AU2784299A - Enhancing the circulating half-life of antibody-based fusion proteins - Google Patents

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

Info

Publication number
AU2784299A
AU2784299A AU27842/99A AU2784299A AU2784299A AU 2784299 A AU2784299 A AU 2784299A AU 27842/99 A AU27842/99 A AU 27842/99A AU 2784299 A AU2784299 A AU 2784299A AU 2784299 A AU2784299 A AU 2784299A
Authority
AU
Australia
Prior art keywords
antibody
fusion protein
protein
based fusion
heavy chain
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.)
Granted
Application number
AU27842/99A
Other versions
AU758240B2 (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
Merck Patent GmbH
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 Merck Patent GmbH filed Critical Merck Patent GmbH
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 5 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 10 more sophisticated with the introduction of genetic engineering. Several techniques have been developed to improve the utility of antibodies. These include: (1) the generation of monoclonal antibodies by cell fusion to create "hyridomas", or by molecular cloning of antibody heavy (H) and light (L) chains from antibody-producing cells; (2) the conjugation of other molecules to antibodies to deliver them to preferred sites in vivo, e.g., radioisotopes, toxic drugs, protein 15 toxins, and cytokines; (3) the manipulation of antibody effector functions to enhance or diminish biological activity; (4) the joining of other protein such as toxins and cytokines with antibodies at the genetic level to produce antibody-based fusion proteins; and (5) 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 20 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 25 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.
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 5 that all of the fusion proteins tested had an c. phase (distribution phase) half-life of less than 1.5 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 half life of antibody-based fusion proteins. 10 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 15 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 20 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 IgG1 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 25 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 30 is reduced by introducing a genetic modification of one or more amino acid in the constant WO 99/43713 PCT/US99/03966 -3 region 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 5 antibody-based fusion protein with enhanced in vivo circulating half-life is obtained by first introducing a mutation, deletion, or insertion in the IgG1 constant region at one or more amino acid selected from Leu 2 34, Leu 2 35 , Gly 236 , Gly 237 , Asn 297 , and Pro 331 , 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 10 region at one or more amino acid selected from Leu 2 8 , Leu282, Gly283, Gly284,Asn344, and Pro378, 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 15 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 (IL-2), hematopoietic factors such as granulocyte-macrophage colony stimulating factor (GM-CSF), tumor necrosis factor (TNF) such as TNFa, and 20 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, (1) block receptor-ligand interactions at the cell surface; or 25 (2) neutralize the biological activity of a molecule (e.g., 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.
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 5 specific for a target antigen and a constant region linked through a peptide bond to a second non immunoglobulin protein. The constant region may be the constant region normally associated with the variable region, or a different one, e.g., 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 10 and variable regions (i.e., 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 15 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 non immunoglobulin protein can exert a localized biological effect. In an alternative preferred embodiment, the antibody-based fusion protein comprises a 20 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 non immunoglobulin protein, but which lack the antigen-binding specificity and activity of an antibody. 25 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.
WO 99/43713 PCT/US99/03966 -5 The invention also features DNA constructs encoding the above-described fusion proteins, and cell lines, e.g., 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. 5 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 10 Cy 1 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 Cy 1,
C
7 2, and Cy 4, aligned to maximize amino acid identity, and wherein non-conserved amino acids are identified by boxes; 15 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; 20 FIG. 5 is a line graph depicting the in vivo plasma concentration of total antibody (free antibody and fusion protein) of hu-KSyI-IL2 (closed diamond) and hu-KSy4-IL2 (closed triangle) and of intact fusion protein of hu-KSy1-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 25 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-KSy I -IL2 (0); mutated hu-KSy 1-IL2 (11) and hu-KSy4-IL2 (A) as a function of time.
