glu Leu Lys Thr Pro Leu Gly Asp Thr 210 215 220 Thr His Thr Cys Pro Arg Cys Pro Glu Pro Lys Ser Cys Asp Thr Pro 225 230 235 240 Pro Pro Cys Pro Arg Cys Pro Glu Pro Lys Ser Cys Asp Thr Pro Pro 245 250 255 Pro Cys Pro Arg Cys Pro Glu Pro Lys Ser Cys Asp Thr Pro Pro Pro 260 265 270 Cys Pro Arg Cys Pro Ala Pro Glu Leu Leu Gly Gly Pro Ser Val Phe 275 280 285 Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu Met Ile Ser Arg Thr Pro 290 295 300 Glu Val Thr Cys Val Val Val Asp Val Ser His Glu Asp Pro Glu Val 305 310 315 320 Gin Phe Lys Trp Tyr Val Asp Gly Val Glu Val His Asn Ala Lys Thr 325 330 335 Lys Pro Arg Glu Glu Gin Tyr Asn Ser Thr Phe Arg Val Val Ser Val 340 345 350 Leu Thr Val Leu His Gin Asp Trp Leu Asn Gly Lys Glu Tyr Lys Cys 355 360 365 Lys Val Ser Asn Lys Ala Leu Pro Ala Pro Ile Glu Lys Thr Ile Ser 370 375 380 Lys Thr Lys Gly Gin Pro Arg Glu Pro Gin Val Tyr Thr Leu Pro Pro 385 390 395 400 Ser Arg Glu Glu Met Thr Lys Asn Gin Val Ser Leu Thr Cys Leu Val 405 410 415 Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val Glu Trp Glu Ser Ser Gly 420 425 430 Gin Pro Glu Asn Asn Tyr Asn Thr Thr Pro Pro Met Leu Asp Ser Asp 435 440 445 Gly Ser Phe Phe Leu Tyr Ser Lys Leu Thr Val Asp Lys Ser Arg Trp 450 455 460 Gin Gin Gly Asn Ile Phe Ser Cys Ser Val Met His Glu Ala Leu His 465 470 475 480 Asn Arg Phe Thr Gin Lys Ser Leu Ser Leu Ser Pro Gly Lys 485 490 SUBSTITUTE SHEET (RULE 26) WO 99/437 13 PTU9/36 PCTIUS99/03966 <210> 4 <211> 444 <212> PRT <213> Homo sapiens <220> <223> IGG-4 CHAIN C REGION <400> 4 Xaa Xaa 1 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Ala Pro 130 Leu Vai 145 Gly Ala Ser Gly Leu Gly Thr Lys 210 Ser Cys 225 Pro Pro Thr Cys Asn Trp Arg Giu 290 Val Leu 305 Ser Asn (aa (aa (a a (a a Xaa X(aa Xa a Xaa 115 Cys Lys Leu Leu Thr 195 Val1 Pro Lys Val Tyr 275 Glu His~ L y, Xaa Xaa Xaa Xaa Xaa Xaa Xaa 100 Xaa Ser Asp Thr Tyr 180 Lys Asp Ala Pro Val1 260 Val Gir G1r Gi' Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Arg Tyr Ser 165 Ser Thr *Lys Pro Lys 245 *Val -Asp SPhe i Asp y Leu 325 ~aa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa (aa (aa (aa (a a 