EP1060194A1 - 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
EP1060194A1
EP1060194A1 EP99908399A EP99908399A EP1060194A1 EP 1060194 A1 EP1060194 A1 EP 1060194A1 EP 99908399 A EP99908399 A EP 99908399A EP 99908399 A EP99908399 A EP 99908399A EP 1060194 A1 EP1060194 A1 EP 1060194A1
Authority
EP
European Patent Office
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.)
Ceased
Application number
EP99908399A
Other languages
German (de)
English (en)
French (fr)
Inventor
Stephen D. Gillies
Kin-Ming Lo
Yan Lan
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 EP1060194A1 publication Critical patent/EP1060194A1/en
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

Definitions

  • 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.
  • 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: (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 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.
  • 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.
  • 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 Fc ⁇ RI, Fc ⁇ RII or Fc ⁇ RIII.
  • 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.
  • heavy chain isotypes as fusion partners that have reduced binding affinity for Fc receptors on cells.
  • both human IgGl and IgG3 have been reported to bind to FcR ⁇ l 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.
  • 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.
  • 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 - 3 - region of the IgGl or IgG3 heavy chains that reduces the binding affinity of these isotypes for Fc receptors.
  • 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.
  • an antibody-based fusion protein with enhanced in vivo circulating half-life is obtained by first introducing a mutation, deletion, or insertion in the IgGl constant region at one or more amino acid selected from Leu 234 , Leu 235 , Gly 236 , Gly 237 , Asn 297 , and Pro 331 , and then linking the resulting immunoglobulin, or portion thereof, to a second non-immunoglobulin protein.
  • the mutation, deletion, or insertion is introduced in the IgG3 constant region at one or more amino acid selected from Leu 28) , Leu 282 , Gly 283 , Gly 284 , Asn 344 , and Pro 378 , 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.
  • the second non-immunoglobulin component of the fusion protein is a cytokine.
  • 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.
  • 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 TNF ⁇ , and lymphokines such as lymphotoxin.
  • IL-2 interleukin-2
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • TNF tumor necrosis factor
  • lymphokines such as lymphotoxin.
  • the antibody-cytokine fusion protein of the present invention displays cytokine biological activity.
  • the second non-immunoglobulin component of the fusion protein is a ligand-binding protein with biological activity.
  • ligand-binding proteins may, for example, (1) block receptor-ligand interactions at the cell surface; or (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.
  • ligand-binding proteins include CD4, CTLA-4, TNF receptors, or interleukin receptors such as the IL-1 and IL-4 receptors.
  • the antibody-receptor fusion protein of the present invention displays the biological activity of the ligand-binding protein.
  • the second non-immunoglobulin component of the fusion protein is a protein toxin.
  • the antibody-toxin fusion protein of the present invention displays the toxicity activity of the protein toxin.
  • 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 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 CHI, CH2, and/or CH3 domains.
  • fusion protein also embraced within the term "fusion protein” are constructs having a binding domain comprising framework regions 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.
  • 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 non- immunoglobulin protein can exert a localized biological effect.
  • 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 non- immunoglobulin protein, but which lack the antigen-binding specificity and activity of an antibody.
  • 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).
  • 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.
  • - 5 The invention also features DNA constructs encoding the above-described fusion proteins, and cell lines, e.g., myelomas, transfected with these constructs.
  • FIG. 1 is a homology alignment of the amino acid sequences of the constant region of C ⁇ l and C ⁇ 3, 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 C ⁇ l, C ⁇ 2, and C ⁇ 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-KS ⁇ l-IL2 and hu-KS ⁇ 4-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-KS ⁇ l-IL2 (closed diamond) and hu-KS ⁇ 4-IL2 (closed triangle) and of intact fusion protein of hu-KS ⁇ l-IL2 (open diamond) and hu-KS ⁇ 4-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-KS ⁇ l-IL2 (0); mutated hu-KS ⁇ l-IL2 (ffl) and hu-KS ⁇ 4-IL2 ( ⁇ ) as a function of time. - 6 - Detailed Description of the Invention