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 5 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 10 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, IgG1 (Cyl) and IgG3 (C 7 3) are known to bind FcRyl with high affinity, whereas IgG4 (C 7 4) has a 10-fold lower binding affinity, and IgG2 (Cy2) does not bind to FcRyl. Thus, an antibody-based fusion protein with reduced binding 15 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 20 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 FcRyl with high affinity, whereas Cy4 and Cy2 have reduced or no binding affinity for FcRyl. A comparison of the Cyl and Cy3 indicates that, with the exception of an extended hinge segment in C 7 3, the amino acid sequence homology between 25 these two isotypes is very high. This is true even in those regions that have been shown to interact with the C 1q fragment of complement and the various FcyR classes. FIG. 1 provides a alignment of the amino acid sequences of Cyl1 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 WO 99/43713 PCT/US99/03966 -7 sequences for FcyR binding are Leu-Leu-Gly-Gly (residues 234 through 237 in Cyl1), 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 5 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 3 3, 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 297 . Enzymatic removal of this structure or mutation 10 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 15 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 20 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 25 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 30 assembled in or inserted into an expression vector for transfection of the appropriate recipient cells where it is expressed.
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 C'y4 IgG constant regions. According to the present invention, antibody-based fusion proteins with enhanced in vivo 5 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 Cy71 constant region (Fc 10 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. Sc. USA 89: 1428-1432 (1992); and U.S. Patent No 5,650,150, the disclosure of which is incorporated herein 15 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 (V) 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 20 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 (H) chain variable (V) regions derived from the mouse antibody KS-1/4. The mouse V regions were humanized by standard methods and their encoding DNA sequences were 25 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 WO 99/43713 PCT/US99/03966 -9 also 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 (CH1), just downstream from a unique Hind III common to all human Cy genes. A 3' untranslated and polyadenylation site from 5 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 Cyl71-containing plasmid DNA with Hind III and Xho I and purifying the large 7.8 kb fragment by 10 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 Cyl 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 15 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 CylI (no Fsp I) and Cy4 (one site) gene inserts. The final vector, containing the Cy4-IL2 heavy chain 20 replacement, was introduced into mouse myeloma cells and transfectants were selected by growth in medium containing methotrexate (0.1 pM). 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 25 measured in a T-cell proliferation assay and found to be identical to that of the C71yl 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 FcRyl that is capable of binding mouse or 30 human IgG of the appropriate subclasses. Likewise, the human K562 erythroleukemic cell line WO 99/43713 PCT/US99/03966 - 10 expresses FcRyII but not FcRyl. 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 - I L2 fusion protein, and a Cy4-IL2 fusion protein for FcRyI were compared in the mouse J774 cell line. 5 The two antibody-IL-2 fusion proteins described in Example 1, hu-KSy1 -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 (Fab 2 ) was added and incubation was continued for an additional 30 min. Unbound antibodies were removed by two washes with PBS-BSA, and 10 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 KSy1 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 15 the Cyl-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. 20 In order to determine whether the greater binding observed with IL-2 fusion proteins was due to the presence of IL-2 receptors or FcRyl receptors on the cells, excess mouse IgG (mIgG) was used to compete the binding at the Fe 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 25 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 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. 