70 Xaa Xa a Al a Ser Phe 150 Gly Leu Tyr Arq Glu 230 Asp Asp Gi Asr Try 31( Prc Xaa X( Xaa Xaa 55 Xaa Xaa Xaa Ser Thr 135 Pro Val Ser Thr Val 215 Phe Thr Val Val Ser 295 Leu Ser a a ~aa 40 (aa (aa (aa (a a rhr 120 Ser G1u His Se r Cys 200 Glu Leu Leu Ser GlL 28( Thi Asi Se~ Xaa X 25 Xaa Xaa Xaa Xaa Xaa 105 Lys Giu Pro Thr Val1 185 Asn *Ser *Gly Met Gin 265 Val Tyr SGly r Ile :aa aa ~aa a a (a a 90 (a a Gly Ser Val1 Phe 170 Val1 Val Lys Giy Ile 25C Glu Arc Ly~ Gl 33~ Xaa Xaa Xaa Xaa 75 Xaa Xaa Pro Thr Thr 155 Pro Thr Asp Tyr Pro 235 Ser Asp Asn ;Vai s Giu 315 u Lys 0 ~aa (aa (aa <aa Kaa Xaa Ser Al a 140 Val Al a Val His Gly 220 Ser Arc Prc Alz Va 30( Ty~ Th: Xaa Xaa Xaa Xaa Xaa Xaa Val1 125 Al a Ser Val1 Pro Lys 205 Pro Val Thr Giu Lys 285 L Ser 0 Lys Ile Xaa Xaa Xaa Xaa Xaa 110 Phe Leu T rp Leu Ser 190 Pro Pro Phe Pro Val 270 Thr Val Cy3 Se2 Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Pro Leu Gly Cys Asn Ser 160 Gin Ser 175 Ser Ser Ser Asn Cys Pro Leu Phe 240 Giu Val 255 Gin Phe *Lys Pro *Leu Thr Lys Val 320 Lys Ala 335 Lys Gly Gin Pro Arg Giu Pro Gin Val Tyr Thr Leu Pro Pro Ser Gin 340 345 350 SUBSTITUTE SHEET (RULE 26) Glu Phe Glu 385 Phe Gly Tyr WO 99/437 Glu Met 355 Tyr Pro 370 Asn Asn Phe Leu Asn Val Thr Gin 435 13 Thr Lys Asn Gin Ser Asp Ile Ala 375 Tyr Lys Thr Thr 390 Tyr Ser Arg Leu 405 Phe Ser Cys Ser 420 Lys Ser Leu Ser -6- Ser Leu Thr Glu Trp Glu Pro Val Leu 395 Val Asp Lys 410 Met His Glu 425 Ser Leu Gly PCT/US99/03966 Cys Leu Val Lys Gly 365 Ser Asn Gly Gin Pro 380 Asp Ser Asp Gly Ser 400 Ser Arg Trp Gin Glu 415 Ala Leu His Asn His 430 Lys <210> <211> 17 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: primer 1 <400> catcggtctt ccccctg <210> 6 <211> <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: primer 2 <400> 6 cggtcctgcg acgggaggtg ctgaggaaga gatgg <210> 7 <211> <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: primer 3 <400> 7 tcttcctcag cacctcccgt cgcaggaccg tcagtcttcc tcttc <210> 8 <211> 17 <212> DNA <213> Artificial Sequence <220> <223> Description of Artificial Sequence: primer 4 <400> 8 gaggcgtggt cttgtag SUBSTITUTE SHEET (RULE 26)