  • 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.
  • 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.
  • IgGl C ⁇ l
  • IgG3 C ⁇ 3
  • IgG4 C ⁇ 4
  • IgG2 C ⁇ 2
  • an antibody-based fusion protein with reduced binding affinity for a Fc receptor could be obtained by constructing a fusion protein with a C ⁇ 2 constant region (Fc region) or a C ⁇ 4 Fc region, and avoiding constructs with a C ⁇ l Fc region or a C ⁇ 3 Fc region.
  • 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.
  • IgG molecules interact with three classes of Fc receptors (FcR), namely Fc ⁇ RI, Fc ⁇ RII, and Fc ⁇ RIII.
  • FcR Fc receptors
  • C ⁇ l and C ⁇ 3 bind FcR ⁇ l with high affinity
  • C ⁇ 4 and C ⁇ 2 have reduced or no binding affinity for FcR ⁇ l.
  • a comparison of the C ⁇ l and C ⁇ 3 indicates that, with the exception of an extended hinge segment in C ⁇ 3, the amino acid sequence homology between these two isotypes is very high.
  • FIG. 1 provides a alignment of the amino acid sequences of C ⁇ l and C ⁇ 3.
  • the other two isotypes of human IgG (C ⁇ 2 and C ⁇ 4) have sequence differences which have been associated with FcR binding.
  • FIG. 2 provides a alignment of the amino acid sequences of C ⁇ l, C ⁇ 2, and C ⁇ 4.
  • the important - 7 - sequences for Fc ⁇ R binding are Leu-Leu-Gly-Gly (residues 234 through 237 in C ⁇ l), located in the CH2 domain adjacent to the hinge.
  • 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.
  • FcRp protection receptor
  • 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.
  • 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. - 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 C ⁇ l to C ⁇ 4 IgG constant regions.
  • 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.
  • an antibody-IL2 fusion protein with a human C ⁇ l constant region was compared to an antibody-IL2 fusion protein with a human C ⁇ 4 Fc 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 C ⁇ l heavy chain fused to human IL-2, was modified by removing the C ⁇ l gene fragment and replacing it with the corresponding sequence from the human C ⁇ 4 gene.
  • V variable
  • KSA pancarcinoma antigen
  • 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 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.
  • the VH encoding sequence was inserted upstream of the DNA encoding the C ⁇ heavy chain gene of choice, fused to human IL-2 encoding sequences.
  • Such C ⁇ genes contain splice acceptor sites for the first heavy chain exon (CHI), just downstream from a unique Hind III common to all human C ⁇ genes.
  • CHI first heavy chain exon
  • 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 C ⁇ l and C ⁇ 4 fragments was accomplished by digesting the original C ⁇ l -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 C ⁇ 4 gene was digested with Hind III and Nsi I and the 1.75 kb fragment was purified.
  • mouse and human cell lines express one or more Fc receptor.
  • the mouse J774 macrophage-like cell line expresses FcR ⁇ l that is capable of binding mouse or human IgG of the appropriate subclasses.
  • the human K562 erythroleukemic cell line - 10 - expresses FcR ⁇ ll but not FcR ⁇ l.
  • the binding affinities of an antibody, a C ⁇ l-IL2 fusion protein, and a C ⁇ 4-IL2 fusion protein for FcR ⁇ l were compared in the mouse J774 cell line.
  • FACS fluorescence-activated cell sorter
  • 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 - 11 - for an Fc receptor will be identified as antibody-based fusion proteins with enhanced in vivo half lives.
  • fusion proteins containing the C ⁇ l isotype heavy chain i.e., hu-KS ⁇ l-IL2
  • fusion proteins containing the C ⁇ 4 isotype heavy chain were compared to fusion proteins containing the C ⁇ 4 isotype heavy chain (i.e., hu-KS ⁇ 4-IL2).
  • KS-1/4-IL2 fusion proteins containing either the C ⁇ l or C ⁇ 4 isotype heavy chain were buffer-exchanged by diafiltration into phosphate buffered saline (PBS) and diluted further to a concentration of -100 ⁇ g/ml.
  • PBS phosphate buffered saline
  • 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°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.
  • HRP horse radish peroxidase
  • the ⁇ phase half-life of the hu-KS ⁇ 4-IL2 fusion protein was significantly longer than the ⁇ phase half-life of the hu-KS ⁇ l-IL2 fusion protein.
  • the increased half-life is best exemplified by the significantly higher concentrations of the hu-KS ⁇ 4-IL2 fusion - 12 - protein (3.3 ⁇ g/ml) compared to the hu-KS ⁇ l-IL2 fusion protein (60 ng/ml) found in mice after 24 hours.
  • the hu-KS ⁇ l-IL2 protein had a rapid distribution ( ⁇ ) phase followed by a slower catabolic ( ⁇ ) phase, as reported earlier for the chimeric 14.18-IL2 fusion protein.