5 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 Cy 1 isotype heavy chain (i.e., hu-KSyI-IL2) were compared to fusion proteins containing the Cy4 isotype heavy chain (i.e., hu-KSy4-IL2). Purified humanized KS-1/4-IL2 fusion proteins containing either the C1yl or Cy4 isotype 10 heavy chain were buffer-exchanged by diafiltration into phosphate buffered saline (PBS) and diluted further to a concentration of -100 pg/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 15 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'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 20 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 25 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 c. phase half-life of the hu-KSy4-IL2 fusion protein was significantly longer than the cc phase half-life of the hu-KSy1-IL2 fusion protein. The increased half-life is best exemplified by the significantly higher concentrations of the hu-KSy4-IL2 fusion WO 99/43713 PCT/US99/03966 -12 protein (3.3 pg/ml) compared to the hu-KSyI-IL2 fusion protein (60 ng/ml) found in mice after 24 hours. The hu-KSy I -IL2 protein had a rapid distribution (a) phase followed by a slower catabolic (3) phase, as reported earlier for the chimeric 14.18-IL2 fusion protein. See, Gillies et 5 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 (1) an antibody-specific ELISA, and (2) a fusion protein specific ELISA (i.e., an ELISA that requires that both the antibody and IL-2 components be 10 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 15 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 20 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 25 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 WO 99/43713 PCT/US99/03966 - 13 circulating half-life compared to antibody-based fusion proteins having binding affinity for a Fc receptor. Example 2 Mutating the human Cyl or Cy3 gene in antibody-based fusion protein constructs to improve their in vivo circulating half-life. 5 IgG molecules interact with several molecules in the circulation, including members of the complement system of proteins (e.g., C1lq fragment), as well as the three classes of FcR. The important residues for Clq binding are residues Glu 318 , Lys 320 , and Lys 32 2 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 1 q binding as mechanisms for rapid clearance, we substituted 10 the more drastically altered Cy2 hinge-proximal segment into the Cy71 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 15 junction of two adjacent PCR fragments spanning a Pst I to Drd I fragment (see FIG. 6). 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 C71yl gene as the template. The cycle conditions for the primary PCR were 35 cycles of: 94oC for 45 sec, annealing at 48oC for 45 seconds, and primer extension at 72oC for 45 sec. The products of each 20 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: 94oC for 1 min, annealing at 51 oC for 1 min, and primer extension at 72oC for 1 min. Joining occurs as a result of the overlapping between the two individual fragments which pairs with the 25 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 30 vector containing the human Cyl gene was digested with Pst I and Xho I to remove the WO 99/43713 PCT/US99/03966 -14 non-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 Cy 1-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 5 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 10 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 of hu-KSy1-
I
L2 at various specified 15 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 20 clearance involve FcR binding rather than C 1q 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 C 7 4 gene in order to further reduce the binding of Cy4 25 fusion proteins to FcR. For example, additional possible mutations include mutations in the hinge proximal amino acid residues, mutating Pro 33 1 , or by mutating the single N-linked glycosylation site in all IgG Fc regions. The latter is located at Asn 297 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 30 amino acid Gln, for example, would have little effect on the protein but would prevent the WO 99/43713 PCT/US99/03966 - 15 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. 5 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 10 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 (i.e., the second non-Ig protein) to more effectively (1) block receptor-ligand interactions at the cell surface; or (2) neutralize the biological activity 15 of a molecule (e.g., 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-antibody based 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 antibody based fusion proteins with human Cy4 Fc regions. 20 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 Cy1 or C7y4 gene downstream of their endogenous Hind III site. 25 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 30 and transfectants are selected by growth in culture medium containing methotrexate (0.1 tM).
WO 99/43713 PCT/US99/03966 - 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) 5 of either the CD4-Cyl or CD4-Cy4 fusion protein and the pharmacokinetics 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.

Claims (1)

