AU597574B2 - Method for enhancing glycoprotein stability - Google Patents

Method for enhancing glycoprotein stability Download PDF

Info

Publication number
AU597574B2
AU597574B2 AU56271/86A AU5627186A AU597574B2 AU 597574 B2 AU597574 B2 AU 597574B2 AU 56271/86 A AU56271/86 A AU 56271/86A AU 5627186 A AU5627186 A AU 5627186A AU 597574 B2 AU597574 B2 AU 597574B2
Authority
AU
Australia
Prior art keywords
glcnac
protein
gal
endo
oligosaccharide
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
AU56271/86A
Other versions
AU5627186A (en
Inventor
Michel Louis Eugene Bergh
S. Catherine Hubbard
James R. Rasmussen
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.)
Massachusetts Institute of Technology
Original Assignee
Massachusetts Institute of Technology
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 Massachusetts Institute of Technology filed Critical Massachusetts Institute of Technology
Publication of AU5627186A publication Critical patent/AU5627186A/en
Application granted granted Critical
Publication of AU597574B2 publication Critical patent/AU597574B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • C07K1/1072General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups
    • C07K1/1077General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups by covalent attachment of residues other than amino acids or peptide residues, e.g. sugars, polyols, fatty acids

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Chemical & Material Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)

Description

AIJ-AI-56271/86 WORLD INELETUrAin G'7ATI t INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOIO N TREATY (PCT) (51) International Patent Classification 4 (11) International Publication Number: WO 87/ 05330 C12P 21/00, C07K 15/14 Al (43) International Publication Date: I1I September 1987 (11.09.87) (21I) International Application Number: PCT/U586/00495 International Filing Date: 7 March 1986 (07.03.86) Published SECTION 34(4)(a) DIRECTION SEE FOLIO 2 Al NAME DIREOTEDPG~Sa14U In4Ae
Q-
-7 -1c0i3as u'aPM- 74Agents: PABST,- Patrea, L.;_Kenway enney, Street, Boston, MA 02109 (US) et al.
(81) Designated States: AT (European patent), AU, BE (Eu- 2 C 1987 ropean patent), CH (European patent), DE (European patent), DK, Fl, FR (European patent), GB (European patent), HU, IT (European patent), JP, KR, LU (European patent), MC, NL (European patent), NO, AUSTRALIAN RO, SE (European patent), SU. 28 SEP 1987 IThis document contains the PAETOFC [amendments made under PAETOFC ,Section 49 and is correct for printing.
(54)Title: METHOD FOR ENHANCING GLYCOPROTEIN STABILITY Abstract 0 II (57) bstrat A Le A method for modifly- A/~e.4 ing eukaryotic and prokaryot- *l&4i 1 ic proteins to extend their in 0 ATe vivo circulatory lifetimes. In Mi" A4 the preferred embodiment, 0AIISri' 0AI400 enzymatic and/or chemical AIO/ S treatments are used to pro- wrduce a modified protein, car- rying. one or more covalently IL AISDS attached trisaccharide, sialic I wxn Iacid-- galactose-- N-ace-
#-AIASF
tylglucosamine-- /MAV.e Gal-- GlcNAc-- or tet-01/ocCAIO rasaccharide Gal--
NISD~
GlcNAc-- GlcNAcbe applied to any natural or fileosri- recombinant protein possess- J.14 n ing asparag ine-linked oligos- rVfXf accharides or to any'nont-gly- II cosylated protein that can be SAlUC/i/fs chemically or enzymatically emnetr oiloOSACCnalOs derivatized with the approprk ate carbohydrate units. Following injection into -an an imal, the modified glycoproteins are protected from premature clearenice by cells of the liver and reticulo-endothelial system which recognize and rapidly internalize circulating glycoproteins with, carbohydrate containing terminal Gal,~ GlcNAc, fucose or, mannose residues. The method can also be used to mask anitigenic determinantF, on foreign proteins -which would otherwise produce an immune response or to "target" a protein for recognition by. sugar-specific cell, surface receptors.
_A14 -1 WO 87/05330 PCT/US86/00495 METHOD FOR ENHANCING GLYCOPROTEIN STABILITY S' BACKGROUND OF THE INVENTION The United States Government has certain rights in this invention by virtue of National Institutes of Health grants No. CA26712, GN31318, and CA14051.
Glycoproteins, proteins with covalently bound sugars, are found in plants, animals, insects, and even many unicellular eukaryotes such as yeast. They occur within cells in both soluble and membrane-bound forms, in the intercellular matrix, and in extracellular fluids. The carbohydrate moieties of these glycoproteins can participate directly in the biological activity of the glycoproteins in a variety of ways: protection from proteolytic degradation, stabilization of protein conformation, and mediation of inter- and intracellular recognition.