  • rapid distribution
  • catabolic
  • 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 physically linked).
  • a fusion protein-specific ELISA i.e., an ELISA that requires that both the antibody and IL-2 components be physically linked.
  • C ⁇ l and C ⁇ 3 have binding affinity for Fc receptors, whereas while C ⁇ 4 has reduced binding affinity and C ⁇ 2 has no binding affinity for Fc receptors.
  • the present Example described methods for producing antibody-based fusion proteins using the C ⁇ 4 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 C ⁇ 2 Fc region, instead of the C ⁇ 4 Fc region, in order to enhance the circulating half-life of fusion proteins.
  • a Hu-KS-IL2 fusion protein utilizing the human C ⁇ 2 region can be constructed using the same restriction fragment replacement and the above-described methods for C ⁇ 4-IL2 fusion protein, and tested using the methods described herein to demonstrate increased circulating half-life.
  • Antibody-based fusion proteins with the C ⁇ 2 Fc region, or any other Fc region having reduced binding affinity or lacking binding affinity for a Fc receptor will have enhanced in vivo - 13 - circulating half-life compared to antibody-based fusion proteins having binding affinity for a Fc receptor.
  • Example 2 Mutating the human C ⁇ l or C ⁇ 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 (e.g., Clq fragment), as well as the three classes of FcR.
  • the important residues for Clq binding are residues Glu 318 , Lys 320 , and Lys 322 which are located in the CH2 domains of human heavy chains.
  • 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 C ⁇ l 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. 6).
  • 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 C ⁇ l gene as the template.
  • the cycle conditions for the primary PCR were 35 cycles of: 94°C for 45 sec, annealing at 48°C for 45 seconds, and primer extension at 72°C for 45 sec.
  • each PCR reaction was 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 72°C 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.
  • a cloning vector containing the human C ⁇ l gene was digested with Pst I and Xho I to remove the - 14 - non-mutated hinge-CH2-CH3 coding sequences.
  • CH2 all of CH3 and the fused human IL-2 coding sequences was prepared from the C ⁇ l-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 C ⁇ l-IL2 gene was used to reassemble the complete hu-KS antibody-IL2 fusion protein expression vector.
  • the in vivo plasma concentration of the mutated hu-KS ⁇ l-IL2 was compared to the plasma concentration of hu-KS ⁇ l-IL2 at various specified times. As illustrated in FIG. 7, the in vivo clearance rates of the mutated hu-KS ⁇ l-IL2 and hu- S ⁇ 4-IL2 were significantly lower than the clearance rate of hu-KS ⁇ l-IL2.
  • mutations to the C ⁇ l or C ⁇ 3 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.
  • mutations can also be introduced into the C ⁇ 4 gene in order to further reduce the binding of C ⁇ 4 fusion proteins to FcR.
  • additional possible mutations include mutations in the hinge proximal amino acid residues, mutating Pro 331 , 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 amino acid Gin would have little effect on the protein but would prevent the - 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.
  • Example 3 Increasing the circulating half-life of receptor-antibody-based fusion proteins.
  • 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
  • ligand-binding protein partner i.e., the second non-Ig protein
  • the ligand-binding protein partner i.e., the second non-Ig protein
  • the biological activity of a molecule e.g., a cytokine
  • receptor-antibody-based fusion proteins with human C ⁇ l Fc regions are compared to antibody- based fusion proteins with human C ⁇ 4 Fc regions.
  • 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).
  • PBMC peripheral blood monocytic cells
  • the expression vector contains a unique Xba I cloning site downstream of the CMV early promoter, and the human C ⁇ l or C ⁇ 4 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-C ⁇ l and CD4-C ⁇ 4.
  • 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 ⁇ M).
  • - 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 ⁇ g/ml.
  • Balb/c mice are injected with 0.2 ml (20 ⁇ g) of either the CD4-C ⁇ l or CD4-C ⁇ 4 fusion protein and the pharmacokinetics are tested as described in Example 1.3.
  • the CD4-C ⁇ 4 fusion protein has a significantly greater half-life than the CD4-C ⁇ l fusion protein.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
EP99908399A 1998-02-25 1999-02-24 Enhancing the circulating half-life of antibody-based fusion proteins Ceased EP1060194A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US7588798P 1998-02-25 1998-02-25
US75887P 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 (1)