  1. - 17 - What is claimed is: 1. An antibody-based fusion protein with an enhanced circulating half-life, comprising at least a portion of an immunoglobulin (Ig) heavy chain having substantially reduced binding affinity for an Fc receptor, said portion of heavy chain being linked to a second non-Ig protein, said antibody-based fusion protein having a longer circulating half-life in vivo than an unlinked second non-Ig protein. 2. The antibody-based fusion protein of claim 1, wherein said portion of heavy chain comprises at least the CH2 domain of an IgG2 or IgG4 constant region. 3. The antibody-based fusion protein of claim 1 , wherein said portion of heavy chain comprises at least a portion of an IgGl constant region having a mutation or a deletion at one or more amino acid selected from the group consisting of Leu234, Leu235, Gly236, Gly237, Asn297, and Pro331. 4. The antibody-based fusion protein of claim 1, wherein said portion of heavy chain comprises at least a portion of an IgG3 constant region having a mutation or a deletion at one or more amino acid selected from the group consisting of Leu281, Leu282, Gly283, Gly284, Asn344, and Pro378. 5. The antibody-based fusion protein of claim 1, wherein said portion of heavy chain further has binding affinity for an immunoglobulin protection receptor. 6. The antibody-based fusion protein of claim 1 , wherein said portion of heavy chain has substantially reduced binding affinity for a Fc receptor selected from the group consisting of FcγRI, FcγRII and FcγRIII. 7. The antibody-based fusion protein of claim 1 , wherein said second non-Ig protein is selected from the group consisting of a cytokine, a ligand-binding protein, and a protein toxin. 8. The antibody-based fusion protein of claim 1, wherein said cytokine is selected from the group consisting of a tumor necrosis factor, an interleukin, and a lymphokine. 9. The antibody-based fusion protein of claim 8, wherein said tumor necrosis factor is tumor necrosis factor alpha. 10. The antibody-based fusion protein of claim 8, wherein said interleukin is interleukin-2. - 18 - 1 1. The antibody-based fusion protein of claim 8, wherein said lymphokine is a lymphotoxin or a colony stimulating factor. 12. The antibody-based fusion protein of claim 1 1 , wherein said colony stimulating factor is a granulocyte-macrophage colony stimulating factor. 13. The antibody-based fusion protein of claim 1 , wherein said ligand-binding protein is selected from the group consisting of CD4, CTLA-4, TNF receptor, and an interleukin receptor. 14. A method of increasing the circulating half-life of an antibody-based fusion protein, comprising the step of linking at least a portion of an Ig heavy chain to a second non-Ig protein, said portion of heavy chain having substantially reduced binding affinity for an Fc receptor, thereby forming an antibody-based fusion protein having a longer circulating half-life in vivo than an unlinked second non-Ig protein. 15. The method of claim 14, wherein said portion of heavy chain comprises at least the CH2 domain of an IgG2 or IgG4 constant region. 16. A method of increasing the circulating half-life of an antibody-based fusion protein, comprising the steps of: (a) introducing a mutation or a deletion at one or more amino acid of an IgGl constant region, said amino acid selected from the group consisting of Leu234, Leu235, Gly236, Gly237, Asn297, and Pro331, thereby producing an Ig heavy chain having substantially reduced binding affinity for an Fc receptor; and (b) linking at least a portion of the heavy chain of step (a) to a second non-Ig protein, thereby forming an antibody-based fusion protein having a longer circulating half-life in vivo than an unlinked second non-Ig protein. 17. A method of increasing the circulating half-life of an antibody -based fusion protein, comprising the steps of: (a) introducing a mutation or a deletion at one or more amino acid of an IgG3 constant region, said amino acid selected from the group consisting of Leu281, Leu282, Gly283, Gly284, Asn344, and Pro378, thereby producing an Ig heavy chain having substantially reduced binding affinity for an Fc receptor; and - 19 - (b) linking at least a portion of the Ig heavy chain of step (a) to a second non-Ig protein, thereby forming an antibody-based fusion protein having a longer circulating half-life in vivo than an unlinked second non-Ig protein. 18. The method of claim 14, 16 or 17, wherein said portion of heavy chain further has binding affinity for an immunoglobulin protection receptor. 19. The method of claim 14, 16 or 17, wherein said portion of heavy chain has substantially reduced binding affinity for a Fc receptor selected from the group consisting of FcγRI, FcγRII and FcγRIII. 20. The method of claim 14, 16 or 17, wherein said second non-Ig protein is selected from the group consisting of a cytokine, a ligand-binding protein, and a protein toxin. 21. The method of claim 14, 16 or 17, wherein said cytokine is selected from the group 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 alpha. 23. The method of claim 21 , wherein said interleukin is interleukin-2. 24. The method of claim 21 , wherein said lymphokine is a lymphotoxin or a colony stimulating factor. 25. The antibody-based fusion protein of claim 24, wherein said colony stimulating factor is a granulocyte-macrophage colony stimulating factor. 26. The method of claim 14, 16 or 17, wherein said ligand-binding protein is selected from the group consisting of CD4, CTLA-4, TNF receptor, and an interleukin receptor.