Claims (4)

17- THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS: 1 I. An antibody-based fusion protein with an enhanced circulating half-life, comprising at 2 least a portion of an immunoglobulin (Ig) heavy chain having substantially reduced 3 binding affinity for an Fc receptor, said portion of heavy chain being linked to a second 4 non-Ig protein, said antibody-based fusion protein having a longer circulating half-life in vivo than an unlinked second non-Ig protein. 1 2. The antibody-based fusion protein of claim 1. wherein said portion of heavy chain 2 comprises at least the CH2 domain of an IgG2 or IgG4 constant region. 1 3. The antibody-based fusion protein of claim 1, wherein said portion of heavy chain 2 comprises at least a portion of an IgGI constant region having a mutation or a deletion at 3 one or more amino acid selected from the group consisting of Leu, 34 Leu, 35 Gly 2 3 6 4 Gly 2 3 7 Asn 2 9 7 and Pro 3 3 1 4. The antibody-based fusion protein of claim 1, wherein said portion of heavy chain 2 comprises at least a portion of an IgG3 constant region having a mutation or a deletion at 3 one or more amino acid selected from the group consisting of Leu 28 Leu 2 2 Gly 2 83 4 Gly 2 4, Asn 3 4 4 and Pro 37 1 5. The antibody-based fusion protein of claim 1, wherein said portion of heavy chain further 2 has binding affinity for an immunoglobulin protection receptor. 1 6. The antibody-based fusion protein of claim 1, wherein said portion of heavy chain has 2 substantially reduced binding affinity for a Fc receptor selected from the group consisting 3 of FcyRI, FcyRII and FcyRIII. 1 7. The antibody-based fusion protein of claim 1, wherein said second non-Ig protein is 2 selected from the group consisting of a cytokine. a ligand-binding protein, and a protein 3 toxin. 1 8. The antibody-based fusion protein of claim 7, wherein said cytokine is selected from the 2 group consisting of a tumor necrosis factor, an interleukin, and a lymphokine. 1 9. The antibody-based fusion protein of claim 8. wherein said tumor necrosis factor is tumor 2 necrosis factor alpha. 1 10. The antibody-based fusion protein of claim 8. wherein said interleukin is interleukin-2.
18- 1 11. The antibody-based fusion protein of claim 8. wherein said lymphokine is a lymphotoxin 2 or a colony stimulating factor. 1 12. The antibody-based fusion protein of claim 11, wherein said colony stimulating factor is 2 a granulocyte-macrophage colony stimulating factor. 1 13. The antibody-based fusion protein of claim 7, wherein said ligand-binding protein is 2 selected from the group consisting of CD4, CTLA-4, TNF receptor, and an interleukin 3 receptor. 1 14. A method of increasing the circulating half-life of an antibody-based fusion protein, 2 comprising the step of linking at least a portion of an Ig heavy chain to a second non-Ig 3 protein, said portion of heavy chain having substantially reduced binding affinity for an S 4 Fc receptor, thereby forming an antibody-based fusion protein having a longer circulating 5 half-life in vivo than an unlinked second non-Ig protein. 1 15. The method of claim 14, wherein said portion of heavy chain comprises at least the CH2 S2 domain of an IgG2 or IgG4 constant region. 1 16. A method of increasing the circulating half-life of an antibody-based fusion protein, 2 comprising the steps of: 3 introducing a mutation or a deletion at one or more amino acid of an IgG 1 4 constant region, said amino acid selected from the group consisting of Leu23., S* 5 Leu 2 3 Gly 2 3 6 Gly,,, Asn 97 and Pro 33 1 thereby producing an Ig heavy chain 6 having substantially reduced binding affinity for an Fc receptor; and 7 linking at least a portion of the heavy chain of step to a second non-Ig protein, 8 thereby forming an antibody-based fusion protein having a longer circulating half-life in 9 vivo than an unlinked second non-Ig protein. 1 17. A method of increasing the circulating half-life of an antibody-based fusion protein. 2 comprising the steps of: 3 introducing a mutation or a deletion at one or more amino acid of an IgG3 4 constant region, said amino acid selected from the group consisting of Leus,, Leu 2 Gly 2 83 Gly 2 4, Asn 3 4 and Pro, 37 thereby producing an Ig heavy chain 6 having substantially reduced binding affinity for an Fc receptor; and
19- 7 linking at least a portion of the Ig heavy chain of step to a second non-Ig 8 protein, 9 thereby forming an antibody-based fusion protein having a longer circulating half-life in vivo than an unlinked second non-Ig protein. 1 18. The method of claim 14, 16 or 17, wherein said portion of heavy chain further has 2 binding affinity for an immunoglobulin protection receptor. 1 19. The method of claim 14, 16 or 17, wherein said portion of heavy chain has substantially 2 reduced binding affinity for a Fc receptor selected from the group consisting of FcyRI. 3 FcyRII and FcyRIII. 1 20. The method of claim 14, 16 or 17, wherein said second non-Ig protein is selected from 2 the group consisting of a cytokine, a ligand-binding protein, and a protein toxin. 1 21. The method of claim 20, wherein said cytokine is selected from the group 2 consisting of a tumor necrosis factor, an interleukin, and a lymphokine.
22. The method of claim 21, wherein said tumor necrosis factor is tumor necrosis factor 2 alpha. 1 23. The method of claim 21, wherein said interleukin is interleukin-2. 1 24. The method of claim 21, wherein said lymphokine is a lymphotoxin or a colony S 2 stimulating factor. 1 25. The method of claim 24, wherein said colony stimulating factor is 2 a granulocyte-macrophage colony stimulating factor. S 1 26. The method of claim 20, wherein said ligand-binding protein is selected from 1 26. 2 the group consisting of CD4, CTLA-4, TNF receptor, and an interleukin receptor. Dated this 8 th day of January 2003. Lexigen Pharmaceuticals Corporation By its Patent Attorneys Davies Collison Cave
AU27842/99A 1998-02-25 1999-02-24 Enhancing the circulating half-life of antibody-based fusion proteins Ceased AU758240B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US7588798P 1998-02-25 1998-02-25
US60/075887 1998-02-25
PCT/US1999/003966 WO1999043713A1 (en) 1998-02-25 1999-02-24 Enhancing the circulating half-life of antibody-based fusion proteins