Publication Number Publication Date
EP1060194A1 true EP1060194A1 (en) 2000-12-20

Family

ID=22128576

Family Applications (1)

Application Number Title Priority Date Filing Date
EP99908399A Ceased EP1060194A1 (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 (hu)
JP (1) JP2002505086A (hu)
CN (1) CN1204147C (hu)
AU (1) AU758240B2 (hu)
BR (1) BR9908226A (hu)
CA (1) CA2320403A1 (hu)
CZ (1) CZ20003099A3 (hu)
HK (1) HK1036286A1 (hu)
HU (1) HUP0100813A3 (hu)
NO (1) NO20004218L (hu)
PL (1) PL199659B1 (hu)
WO (1) WO1999043713A1 (hu)

Families Citing this family (130)

* 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
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
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
US6737056B1 (en) 1999-01-15 2004-05-18 Genentech, Inc. Polypeptide variants with altered effector function
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
PL209786B1 (pl) 1999-01-15 2011-10-31 Genentech Inc Przeciwciało zawierające wariant regionu Fc ludzkiej IgG1, przeciwciało wiążące czynnik wzrostu śródbłonka naczyń oraz immunoadhezyna
GB9926084D0 (en) * 1999-11-03 2000-01-12 King S College London Recombinant fusion molecules
ES2269366T3 (es) * 2000-02-11 2007-04-01 Merck Patent Gmbh Mejoramiento de la vida media en circulacion de proteinas de fusion basadas en anticuerpos.
EP1272526A4 (en) 2000-04-13 2004-10-13 Univ Rockefeller REINFORCING ANTIBODY-IMMUNE RESPONSE
DE10045591A1 (de) * 2000-09-15 2002-04-04 Klaus Pfizenmaier Ortsspezifische, antikörpervermittelte Aktivierung proapoptotischer Zytokine - AMAIZe (Antibody-Mediated Apoptosis Inducing Zytokine)
EP2357187A1 (en) * 2000-12-12 2011-08-17 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
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
DK1366067T3 (da) * 2001-03-07 2012-10-22 Merck Patent Gmbh Ekspressionsteknologi for proteiner indeholdende en hybrid isotype-antistof-enhed
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
EP1495055B1 (en) * 2002-04-18 2013-08-14 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
US8968730B2 (en) 2002-08-14 2015-03-03 Macrogenics Inc. FcγRIIB specific antibodies and methods of use thereof
US8946387B2 (en) 2002-08-14 2015-02-03 Macrogenics, Inc. FcγRIIB specific antibodies and methods of use thereof
PT1562972E (pt) * 2002-10-15 2010-11-10 Facet Biotech Corp Alteração de afinidades de ligação ao fcrn ou semi-vidas séricas de anticorpos por mutagénese
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
US7355008B2 (en) 2003-01-09 2008-04-08 Macrogenics, Inc. Identification and engineering of antibodies with variant Fc regions and methods of using same
EP2298347B1 (en) 2003-05-06 2015-09-30 Biogen Hemophilia Inc. Clotting factor chimeric proteins for treatment of a hemostatic disorder
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
JP2005073528A (ja) * 2003-08-28 2005-03-24 Genetics Inst Llc 生物学的系における生物学的組織への血球の接着を阻害する方法、ならびにその方法に使用するための組成物
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