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 true AU2784299A (en) 1999-09-15
AU758240B2 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
US7749498B2 (en) 1997-03-10 2010-07-06 Genentech, Inc. Antibodies for inhibiting blood coagulation and methods of use thereof
US20030109680A1 (en) 2001-11-21 2003-06-12 Sunol Molecular Corporation Antibodies for inhibiting blood coagulation and methods of use thereof
US5986065A (en) 1997-03-10 1999-11-16 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
US6528624B1 (en) 1998-04-02 2003-03-04 Genentech, Inc. Polypeptide variants
US6194551B1 (en) 1998-04-02 2001-02-27 Genentech, Inc. Polypeptide variants
US6242195B1 (en) 1998-04-02 2001-06-05 Genentech, Inc. Methods for determining binding of an analyte to a receptor
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
EP2386574A3 (en) 1999-01-15 2012-06-27 Genentech, Inc. Polypeptide variants with altered effector function
GB9926084D0 (en) * 1999-11-03 2000-01-12 King S College London Recombinant fusion molecules
HUP0204475A2 (en) * 2000-02-11 2003-04-28 Merck Patent Gmbh Enhancing the circulating half-life of antibody-based fusion proteins
EP2264072A1 (en) 2000-04-13 2010-12-22 The Rockefeller University Enhancement of antibody-mediated cytotoxicity.
DE10045591A1 (en) * 2000-09-15 2002-04-04 Klaus Pfizenmaier Site-specific, antibody-mediated activation of proapoptotic cytokines - AMAIZe (Antibody-Mediated Apoptosis Inducing Cytokines)
US7658921B2 (en) 2000-12-12 2010-02-09 Medimmune, Llc Molecules with extended half-lives, compositions and uses thereof
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
EP2357187A1 (en) * 2000-12-12 2011-08-17 MedImmune, LLC Molecules with extended half-lives, compositions and uses thereof
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
KR100900176B1 (en) * 2001-03-07 2009-06-02 메르크 파텐트 게엠베하 Expression technology for proteins containing a hybrid isotype antibody moiety
US6992174B2 (en) 2001-03-30 2006-01-31 Emd Lexigen Research Center 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
AU2003222664A1 (en) * 2002-04-18 2003-11-03 Genencor International, Inc. 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
PT1562972E (en) * 2002-10-15 2010-11-10 Facet Biotech Corp 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
US7960512B2 (en) 2003-01-09 2011-06-14 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
EP2368578A1 (en) 2003-01-09 2011-09-28 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
ES2361036T3 (en) 2003-05-06 2011-06-13 Syntonix Pharmaceuticals, Inc. CHEMICAL PROTEINS VII-FC OF THE COAGULATION FACTOR FOR THE TREATMENT OF A HEMOSTATIC DISORDER.
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
EP1673395A1 (en) 2003-10-15 2006-06-28 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
US8420087B2 (en) 2004-01-05 2013-04-16 Antisoma Research Limited Interleukin-12 targeted to oncofoetal fibronectin
EP1810035A4 (en) 2004-11-10 2010-03-17 Macrogenics Inc Engineering fc antibody regions to confer effector function
US11254748B2 (en) 2005-04-15 2022-02-22 Macrogenics, Inc. Covalent diabodies and uses thereof
US9963510B2 (en) 2005-04-15 2018-05-08 Macrogenics, Inc. Covalent diabodies and uses thereof
WO2006113665A2 (en) 2005-04-15 2006-10-26 Macrogenics, Inc. Covalent diabodies and uses thereof
US9284375B2 (en) 2005-04-15 2016-03-15 Macrogenics, Inc. Covalent diabodies and uses thereof
ES2460517T3 (en) 2005-07-25 2014-05-13 Emergent Product Development Seattle, Llc Reduction of b cells by using cd37 specific binding and cd20 specific binding molecules
HUE029465T2 (en) 2005-08-10 2017-02-28 Macrogenics Inc Identification and engineering of antibodies with variant Fc regions and methods of using same
AU2007257692B2 (en) 2006-06-12 2013-11-14 Aptevo Research And Development Llc Single-chain multivalent binding proteins with effector function
AU2007281876B2 (en) 2006-06-26 2013-09-05 Macrogenics, Inc. FcgammaRIIB-specific antibodies and methods of use thereof
EP2032159B1 (en) 2006-06-26 2015-01-07 MacroGenics, Inc. Combination of fcgammariib antibodies and cd20-specific antibodies and methods of use thereof
ES2352561T3 (en) * 2006-06-30 2011-02-21 Conaris Research Institute Ag IMPROVED SGP DIMMERS 130FC.