Examples of glycoproteins include enzymes, serum proteins such as immunoglobulins and blood clotting factors, cell surface receptors for growth factors and infectious agents, hormones, toxins, lectins and structural proteins.
Natural and recombinant proteins are being used as therapeutic agents in humans and animals. In many cases a therapeutic protein will be most efficacious if it has an appreciable circulatory lifetime. At least four general mechanisms can contribute to a shortened circulatory lifetime for an exogenous protein: proteolytic degradation, clearance by the immune system if the protein is antigenic or immunogenic, clearance by cells of the liver or reticulo-endothelial system that recognize specific exposed sugar units on a glycoprotein, and clearance through the glomerular basement membrane of the kidney if the protein is of low molecular weight. The oligosaccharides of a glycoprotein can exert a strong effect on the first three of these clearance mechanisms.
S; The oligosaccharide chains of glycoproteins are attached to the polypeptide backbone by either N- or C-glycosidic linkages. In the case of N-linked glycans, there is an amide bond connecting the anomeric carbon of a reducing-terminal N-acetylglucosamine (GlcNAc) residue of the oligosaccharide and a nitrogen of an aspara- 4 WO 87/05330 PCT/US86/00495 -2gine (Asn) residue of the polypeptide. In animal cells, 0-linked glycans are attached via a glycosidic bond between N-acetylgalactosamine (GalNAc), galactose (Gal), or xylose and one of several hydroxyamino acids, most commonly serine (Ser) or threonine (Thr), but also hydroxyproline or hydroxylysine in some cases. The 0-linked glycans in the yeast Saccharomyces cerevisiae are also attached to serine or threonine residues, but, unlike the glycans of animals, they consist of one to several a-linked mannose (Man) residues. Mannose residues have not been found in the O-linked oligosaccharides of animal cells.
The biosynthetic pathways of N- and 0-linked oligosaccharides are quite different. 0-Linked glycan synthesis is relatively simple, consisting of a step-by-step transfer of single sugar residues from nucleotide sugars by a series of specific glycosyltransferases. The nucleotide sugars which function as the monosaccharide donors are uridine-diphospho-GalNAc (UDP-GalNAc), UDP-GlcNAc, UDP-Gal, guanidinediphospho-fucose (GDP-Fuc), and cytidine-monophospho-sialic acid (CMP-SA). N-Linked oligosaccharide synthesis, which is much more complex, is described below.
The initial steps in the biosynthesis of N-linked glycans have been preserved with little change through evolution from the level of unicellular eukaryotes such as yeast to higher plants and man. For all of these organisms, initiation of N-linked oligosaccharide assembly does not occur directly on the Asn residues of the protein, but rather involves preassembly of a lipid-linked precursor oligosaccharide which is then transferred to the protein during or very soon after its translation from mRNA. This precursor oligosaccharide, which has the composition Glc 3 Man GlcNAc 2 and the structure shown in Fig. 1A, is synthesized while attached via a pyrophosphate bridge to a polyisoprenoid carrier lipid, a dolichol. This assembly is a complex process involving at least six distinct membrane-bound glycosyltransferases. Some of these enzymes transfer monosaccharides from nucleotide sugars, while others utilize dolichol-linked monosaccharides as sugar donors. After assembly of the lipid-linked precursor is complete, another membrane-bound enzyme transfers it to sterically accessible Asn residues which occur as part of the sequence -Asn-X- Ser/Thr-. The requirement for steric accessibility is presumably responsible for the observation that denaturation is usually required a- 1
'W
WO 87/05330 PCT/US86/00495 -3for in vitro transfer of precursor oligosaccharide to exogenous proteins.
Glycosylated Asn residues of newly-synthesized glycoproteins transiently carry only one type of oligosaccharide, Glc 3 ManoGlcNAc 2 Modification, or "processing," of this structure generates the great diversity of structures found on mature glycoproteins, and it is the variation in the type or extent of this processing which accounts for the observation that different cell types often glycosylate even the same polypeptide differeltly.
The processing of N-linked oligosaccharides is accomplished.by the sequential action of a number of membrane-bound enzymes and begins immediately after transfer of the precursor oligosaccharide Glc 3 ManQ- GlcNAc 2 to the protein. In broad terms, N-linked oligosaccharide processing can be divided into three stages: removal of the three glucose residues, removal of a variable number of mannose residues, and addition of various sugar residues to the resulting trimmed "core," the Man 3 GlcNAc 2 portion of the original oligosaccharide closest to the polypeptide backbone. A simplified outline of tne processing pathway is shown in Fig. 2.