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 EFFECTOR FUNCTION OBTAINED BY CREATION BY BIOLOGICAL GENE OF FC ANTIBODY REGIONS
US9284375B2 (en) 2005-04-15 2016-03-15 Macrogenics, Inc. Covalent diabodies and uses thereof
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
US9296816B2 (en) 2005-04-15 2016-03-29 Macrogenics, Inc. Covalent diabodies and uses thereof
EP2298815B1 (en) 2005-07-25 2015-03-11 Emergent Product Development Seattle, LLC B-cell reduction using CD37-specific and CD20-specific binding molecules
DK1919503T3 (en) 2005-08-10 2014-12-15 Macrogenics Inc Identification and preparation of antibodies with variant fc regions and methods of use thereof
WO2007146968A2 (en) 2006-06-12 2007-12-21 Trubion Pharmaceuticals, Inc. Single-chain multivalent binding proteins with effector function
HUE030269T2 (hu) 2006-06-26 2017-04-28 Macrogenics Inc FC RIIB-specifikus ellenanyagok és eljárások az alkalmazásukra
EP2032159B1 (en) 2006-06-26 2015-01-07 MacroGenics, Inc. Combination of fcgammariib antibodies and cd20-specific antibodies and methods of use thereof
ATE480568T1 (de) * 2006-06-30 2010-09-15 Conaris Res Inst Ag Verbesserte sgp 130fc dimere
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
PE20120259A1 (es) 2007-08-09 2012-04-04 Boehringer Ingelheim Int Anticuerpos anti-cd37
JP5809415B2 (ja) 2007-11-09 2015-11-10 ペレグリン ファーマシューティカルズ,インコーポレーテッド 抗vegf抗体の組成物および方法
WO2009117030A2 (en) 2007-12-19 2009-09-24 Macrogenics, Inc. Improved compositions for the prevention and treatment of smallpox
CA2711736A1 (en) 2008-01-18 2009-07-23 Medimmune, Llc Cysteine engineered antibodies for site-specific conjugation
KR101614494B1 (ko) 2008-04-02 2016-04-22 마크로제닉스, 인크. Bcr-복합체-특이적 항체 및 그것의 사용 방법
JP5555223B2 (ja) 2008-04-02 2014-07-23 マクロジェニクス,インコーポレーテッド HER2/neu特異的抗体およびその使用方法
NZ603059A (en) 2008-04-11 2014-07-25 Emergent Product Dev Seattle Cd37 immunotherapeutic and combination with bifunctional chemotherapeutic thereof
TWI440469B (zh) 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
AU2010225951B2 (en) * 2009-03-19 2014-03-13 Chugai Seiyaku Kabushiki Kaisha Pharmaceutical formulation containing improved antibody molecules
EP2233500A1 (en) 2009-03-20 2010-09-29 LFB Biotechnologies Optimized Fc variants
EP2421896A1 (en) 2009-04-22 2012-02-29 Merck Patent GmbH Antibody fusion proteins with modified fcrn binding sites
US20120134984A1 (en) 2009-06-01 2012-05-31 Olga Lubman Molecules with extended half-lives and uses thereof
CA2766405A1 (en) 2009-06-22 2011-01-13 Medimmune, Llc Engineered fc regions for site-specific conjugation
RU2583298C2 (ru) 2009-10-07 2016-05-10 Макродженикс, Инк. ПОЛИПЕПТИДЫ, СОДЕРЖАЩИЕ Fc-УЧАСТОК, КОТОРЫЕ ДЕМОНСТРИРУЮТ ПОВЫШЕННУЮ ЭФФЕКТОРНУЮ ФУНКЦИЮ БЛАГОДАРЯ ИЗМЕНЕНИЯМ СТЕПЕНИ ФУКОЗИЛИРОВАНИЯ, И СПОСОБЫ ИХ ПРИМЕНЕНИЯ
CA2782320A1 (en) 2009-12-02 2011-06-09 Acceleron Pharma Inc. Compositions and methods for increasing serum half-life of fc fusion proteins
TWI505838B (zh) 2010-01-20 2015-11-01 Chugai Pharmaceutical Co Ltd Stabilized antibody solution containing