Publications (2)

Publication Number Publication Date
AU2784299A AU2784299A (en) 1999-09-15
AU758240B2 true AU758240B2 (en) 2003-03-20

Family

ID=22128576

Family Applications (1)

Application Number Title Priority Date Filing Date
AU27842/99A Ceased AU758240B2 (en) 1998-02-25 1999-02-24 Enhancing the circulating half-life of antibody-based fusion proteins

Country Status (12)

Country Link
EP (1) EP1060194A1 (en)
JP (1) JP2002505086A (en)
CN (1) CN1204147C (en)
AU (1) AU758240B2 (en)
BR (1) BR9908226A (en)
CA (1) CA2320403A1 (en)
CZ (1) CZ20003099A3 (en)
HK (1) HK1036286A1 (en)
HU (1) HUP0100813A3 (en)
NO (1) NO20004218L (en)
PL (1) PL199659B1 (en)
WO (1) WO1999043713A1 (en)

Families Citing this family (128)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030109680A1 (en) 2001-11-21 2003-06-12 Sunol Molecular Corporation Antibodies for inhibiting blood coagulation and methods of use thereof
US20060235209A9 (en) 1997-03-10 2006-10-19 Jin-An Jiao Use of anti-tissue factor antibodies for treating thromboses
US5986065A (en) 1997-03-10 1999-11-16 Sunol Molecular Corporation Antibodies for inhibiting blood coagulation and methods of use thereof
US7749498B2 (en) 1997-03-10 2010-07-06 Genentech, Inc. Antibodies for inhibiting blood coagulation and methods of use thereof
US6242195B1 (en) 1998-04-02 2001-06-05 Genentech, Inc. Methods for determining binding of an analyte to a receptor
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
NZ539776A (en) 1999-01-15 2006-12-22 Genentech Inc Polypeptide variants with altered effector function
GB9926084D0 (en) * 1999-11-03 2000-01-12 King S College London Recombinant fusion molecules
DK1252192T3 (en) * 2000-02-11 2006-11-20 Merck Patent Gmbh Enhancement of the serum half-life of antibody-based fusion proteins
US7416726B2 (en) 2000-04-13 2008-08-26 The Rockefeller University Enhancement of antibody-mediated immune responses
DE10045591A1 (en) * 2000-09-15 2002-04-04 Klaus Pfizenmaier Site-specific, antibody-mediated activation of proapoptotic cytokines - AMAIZe (Antibody-Mediated Apoptosis Inducing Cytokines)
AU2011253690C1 (en) * 2000-12-12 2018-01-18 Board Of Regents, The University Of Texas System Molecules with extended half-lives, compositions and uses thereof
EP2354149B1 (en) 2000-12-12 2017-08-30 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
US7658921B2 (en) 2000-12-12 2010-02-09 Medimmune, Llc Molecules with extended half-lives, compositions and uses thereof
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
MXPA03008031A (en) * 2001-03-07 2003-12-04 Merck Patent Gmbh Expression technology for proteins containing a hybrid isotype antibody moiety.
WO2002079415A2 (en) 2001-03-30 2002-10-10 Lexigen Pharmaceuticals Corp. Reducing the immunogenicity of fusion proteins
TWI338009B (en) * 2001-10-29 2011-03-01 Genentech Inc Antibodies for inhibiting blood coagulation and methods of use thereof
US20040018573A1 (en) * 2002-04-18 2004-01-29 Power Scott D Production of functional antibodies in filamentous fungi
US7132100B2 (en) 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
US8946387B2 (en) 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
US8968730B2 (en) 2002-08-14 2015-03-03 Macrogenics Inc. FcγRIIB specific antibodies and methods of use thereof
CA2502904C (en) * 2002-10-15 2013-05-28 Protein Design Labs, Inc. Alteration of fcrn binding affinities or serum half-lives of antibodies by mutagenesis
US7217797B2 (en) * 2002-10-15 2007-05-15 Pdl Biopharma, Inc. Alteration of FcRn binding affinities or serum half-lives of antibodies by mutagenesis
AU2004204494B2 (en) 2003-01-09 2011-09-29 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
ATE497783T1 (en) 2003-05-06 2011-02-15 Syntonix Pharmaceuticals Inc CLOTTING FACTOR VII-FC CHIMERIC PROTEINS FOR THE TREATMENT OF HEMOSTATIC DISEASES
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
JP2005073528A (en) * 2003-08-28 2005-03-24 Genetics Inst Llc Method for inhibiting adhesion of blood to biological tissue in biological system, and composition for being used for the method