Like the assembly of the precursor oligosaccharide, the removal of the glucose residues in the first stage of processing has been preserved through evolution. In yeast and in vertebrates, all three glucose residues are trimmed to generate N-linked MangGlcNAc. Processing sometimes stops with this structure, but usually it continues to the second stage with removal of mannose residues. Here the pathway for yeast diverges from that in vertebrate cells.
As shown in Fig. 1B, four of tne mannose residues of the MangGlcNAc 2 moiety are bound by al-->2 linkages. By convention the arrow points toward the reducing terminus of an oligosaccharide, or in this case, toward the protein-bound end of the glycan; a or B indicate the anomeric configuration of the glycosidic bond; and the two numbers indicate which carbon atoms on each monosaccharide are involved in the bond. The four al-->2-linked mannose residues can be removed by Mannosidase I to generate N-linked Mans- 8 GlcNAc 2 all of which are commonly found on vertebrate glycoproteins. Oligosaccharides with the composition Man 5 9 GlcNAc 2 are said to be of the "high-mannose" type.
As shown in Fig. 2, protein-linked Man 5 GlcNAc 2 (Structure M-c) can A I WO 87/05330 PCT/US86/00495 -4serve as a substrate for GlcNAc transferase I, which transfers a 1-->2-linked GlcNAc residue from UDP-GlcNAc to the al-->3-linKed mannose residue to form GlcNAcMan 5 GlcNAc 2 (Structure Mannosidase II can then complete the trimming phase of the processing pathway by removing two mannose residues to generate a protein-linked oligosaccharide with the composition GlcNAcMan 3 GlcNAc 2 (Structure M-e).
This structure is a substrate for GlcNAc transferase II, which can transfer a B1-->2-linked GlcNAc residue to the al-->6-linked mannose residue (not shown).
It is at this stage that the true complexity of the processing pathway begins to unfold. Simply stated, monosaccharides are sequentially added to the growing oligosaccharide chain by a series of membrane-bound Golgi glycosyltransferases, each of which is highly specific with respect to the acceptor oligosaccharide, the donor sugar, and the type of linkage formed between the sugars. Each type of cell has an extensive but discrete set of these glycosyltransferases. These can include at least four more distinct GlcNAc transferases (producing or 81-->6 linkages); three galactosyltransferases (producing and al-->3 linkages); two sialyltransferases (one producing c2-->3 and another, a2-->6 linkages); three fucosyltransferases (producing al-->3, 1a-->4 or al-->6 linkages); and a growing list of other enzymes responsible for a variety of unusual linkages. The cooperative action of these glycosyltransferases produces a diverse family of structures collectively referred to as "complex" oligosaccharides. These may contain two (for example, Structure M-f in Fig. three (for example, Fig. 1C or Structure M-g in Fig. or four outer branches Sattached to the invariant core pentasaccharide, Man 3 GlcNAc 2 These structures are referred to in terms of the number of their outer branches: biantennary (two branches), triantennary (three branches) or tetraantennary (four branches). The size of these complex glycans varies from a hexasaccharide (on rhodopsin) to very large polylactosaminylglycans, which contain one or more outer branches with repeating (GaBl'-->4G1cNAcB1-->3) units (on several cell surface glycoproteins such as the erythrocyte glycoprotein Band 3 and the macrophage antigen SMac-2). Despite this diversity, the specificities of the glycosyltransferases do produce some frequently recurring structures. For 1 1 1 1 1 1 1 1 1 1 1 '7 WO 87/05330 87/05330 PCT/US86/00495 example, the outer branches of many complex N-linked oligosaccharides consist of all or part of the sequence SAa2-->3(6)GalBl-->4G1cNAc1-->.
One or two of these trisaccharide moieties may be attached to each of the two a-linked mannose residues of the core pentasaccharide, as in Structures M-f and M-g of Fig. 2.
Unlike transcription of DNA or translation of mRNA, which are highly reproducible events, oligosaccharide biosynthesis does not take place on a template. As a consequence, considerable heterogeneity is usually observed in the oligosaccharide structures of every glycoprotein. The differences are most commonly due to variations in the extent of processing. The single glycosylation site of the chicken egg glycoprotein ovalbumin, for example, contains a structurally related "family" of at least 18 different oligosaccharides, the great majority of which are of the high-mannose or related "hybrid" type (for example, Structure M-h in Fig. Many glycoproteins contain multiple glycosylated Asn residues, and each of these may carry a distinct family of oligosaccnarides. For example, one site may carry predominantly high-mannose glycans, another may carry mostly fucosylated biantennary complex chains, and a third may carry fucose-free tri- and tetraantennary complex structures. Again, all of these glycans will contain the invariant Man.GlcNAc 2 core.