AU2011223782B2 (en) 2010-03-04 2014-09-18 Macrogenics, Inc. Antibodies reactive with B7-H3, immunologically active fragments thereof and uses thereof
US8802091B2 (en) 2010-03-04 2014-08-12 Macrogenics, Inc. Antibodies reactive with B7-H3 and uses thereof
CA2807127C (en) 2010-08-02 2019-02-12 Leslie S. Johnson Covalent diabodies and uses thereof
UA112062C2 (uk) 2010-10-04 2016-07-25 Бьорінгер Інгельхайм Інтернаціональ Гмбх Cd33-зв'язувальний агент
TR201810773T4 (tr) 2010-11-23 2018-08-27 Glaxo Group Ltd Onkostatin m?ye (osm) antijen bağlayıcı proteinler.
CN103459422A (zh) 2010-11-24 2013-12-18 葛兰素集团有限公司 靶向hgf的多特异性抗原结合蛋白
EP2654790B1 (en) 2010-12-22 2019-02-06 Teva Pharmaceuticals Australia Pty Ltd Modified antibody with improved half-life
PT2691417T (pt) 2011-03-29 2018-10-31 Roche Glycart Ag Variantes de fc de anticorpos
EA201892619A1 (ru) 2011-04-29 2019-04-30 Роше Гликарт Аг Иммуноконъюгаты, содержащие мутантные полипептиды интерлейкина-2
UA111612C2 (uk) 2011-05-21 2016-05-25 Макродженікс, Інк. Домени, які зв'язуються з деімунізованою сироваткою, і їхнє застосування для збільшення часу напівжиття в сироватці
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
BR112014008804A2 (pt) 2011-10-11 2017-06-13 Medimmune Llc suportes derivados de tn3 específicos para cd40l e métodos de uso dos mesmos
CA2859755C (en) 2011-12-23 2021-04-20 Pfizer Inc. Engineered antibody constant regions for site-specific conjugation and methods and uses therefor
HUE056217T2 (hu) 2012-07-13 2022-02-28 Roche Glycart Ag Bispecifikus anti-VEGF/anti-ANG-2 antitestek és ezek alkalmazása szemészeti érbetegségek kezelésében
SG11201408530YA (en) * 2012-08-02 2015-03-30 Hoffmann La Roche 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
WO2014029752A1 (en) 2012-08-22 2014-02-27 Glaxo Group Limited Anti lrp6 antibodies
TWI682941B (zh) 2013-02-01 2020-01-21 美商再生元醫藥公司 含嵌合恆定區之抗體
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
DK2968520T3 (da) 2013-03-14 2021-08-09 Macrogenics Inc Bispecifikke molekyler som er immunoreaktive med immuneffektorceller der udtrykker en aktiverende receptor
UA118750C2 (uk) 2013-03-15 2019-03-11 Ґлаксосмітклайн Інтеллектуал Проперті Дивелопмент Лімітед Антитіло до lag-3
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 (uk) 2013-08-09 2018-03-26 Макродженікс, Інк. БІСПЕЦИФІЧНЕ МОНОВАЛЕНТНЕ Fc-ДІАТІЛО, ЯКЕ ОДНОЧАСНО ЗВ'ЯЗУЄ CD32B I CD79b, ТА ЙОГО ЗАСТОСУВАННЯ
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
NZ719654A (en) 2014-02-06 2020-06-26 Hoffmann La Roche Interleukin-2 fusion proteins and uses thereof
GB201403775D0 (en) 2014-03-04 2014-04-16 Kymab Ltd Antibodies, uses & methods
TWI754319B (zh) 2014-03-19 2022-02-01 美商再生元醫藥公司 用於腫瘤治療之方法及抗體組成物
ES2869459T3 (es) 2014-05-16 2021-10-25 Medimmune Llc Moléculas con unión a receptor de fc de neonato alterada que tiene propiedades terapéuticas y de diagnóstico potenciadas
US10562946B2 (en) 2014-06-20 2020-02-18 Genentech, Inc. Chagasin-based scaffold compositions, methods, and uses