WO2005037867A1 (en) 2003-10-15 2005-04-28 Pdl Biopharma, Inc. ALTERATION OF Fc-FUSION PROTEIN SERUM HALF-LIVES BY MUTAGENESIS OF POSITIONS 250, 314 AND/OR 428 OF THE HEAVY CHAIN CONSTANT REGION OF IG
AU2005203962C1 (en) 2004-01-05 2012-11-08 Antisoma Research Limited Interleukin-12 targeted to oncofoetal fibronectin
AU2005335714B2 (en) 2004-11-10 2012-07-26 Macrogenics, Inc. Engineering Fc antibody regions to confer effector function
ES2707152T3 (en) 2005-04-15 2019-04-02 Macrogenics Inc Covalent diabodies and uses thereof
US11254748B2 (en) 2005-04-15 2022-02-22 Macrogenics, Inc. Covalent diabodies and uses thereof
US9284375B2 (en) 2005-04-15 2016-03-15 Macrogenics, Inc. Covalent diabodies and uses thereof
US9963510B2 (en) 2005-04-15 2018-05-08 Macrogenics, Inc. Covalent diabodies and uses thereof
SI2298815T1 (en) 2005-07-25 2015-08-31 Emergent Product Development Seattle, Llc B-cell reduction using CD37-specific and CD20-specific binding molecules
DK2573114T3 (en) 2005-08-10 2016-07-04 Macrogenics Inc The identification and production of antibodies with variant Fc regions, and methods of using same
NZ573646A (en) 2006-06-12 2012-04-27 Wyeth Llc Single-chain multivalent binding proteins with effector function
ES2599319T3 (en) 2006-06-26 2017-02-01 Macrogenics, Inc. Fc RIIB specific antibodies and their methods of use
EP2032159B1 (en) 2006-06-26 2015-01-07 MacroGenics, Inc. Combination of fcgammariib antibodies and cd20-specific antibodies and methods of use thereof
ATE480568T1 (en) * 2006-06-30 2010-09-15 Conaris Res Inst Ag IMPROVED SGP 130FC DIMERE
WO2008140603A2 (en) 2006-12-08 2008-11-20 Macrogenics, Inc. METHODS FOR THE TREATMENT OF DISEASE USING IMMUNOGLOBULINS HAVING FC REGIONS WITH ALTERED AFFINITIES FOR FCγR ACTIVATING AND FCγR INHIBITING
TW200907056A (en) * 2007-03-28 2009-02-16 Astrazeneca Ab New method
PE20120259A1 (en) 2007-08-09 2012-04-04 Boehringer Ingelheim Int ANTI-CD37 ANTIBODIES
NZ586053A (en) 2007-11-09 2012-09-28 Peregrine Pharmaceuticals Inc Anti-vegf antibody compositions and methods
US8795667B2 (en) 2007-12-19 2014-08-05 Macrogenics, Inc. Compositions for the prevention and treatment of smallpox
WO2009092011A1 (en) 2008-01-18 2009-07-23 Medimmune, Llc Cysteine engineered antibodies for site-specific conjugation
EP3045475B1 (en) 2008-04-02 2017-10-04 MacroGenics, Inc. Bcr-complex-specific antibodies and methods of using same
US8802093B2 (en) 2008-04-02 2014-08-12 Macrogenics, Inc. HER2/neu-specific antibodies and methods of using same
WO2009126944A1 (en) 2008-04-11 2009-10-15 Trubion Pharmaceuticals, Inc. Cd37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
TWI440469B (en) 2008-09-26 2014-06-11 Chugai Pharmaceutical Co Ltd Improved antibody molecules
EP2373689A1 (en) 2008-12-12 2011-10-12 MedImmune, LLC Crystals and structure of a human igg fc variant with enhanced fcrn binding
JP4885308B2 (en) * 2009-03-19 2012-02-29 中外製薬株式会社 Formulations containing improved antibody molecules
EP2233500A1 (en) 2009-03-20 2010-09-29 LFB Biotechnologies Optimized Fc variants
AU2010238858A1 (en) 2009-04-22 2011-12-08 Merck Patent Gmbh Antibody fusion proteins with modified FcRn binding sites
WO2010141329A1 (en) 2009-06-01 2010-12-09 Medimmune, Llc Molecules with extended half-lives and uses thereof
US20120213705A1 (en) 2009-06-22 2012-08-23 Medimmune, Llc ENGINEERED Fc REGIONS FOR SITE-SPECIFIC CONJUGATION
JP5898082B2 (en) 2009-10-07 2016-04-06 マクロジェニクス,インコーポレーテッド Fc region-containing polypeptide exhibiting improved effector function by changing the degree of fucosylation and use thereof
RU2012127383A (en) 2009-12-02 2014-01-10 Акселерон Фарма Инк. COMPOSITIONS AND METHODS FOR INCREASING THE HALF TIME OF FC-FILLED PROTEINS IN SERUM
TWI505838B (en) 2010-01-20 2015-11-01 Chugai Pharmaceutical Co Ltd Stabilized antibody
PE20130479A1 (en) 2010-03-04 2013-05-12 Macrogenics Inc ANTIBODIES REACTIVE WITH B7-H3, IMMULOGICALLY ACTIVE FRAGMENTS OF THE SAME AND USES OF THE SAME