US8652466B2 (en) 2006-12-08 2014-02-18 Macrogenics, Inc. Methods for the treatment of disease using immunoglobulins having Fc regions with altered affinities for FcγRactivating and FcγRinhibiting
TW200907056A (en) * 2007-03-28 2009-02-16 Astrazeneca Ab New method
PE20140196A1 (en) 2007-08-09 2014-03-19 Boehringer Ingelheim Int ANTI-CD37 ANTIBODIES
KR101502267B1 (en) 2007-11-09 2015-03-18 페레그린 파마수티컬즈, 인크 Anti-vegf antibody compositions and methods
US8795667B2 (en) 2007-12-19 2014-08-05 Macrogenics, Inc. Compositions for the prevention and treatment of smallpox
ES2613963T3 (en) 2008-01-18 2017-05-29 Medimmune, Llc Cysteine manipulated antibodies for site specific conjugation
BRPI0906309A2 (en) 2008-04-02 2020-05-26 Macrogenics, Inc IMMUNOGLOBULIN, ANTIBODY, USE OF ANTIBODY AND PHARMACEUTICAL COMPOSITION
CN102046655B (en) 2008-04-02 2016-09-14 宏观基因有限公司 BCR-complex-specific antibody and its using method
DK2132228T3 (en) 2008-04-11 2011-10-10 Emergent Product Dev Seattle CD37 immunotherapeutic agent and combination with its bifunctional chemotherapeutic agent
TWI440469B (en) 2008-09-26 2014-06-11 Chugai Pharmaceutical Co Ltd Improved antibody molecules
WO2010068722A1 (en) 2008-12-12 2010-06-17 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
SG175233A1 (en) 2009-04-22 2011-11-28 Merck Patent Gmbh Antibody fusion proteins with modified fcrn binding sites
EP2437767B1 (en) 2009-06-01 2015-07-08 MedImmune, LLC Molecules with extended half-lives and uses thereof
EP2445520A4 (en) 2009-06-22 2013-03-06 Medimmune Llc ENGINEERED Fc REGIONS FOR SITE-SPECIFIC CONJUGATION
ES2672121T3 (en) 2009-10-07 2018-06-12 Macrogenics, Inc. Polypeptides containing Fc region that have an enhanced effector function due to alterations in the degree of fucosylation, and methods for their use
WO2011068993A1 (en) 2009-12-02 2011-06-09 Acceleron Pharma Inc. Compositions and methods for increasing serum half-life of fc fusion proteins.
AR080428A1 (en) 2010-01-20 2012-04-11 Chugai Pharmaceutical Co Ltd FORMULATIONS STABILIZED LIQUID CONTAINERS OF ANTIBODIES
US8802091B2 (en) 2010-03-04 2014-08-12 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
PL2542256T3 (en) 2010-03-04 2020-01-31 Macrogenics, Inc. Antibodies reactive with b7-h3, immunologically active fragments thereof and uses thereof
EP2601216B1 (en) 2010-08-02 2018-01-03 MacroGenics, Inc. Covalent diabodies and uses thereof
UA112062C2 (en) 2010-10-04 2016-07-25 Бьорінгер Інгельхайм Інтернаціональ Гмбх CD33-Binding Agent
US8916695B2 (en) 2010-11-23 2014-12-23 Glaxo Group Limited Antigen binding proteins to oncostatin M (OSM)
WO2012069557A1 (en) 2010-11-24 2012-05-31 Glaxo Group Limited 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
RS57895B1 (en) 2011-03-29 2019-01-31 Roche Glycart Ag Antibody fc variants
EA201892619A1 (en) 2011-04-29 2019-04-30 Роше Гликарт Аг IMMUNOCONJUGATES CONTAINING INTERLEUKIN-2 MUTANT POLYPETIPS
KR102030531B1 (en) 2011-05-21 2019-10-10 마크로제닉스, 인크. Deimmunized serum-binding domains and their use for extending serum half-life
WO2012170938A1 (en) 2011-06-08 2012-12-13 Acceleron Pharma Inc. Compositions and methods for increasing serum half-life
CA2842099A1 (en) 2011-07-27 2013-01-31 Glaxo Group Limited Antigen binding constructs
WO2013055745A2 (en) 2011-10-11 2013-04-18 Medimmune, Llc Cd40l-specific tn3-derived scaffolds and methods of use thereof