KR20170035910A (ko) 2014-08-11 2017-03-31 데리니아, 인크. 자가면역 질환의 치료를 위한 조절 t 세포를 선택적으로 활성화시키는 변형된 il-2 변이체
WO2016054101A1 (en) 2014-09-29 2016-04-07 Duke University Bispecific molecules comprising an hiv-1 envelope targeting arm
PL3221359T3 (pl) 2014-11-17 2020-11-16 Regeneron Pharmaceuticals, Inc. Sposoby leczenia nowotworów przy użyciu dwuswoistego przeciwciała CD3XCD20
KR101892883B1 (ko) 2015-02-27 2018-10-05 추가이 세이야쿠 가부시키가이샤 Il-6 관련 질환 치료용 조성물
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
WO2017019729A1 (en) 2015-07-27 2017-02-02 The General Hospital Corporation Antibody derivatives with conditionally enabled effector function
EP3798234A1 (en) 2015-09-02 2021-03-31 Immutep S.A.S. Anti-lag-3 agonistic antibodies
TWI799366B (zh) 2015-09-15 2023-04-21 美商建南德克公司 胱胺酸結骨架平臺
SG10202002577XA (en) 2015-09-21 2020-04-29 Aptevo Res & Development Llc Cd3 binding polypeptides
WO2017093947A1 (en) 2015-12-04 2017-06-08 Novartis Ag Antibody cytokine engrafted compositions and methods of use for immunoregulation
US20170204154A1 (en) 2016-01-20 2017-07-20 Delinia, Inc. Molecules that selectively activate regulatory t cells for the treatment of autoimmune diseases
TWI781098B (zh) 2016-04-15 2022-10-21 美商宏觀基因股份有限公司 新穎的b7-h3-結合分子、其抗體藥物綴合物及其使用方法
US9567399B1 (en) 2016-06-20 2017-02-14 Kymab Limited Antibodies and immunocytokines
WO2018049275A1 (en) 2016-09-09 2018-03-15 Genentech, Inc. Selective peptide inhibitors of frizzled
WO2018073365A1 (en) * 2016-10-19 2018-04-26 F. Hoffmann-La Roche Ag Method for producing an immunoconjugate
WO2018083248A1 (en) 2016-11-03 2018-05-11 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses & methods
EP3538548A4 (en) 2016-11-08 2020-08-19 Delinia, Inc. IL-2 VARIANTS FOR THE TREATMENT OF AUTOIMMUNE DISEASES
WO2018203545A1 (ja) 2017-05-02 2018-11-08 国立研究開発法人国立精神・神経医療研究センター Il-6及び好中球の関連する疾患の治療効果の予測及び判定方法
JOP20190271A1 (ar) 2017-05-24 2019-11-21 Novartis Ag بروتينات مطعّمة بسيتوكين- الجسم المضاد وطرق الاستخدام للاضطرابات المتعلقة بالمناعة
EP3694885A1 (en) 2017-10-14 2020-08-19 CytomX Therapeutics, Inc. Antibodies, activatable antibodies, bispecific antibodies, and bispecific activatable antibodies and methods of use thereof
MX2020006322A (es) 2017-12-19 2020-09-18 Xencor Inc Proteinas de fusion il-2 fc modificadas.
KR20210054528A (ko) 2018-08-31 2021-05-13 리제너론 파아마슈티컬스, 인크. Cd3/c20 이중특이적 항체에 대한 사이토카인 방출 증후군을 경감시키는 투약 전략
CN112142847B (zh) * 2019-02-22 2023-05-05 武汉友芝友生物制药股份有限公司 改造的Fc片段,包含其的抗体及其应用
CN114502593A (zh) 2019-08-06 2022-05-13 葛兰素史密斯克莱知识产权发展有限公司 生物药物组合物和相关方法
EP4347656A1 (en) 2021-05-28 2024-04-10 GlaxoSmithKline Intellectual Property Development Limited Combination therapies for treating cancer
WO2023012669A2 (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
WO2024083945A1 (en) 2022-10-20 2024-04-25 Glaxosmithkline Intellectual Property (No.3) Limited Antigen binding proteins
US20240166728A1 (en) 2022-11-02 2024-05-23 VIIV Healthcare UK (No.5) Limited Antigen binding proteins