US8802091B2 (en) 2010-03-04 2014-08-12 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
CN103154025B (en) 2010-08-02 2015-07-01 宏观基因有限公司 Covalent diabodies and uses thereof
UA112062C2 (en) 2010-10-04 2016-07-25 Бьорінгер Інгельхайм Інтернаціональ Гмбх CD33-Binding Agent
PL2643352T3 (en) 2010-11-23 2018-10-31 Glaxo Group Limited Antigen binding proteins to oncostatin m (osm)
US20130236467A1 (en) 2010-11-24 2013-09-12 Jeremy Griggs Multispecific antigen binding proteins targeting hgf
KR101941514B1 (en) 2010-12-22 2019-01-23 테바 파마슈티컬즈 오스트레일리아 피티와이 엘티디 Modified antibody with improved half-life
MX336740B (en) 2011-03-29 2016-01-29 Roche Glycart Ag Antibody fc variants.
EA201892619A1 (en) 2011-04-29 2019-04-30 Роше Гликарт Аг IMMUNOCONJUGATES CONTAINING INTERLEUKIN-2 MUTANT POLYPETIPS
MX347818B (en) 2011-05-21 2017-05-15 Macrogenics Inc Deimmunized serum-binding domains and their use for extending serum half-life.
US8883982B2 (en) 2011-06-08 2014-11-11 Acceleron Pharma, Inc. Compositions and methods for increasing serum half-life
BR112014001855A2 (en) 2011-07-27 2017-02-21 Glaxo Group Ltd antigen-binding construct and protein, dimer, pharmaceutical composition, polynucleotide sequence, host cell and method for construct production
RU2019133467A (en) 2011-10-11 2020-07-06 МЕДИММЬЮН, ЭлЭлСи CD40L-SPECIFIC FRAME STRUCTURES ORIGINING FROM TN3, AND METHODS OF APPLICATION
US11147852B2 (en) 2011-12-23 2021-10-19 Pfizer Inc. Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
HRP20211641T1 (en) 2012-07-13 2022-02-04 Roche Glycart Ag Bispecific anti-vegf/anti-ang-2 antibodies and their use in the treatment of ocular vascular diseases
JP6388581B2 (en) 2012-08-02 2018-09-12 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Method for producing monomer and multimeric molecules and uses thereof
US20140044675A1 (en) 2012-08-10 2014-02-13 Roche Glycart Ag Interleukin-2 fusion proteins and uses thereof
WO2014029752A1 (en) 2012-08-22 2014-02-27 Glaxo Group Limited Anti lrp6 antibodies
TWI682941B (en) 2013-02-01 2020-01-21 美商再生元醫藥公司 Antibodies comprising chimeric constant domains
US9487587B2 (en) 2013-03-05 2016-11-08 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells of a companion animal that express an activating receptor and cells that express B7-H3 and uses thereof
US9908938B2 (en) 2013-03-14 2018-03-06 Macrogenics, Inc. Bispecific molecules that are immunoreactive with immune effector cells that express an activating receptor and an antigen expressed by a cell infected by a virus and uses thereof
ME03796B (en) 2013-03-15 2021-04-20 Glaxosmithkline Ip Dev Ltd Anti-lag-3 binding proteins
UA116479C2 (en) 2013-08-09 2018-03-26 Макродженікс, Інк. Bi-specific monovalent fc diabodies that are capable of binding cd32b and cd79b and uses thereof
US11384149B2 (en) 2013-08-09 2022-07-12 Macrogenics, Inc. Bi-specific monovalent Fc diabodies that are capable of binding CD32B and CD79b and uses thereof
EP2840091A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific diabodies that are capable of binding gpA33 and CD3 and uses thereof
EP2839842A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific monovalent diabodies that are capable of binding CD123 and CD3 and uses thereof
SI3102595T1 (en) 2014-02-06 2019-02-28 F. Hoffmann-La Roche Ag Interleukin-2 fusion proteins and uses thereof
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
TWI701042B (en) 2014-03-19 2020-08-11 美商再生元醫藥公司 Methods and antibody compositions for tumor treatment
WO2015175874A2 (en) 2014-05-16 2015-11-19 Medimmune, Llc Molecules with altered neonate fc receptor binding having enhanced therapeutic and diagnostic properties
US10562946B2 (en) 2014-06-20 2020-02-18 Genentech, Inc. Chagasin-based scaffold compositions, methods, and uses
SI3482766T1 (en) 2014-08-11 2020-09-30 Delinia, Inc. Modified il-2 variants that selectively activate regulatory t cells for the treatment of autoimmune diseases