As discussed above, the initial stages of N-linked oligosaccharide synthesis in the yeast Saccharomyces cerevisiae closely resemble those occurring in vertebrate cells. As in higher organisms, lipid-linked Glc3Man 9 GlcNAc 2 is assembled, its oligosaccharide chain transferred to acceptor Asn residues of proteins, and its three glucose res.idues are removed soon after transfer. Yeast cells can remove only a single mannose residue, however, so that the smallest and least-processed N-linked glycans have the composition Man g 9 GlcNAc 2 Processing can stop at this stage or continue with the addition of as many as 50 or more a-linked mannose residues to MangGlcNAc 2 (Fig. 2, Structure Y-c) to generate a mannan (for example, Structure Just as glycoproteins in mammalian cells may have predominantly high-mannose oligosaccharides at one glycosylated Asn residue and highly processed complex glycans at anothe'r, yeast glycoproteins such as external invertase commonly have some glycosylation sites with Man 8 oGlcNAc 2 chains, 4 l" l y iv WO 87/05330 PCT/US86/00495 -6while other sites carry mannans.
Unlike eukaryotic cells, bacteria lack the enzymatic machinery to assemble lipid-linked Glc 3 MangGlcNAc 2 or transfer it to proteins.
Thus, although proteins synthesized in E. coli contain many -Asn-X- Ser/Thr- sequences, they are not glycosylated.
From the foregoing discussion, it is apparent that the glycosylation status of a glycoprotein will depend on the cell in which it is produced. The glycans of a protein synthesized in cultured mammalian cells will resemble those of the same protein isolated from a natural animal source such as a tissue but are unlikely to be identical. Proteins glycosylated by yeast contain high-mannose oligosaccharides and mannans, and proteins synthesized in a bacterium such as E. coli will not be glycosylated because the necessary enzymes are absent.
The precise composition and structure of the carbohydrate chain(s) on a glycoprotein can directly influence its serum lifetime, since cells in the liver and reticulo-endothelial system can bind and internalize circulating glycoproteins with specific carbohydrates. Hepatocytes have receptors on their surfaces that recognize oligosaccharide chains with terminal at the outermost end(s) of glycans relative to the polypeptide) Gal residues, macrophages contain receptors for terminal Man or GlcNAc residues, and hepatocytes and lymphocytes have receptors for exposed fucose residues. No sialic acid-specific receptors have been found, however. Although somewhat dependent on the spatial arrangement of the oligosaccharides, as a general rule, the greater the number of exposed sugar residues recognized by cell surface receptors in the liver and reticulo-endothelial system, the (a more rapidly a glycoprotein will be cleared from the serum. Because of the absence of sialic acid-specific receotors, however, oligosaccharides with all branches-'terminated, or "capped," with sialic acid will not promote the clearance of the protein to which they are attached.
The presence and nature of the oligosaccharide chain(s) on a glycoprotein can also affect important biochemical properties in addition to its recognition by sugar-specific receptors on liver and reticulo-endothelial cells. Removal of the carbohydrate from a glycoprotein will usually decrease its solubility, and it may also increase its susceptibility to proteolytic degradation by destabiq 1 1 i I 1 I I ;j I~ 1 WO 87/05330 PCT/US86/00495 lizing the correct polypeptide folding pattern and/or unmasking protease-sensitive sites. For similar reasons, the glycosylation status of a protein can affect its recognition by the immune system.
It is therefore an objective of the present invention to provide a method for modifying oligosaccharide chains of glycoproteins isolated from natural sources or produced from recombinant DNA in yeast, insect, plant or vertebrate cells in a manner that increases serum lifetime or targets the protein to specific cell types.
It is another objective of the invention to provide an in vitro method for glycosylating proteins produced from bacterial, yeast, plant, viral or animal DNA in a manner that enhances stability and effective biological activity.