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

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9943713A1 *

Also Published As

Publication number Publication date
WO1999043713A1 (en) 1999-09-02
AU2784299A (en) 1999-09-15
AU758240B2 (en) 2003-03-20
CA2320403A1 (en) 1999-09-02
HUP0100813A2 (hu) 2001-06-28
PL342497A1 (en) 2001-06-04
JP2002505086A (ja) 2002-02-19
HUP0100813A3 (en) 2003-08-28
CZ20003099A3 (cs) 2002-04-17
PL199659B1 (pl) 2008-10-31
CN1291995A (zh) 2001-04-18
NO20004218L (no) 2000-10-24
NO20004218D0 (no) 2000-08-23
CN1204147C (zh) 2005-06-01
HK1036286A1 (en) 2001-12-28
BR9908226A (pt) 2000-10-24

Similar Documents

Publication Publication Date Title
AU758240B2 (en) Enhancing the circulating half-life of antibody-based fusion proteins
US20060194952A1 (en) Enhancing the circulating half-life of antibody-based fusion proteins
JP4234438B2 (ja) ハイブリッド・イソタイプ抗体部分を含有する蛋白質の発現技術
US5892019A (en) Production of a single-gene-encoded immunoglobulin
US7053202B2 (en) Immunoglobulin DNA cassette molecules, monobody constructs, methods of production, and methods of use therefor
EP1514934B1 (en) Biosynthetic binding protein for cancer marker
JP5572665B2 (ja) 抗体ベース融合タンパク質の循環系内半減期の増強
JP3081641B2 (ja) 抗体の調製
CA1341351C (en) Production of chimeric mouse-human antibodies with specificity to human tumor antigens
AU2002248571A1 (en) Expression technology for proteins containing a hybrid isotype antibody moiety
CA2328606A1 (en) Antibodies to cd23, derivatives thereof, and their therapeutic uses
CA2582157A1 (en) Methods and compositions for improving recombinant protein production
TW201932142A (zh) 自單特異性抗體產生多特異性抗體之方法
Shin et al. Hybrid antibodies
WO1995014779A1 (en) Mutated hig-e fragments and derivative thereof
US20210253741A1 (en) Bispecific antibodies
CA2131355C (en) Production of a single-gene-encoded immunoglobulin
MXPA00008312A (es) Metodos para mejorar la vida media en circulacion de proteinas de fusion a base de anticuerpo
AU2001243148B2 (en) Enhancing the circulating half-life of antibody-based fusion proteins
AU2001243148A1 (en) Enhancing the circulating half-life of antibody-based fusion proteins

Legal Events

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

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20001011

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LI LU NL PT SE

AX Request for extension of the european patent

Free format text: LT PAYMENT 20001011;LV PAYMENT 20001011;RO PAYMENT 20001011;SI PAYMENT 20001011

17Q First examination report despatched

Effective date: 20030804

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

Owner name: MERCK PATENT GMBH

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

Free format text: STATUS: THE APPLICATION HAS BEEN REFUSED

18R Application refused

Effective date: 20090925