SG11201702544WA (en) 2014-09-29 2017-04-27 Univ Duke Bispecific molecules comprising an hiv-1 envelope targeting arm
CA2967820A1 (en) 2014-11-17 2016-05-26 Regeneron Pharmaceuticals, Inc. Methods for tumor treatment using cd3xcd20 bispecific antibody
CA2972393A1 (en) 2015-02-27 2016-09-01 Chugai Seiyaku Kabushiki Kaisha Composition for treating il-6-related diseases
CA2981312C (en) 2015-03-30 2023-09-26 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to fc gamma receptors
GB201506393D0 (en) * 2015-04-15 2015-05-27 Berkel Patricius H C Van And Howard Philip W Site-specific antibody-drug conjugates
GB201506389D0 (en) * 2015-04-15 2015-05-27 Berkel Patricius H C Van And Howard Philip W Site-specific antibody-drug conjugates
US11466093B2 (en) 2015-07-27 2022-10-11 The General Hospital Corporation Antibody derivatives with conditionally enabled effector function
JP7074341B2 (en) 2015-09-02 2022-05-24 イムテップ エス.アー.エス. Anti-LAG-3 antibody
TWI799366B (en) 2015-09-15 2023-04-21 美商建南德克公司 Cystine knot scaffold platform
JP7002446B2 (en) 2015-09-21 2022-03-04 アプティーボ リサーチ アンド デベロップメント エルエルシー CD3 binding polypeptide
UY37003A (en) 2015-12-04 2017-06-30 Novartis Ag COMPOSITIONS OF ANTIBODY INTEGRATED WITH CYTOQUINE AND METHODS FOR USE IN IMMUNOR REGULATION
US20170204154A1 (en) 2016-01-20 2017-07-20 Delinia, Inc. Molecules that selectively activate regulatory t cells for the treatment of autoimmune diseases
IL262396B2 (en) 2016-04-15 2023-09-01 Macrogenics Inc Novel b7-h3 binding molecules, antibody drug conjugates thereof and methods of use thereof
US9567399B1 (en) 2016-06-20 2017-02-14 Kymab Limited Antibodies and immunocytokines
EP3509616A1 (en) 2016-09-09 2019-07-17 H. Hoffnabb-La Roche Ag Selective peptide inhibitors of frizzled
WO2018073365A1 (en) * 2016-10-19 2018-04-26 F. Hoffmann-La Roche Ag Method for producing an immunoconjugate
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods
CN110167957A (en) 2016-11-08 2019-08-23 德里尼亚公司 For treating the IL-2 variant of autoimmune disease
US11851486B2 (en) 2017-05-02 2023-12-26 National Center Of Neurology And Psychiatry Method for predicting and evaluating therapeutic effect in diseases related to IL-6 and neutrophils
JOP20190271A1 (en) 2017-05-24 2019-11-21 Novartis Ag Antibody-cytokine engrafted proteins and methods of use for immune related disorders
AU2018347607A1 (en) 2017-10-14 2020-03-26 Cytomx Therapeutics, Inc. Antibodies, activatable antibodies, bispecific antibodies, and bispecific activatable antibodies and methods of use thereof
CA3086199A1 (en) 2017-12-19 2019-06-27 Xencor, Inc. Engineered il-2 fc fusion proteins
MX2021002190A (en) 2018-08-31 2021-05-14 Regeneron Pharma Dosing strategy that mitigates cytokine release syndrome for cd3/c20 bispecific antibodies.
CN112142847B (en) * 2019-02-22 2023-05-05 武汉友芝友生物制药股份有限公司 Engineered Fc fragments, antibodies comprising same and uses thereof
BR112022002236A2 (en) 2019-08-06 2022-05-03 Glaxosmithkline Ip Dev Ltd Compositions, pharmaceutical compositions and use thereof to treat cancer, as well as formulations comprising the same
BR112023024804A2 (en) 2021-05-28 2024-02-15 Glaxosmithkline Ip Dev Ltd COMBINATION THERAPIES TO TREAT CANCER
CA3227515A1 (en) 2021-08-03 2023-02-09 Glaxosmithkline Intellectual Property Development Limited Biopharmaceutical compositions and stable isotope labeling peptide mapping method
CA3233953A1 (en) 2021-10-05 2023-04-13 Matthew Bruce Combination therapies for treating cancer
WO2023114951A1 (en) 2021-12-17 2023-06-22 Viiv Healthcare Company Combination therapies for hiv infections and uses thereof
WO2023212304A1 (en) 2022-04-29 2023-11-02 23Andme, Inc. Antigen binding proteins
WO2024042112A1 (en) 2022-08-25 2024-02-29 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins and uses thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9511935D0 (en) * 1995-06-13 1995-08-09 Smithkline Beecham Plc Novel compound
AU2527397A (en) * 1996-03-13 1997-10-01 Protein Design Labs, Inc. Fas ligand fusion proteins and their uses