US11147852B2 (en) 2011-12-23 2021-10-19 Pfizer Inc. Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
CN104428315B (en) 2012-07-13 2017-09-29 罗氏格黎卡特股份公司 Bispecific anti-vegf/antibody of anti-ANG 2 and its application in treatment Ocular Vessels disease
EP2880169B1 (en) 2012-08-02 2017-05-17 F. Hoffmann-La Roche AG Method for producing monomeric and multimeric molecules and uses thereof
US20140044675A1 (en) * 2012-08-10 2014-02-13 Roche Glycart Ag Interleukin-2 fusion proteins and uses thereof
EP2888279A1 (en) 2012-08-22 2015-07-01 Glaxo Group Limited Anti lrp6 antibodies
TWI635098B (en) 2013-02-01 2018-09-11 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
UA118750C2 (en) 2013-03-15 2019-03-11 Ґлаксосмітклайн Інтеллектуал Проперті Дивелопмент Лімітед Anti-lag-3 binding proteins
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
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
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
EP2840091A1 (en) 2013-08-23 2015-02-25 MacroGenics, Inc. Bi-specific diabodies that are capable of binding gpA33 and CD3 and uses thereof
MX370283B (en) 2014-02-06 2019-12-09 Hoffmann La Roche 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
CN106573961B (en) 2014-06-20 2022-01-04 豪夫迈·罗氏有限公司 CHAGASIN-based scaffold compositions, methods and uses
HUE051414T2 (en) 2014-08-11 2021-03-01 Delinia Inc Modified il-2 variants that selectively activate regulatory t cells for the treatment of autoimmune diseases
JP6932639B2 (en) 2014-09-29 2021-09-08 デューク ユニバーシティ Bispecific molecule with HIV-1 envelope targeting arm
EP3699198A1 (en) 2014-11-17 2020-08-26 Regeneron Pharmaceuticals, Inc. Methods for tumor treatment using cd3xcd20 bispecific antibody
AU2016224409B2 (en) 2015-02-27 2021-01-28 Chugai Seiyaku Kabushiki Kaisha Composition for treating IL-6-related diseases
EP3277725B1 (en) 2015-03-30 2020-11-25 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to fc gamma receptors
GB201506389D0 (en) * 2015-04-15 2015-05-27 Berkel Patricius H C Van And Howard Philip W Site-specific antibody-drug conjugates
GB201506393D0 (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
SG10201913864RA (en) 2015-09-02 2020-03-30 Immutep Sas Anti-LAG-3 Antibodies
TWI799366B (en) 2015-09-15 2023-04-21 美商建南德克公司 Cystine knot scaffold platform
UA126278C2 (en) 2015-09-21 2022-09-14 Аптево Рісьорч Енд Девелопмент Ллс Cd3 binding polypeptides
JP6794448B2 (en) 2015-12-04 2020-12-02 ノバルティス アーゲー Antibody Cytokine Graft Composition and Usage for Immunomodulation
US20170204154A1 (en) 2016-01-20 2017-07-20 Delinia, Inc. Molecules that selectively activate regulatory t cells for the treatment of autoimmune diseases
BR112018071105A2 (en) 2016-04-15 2019-02-26 Macrogenics, Inc. drug and antibody conjugate, binding molecule, pharmaceutical composition and use
US9567399B1 (en) 2016-06-20 2017-02-14 Kymab Limited Antibodies and immunocytokines
CN110198729A (en) 2016-09-09 2019-09-03 豪夫迈·罗氏有限公司 The selective inhibitor peptides of frizzled protein
EP3529350A1 (en) * 2016-10-19 2019-08-28 H. Hoffnabb-La Roche Ag Method for producing an immunoconjugate
EP3534947A1 (en) 2016-11-03 2019-09-11 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses & methods
AU2017359172A1 (en) 2016-11-08 2019-05-16 Delinia, Inc. IL-2 variants for the treatment of autoimmune diseases
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