Also Published As

Publication number Publication date
HUP0100813A3 (en) 2003-08-28
JP2002505086A (en) 2002-02-19
NO20004218D0 (en) 2000-08-23
BR9908226A (en) 2000-10-24
PL199659B1 (en) 2008-10-31
CZ20003099A3 (en) 2002-04-17
CA2320403A1 (en) 1999-09-02
PL342497A1 (en) 2001-06-04
WO1999043713A1 (en) 1999-09-02
CN1204147C (en) 2005-06-01
EP1060194A1 (en) 2000-12-20
NO20004218L (en) 2000-10-24
CN1291995A (en) 2001-04-18
HUP0100813A2 (en) 2001-06-28
AU2784299A (en) 1999-09-15
HK1036286A1 (en) 2001-12-28

Similar Documents

Publication Publication Date Title
AU758240B2 (en) Enhancing the circulating half-life of antibody-based fusion proteins
US20030105294A1 (en) Enhancing the circulating half life of antibody-based fusion proteins
EP1366067B1 (en) Expression technology for proteins containing a hybrid isotype antibody moiety
AU2002248571A1 (en) Expression technology for proteins containing a hybrid isotype antibody moiety
EP3433273B1 (en) Binding molecules to cd38 and pd-l1
JPH05502384A (en) Preparation of antibodies
CA2328606A1 (en) Antibodies to cd23, derivatives thereof, and their therapeutic uses
TW201932142A (en) Method for generating multispecific antibodies from monospecific antibodies
US11560437B2 (en) Stable multispecific antibodies
EP2496603B1 (en) Glycosylated repeat-motif-molecule conjugates
CN112243444A (en) Peptide linker with reduced post-translational modifications
CN114249818A (en) Truncated multivalent multimers
EP0730649A1 (en) Mutated hig-e fragments and derivative thereof
WO2020010077A1 (en) Bispecific antibodies
MXPA00008312A (en) Enhancing the circulating half-life of antibody-based fusion proteins
RU2779602C2 (en) Stable multi-specific antibodies
CN117320752A (en) Modified FcRn binding fragments with improved half-life

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)