TW201927816A (en) 2017-10-14 2019-07-16 美商Cytomx生物製藥公司 Antibodies, activatable antibodies, bispecific antibodies, and bispecific activatable antibodies and methods of use thereof
SG11202005732XA (en) 2017-12-19 2020-07-29 Xencor Inc Engineered il-2 fc fusion proteins
WO2020047389A1 (en) 2018-08-31 2020-03-05 Regeneron Pharmaceuticals, Inc. Dosing strategy that mitigates cytokine release syndrome for cd3/c20 bispecific antibodies
KR20210143192A (en) * 2019-02-22 2021-11-26 우한 이지 바이오파마 씨오., 엘티디. Modified Fc fragments, antibodies comprising same, and applications thereof
US20220289859A1 (en) 2019-08-06 2022-09-15 Glaxosmithkline Intellectual Property Development Limited Biopharmacuetical Compositions and Related Methods
CN117396513A (en) 2021-05-28 2024-01-12 葛兰素史密斯克莱知识产权发展有限公司 Combination therapy for the treatment of cancer
KR20240040786A (en) 2021-08-03 2024-03-28 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 Biopharmaceutical compositions and stable isotope labeled peptide mapping methods
WO2023057893A1 (en) 2021-10-05 2023-04-13 Glaxosmithkline Intellectual Property Development Limited Combination therapies for treating cancer
WO2023114951A1 (en) 2021-12-17 2023-06-22 Viiv Healthcare Company Combination therapies for hiv infections and uses thereof
US20230348604A1 (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
US6046310A (en) * 1996-03-13 2000-04-04 Protein Design Labs., Inc. FAS ligand fusion proteins and their uses

Also Published As

Publication number Publication date
CN1204147C (en) 2005-06-01
WO1999043713A1 (en) 1999-09-02
PL342497A1 (en) 2001-06-04
CN1291995A (en) 2001-04-18
HUP0100813A2 (en) 2001-06-28
NO20004218L (en) 2000-10-24
PL199659B1 (en) 2008-10-31
AU758240B2 (en) 2003-03-20
JP2002505086A (en) 2002-02-19
HUP0100813A3 (en) 2003-08-28
NO20004218D0 (en) 2000-08-23
CA2320403A1 (en) 1999-09-02
EP1060194A1 (en) 2000-12-20
BR9908226A (en) 2000-10-24
CZ20003099A3 (en) 2002-04-17
HK1036286A1 (en) 2001-12-28

Similar Documents

Publication Publication Date Title
AU758240B2 (en) Enhancing the circulating half-life of antibody-based fusion proteins
US20090088561A1 (en) Enhancing the circulating half-life of antibody-based fusion proteins
JP4234438B2 (en) Expression technology of protein containing hybrid isotype antibody part
Coloma et al. Design and production of novel tetravalent bispecific antibodies
JP3081641B2 (en) Preparation of antibodies
US7053202B2 (en) Immunoglobulin DNA cassette molecules, monobody constructs, methods of production, and methods of use therefor
AU2002248571A1 (en) Expression technology for proteins containing a hybrid isotype antibody moiety
JP2012176969A (en) Enhancing the circulating half-life of antibody-based fusion protein
TW201932142A (en) Method for generating multispecific antibodies from monospecific antibodies
US11560437B2 (en) Stable multispecific antibodies
CN112243444A (en) Peptide linker with reduced post-translational modifications
Shin et al. Hybrid antibodies
US20210253741A1 (en) Bispecific antibodies
EP0730649A1 (en) Mutated hig-e fragments and derivative thereof
CN114249818A (en) Truncated multivalent multimers
WO2019178539A1 (en) Bispecific antibodies
MXPA00008312A (en) Enhancing the circulating half-life of antibody-based fusion proteins
CA2131355C (en) Production of a single-gene-encoded immunoglobulin
US11958913B2 (en) Polypeptide linker for preparing multispecific antibodies

Legal Events

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