It is a further objective of the invention to provide a method for glycosylation of proteins or modification of oligosaccharide chains on glycoproteins which is efficient, reproducible and cost-effective.
i; Y-
I
I
WO 87/05330 PCT/US86/00495 -8- SU~MARY OF THE INVENTION A method for modifying eukaryotic and prokaryotic proteins to extend their in vivo circulatory lifetimes or to control their site of cellular uptake in the body. In preferred embodiments, enzymatic and/or chemical treatments are used to produce a modified protein carrying one or more covalently attached trisaccharide SAa2-->6(3)GalBl-->4(3)GlcNAc--> or tetrasaccharide SAa2-->6(3)GalB1-->4(3)GlcNAc1 >4GlcNAc--> moieties. In alternative embodiments, one or two GlcNAc residues bound to the protein are used as a basis for construction of other oligosaccharides by elongation with the appropriate glycosyltransferases. The method can be applied to any natural or recombinant protein possessing Asn-linked oligosaccharides or to any non-glycosylated protein that can be chemically or enzymatically derivatized with the appropriate carbohydrate residues.
Generation of glycoproteins containing Asn-linked SA-->Gal-->GlcNAc--> The preferred oligosaccharide modification scheme consists of the following steps wherein all but the Asn-linked GlcNAc of the N-.linked oligosaccharide chains are enzymatically or chemically removed from the protein and a trisaccharide constructed in its place: Step 1. Generation of GlcNAc-->Asn(protein). The initial step is cleavage of the glycosidic bond connecting tne two innermost core GlcNAc residues of some or all N-linked oligosaccharide chains of a.
glycoprotein with an appropriate endo-B-N-acetylglucosaminidase such as Endo H or Endo F. Endo H cleaves the high-mannose and hybrid oligosaccharide chains of glycoproteins produced in eukaryotic cells as well as the mannans produced in yeast such as Saccharomyces cerevisiae, removing all but a single GlcNAc residue attached to each glycosylated Asn residue of the polypeptide backbone. Endo F can Scleave both high-mannose and biantennary complex chains of N-linked oligosaccharides, again leaving a single GlcNAc residue attached at each glycosylation site., If a given glycoprotein contains complex oligosaccharides such as tri- or tetraantennary chains which are Sinefficiently cleaved by known endoglycosidases, these chains can be
S.I
I:
WO 87/05330 PCTUS86/00495 -9trimmed with exoglycosidases such as sialidase, 8- and a-galactosidase, a-fucosidase and 0-hexosaminidase. The innermost GlcNAc residue of the resulting core can be then be exposed by any of several procedures. One procedure is digestion with Endo F or other endo-B-Nacetylglucosaminidases such as Endo D. A second procedure is digestion with a-mannosidase followed by digestion with either Endo L or with B-mannosidase and B-hexosaminidase.
Alternatively, glycoproteins normally bearing complex Asn-linked oligosaccharides can be produced in mammalian cell culture in the presence of a processing inhibitor such as swainsonine or deoxymannojirimycin. The resulting glycoprotein will bear hybrid or highmannose chains susceptible to cleavage by Endo H, thereby eliminating the need for an initial treatment of the glycoprotein with exoglycosidases. In a related variation, the glycoprotein may be produced in a mutant cell line that is incapable of synthesizing complex N-linked chains resistant to endoglycosidases such as Endo H or Endo F.
All sugars other than the N-linked GicNAc residues may also be removed chemically rather than enzymatically by treatment with trifluoromethanesulfonic acid or hydrofluoric acid. In general, chemical cleavage can be expected to be less useful than enzymatic methods because of the denaturing effects of the relatively harsh conditions used.
Step 2. Attachment of Gal to GlcNAc-->Asn(protein). The second step is the enzymatic addition of a Gal residue to the residual GlcNAc on the protein by the action of a galactosyltransferase. The preferred galactosyltransferase is a bovine milk enzyme which transfers Gal to GlcNAc in the presence of the sugar donor UDP-Gal to form a 81-->4 linkage. In another variation, galactose can be added to the GlcNAc residue with a 81-->3 linkage by the use of a galactosyltransferase from a source such as pig trachea.
Step 3. Attachment of SA to Gal-->GlcNAc-->Asn(protein). The final step is the enzymatic addition of a sialic acid residue to Gal 1-->4(3)G1cNAc-->Asn(protein). This reaction can be carried out with an a2-->6-sialy'Ttransferase isolated, for example, from bovine colos''rum or rat liver, which transfers SA from CMP-SA to form an a2->6 linkage to the terinal galactose residue of Ga181-->4(3)- Gl1NAc-->Asn(protein). Alternatively, an a2-->3-sialyltransferase may a
Q-
WO 87/05330 PCT/US86/00495 be used to form an a2-->3 linkage to each terminal Gal residue.
Although the preferred sialic acid is N-acetylneuraminic acid (NeuAc), any naturally occurring or chemically synthesized sialic acid which the sialyltransferase can transfer from the CPM-SA derivative to galactose may be used, for exapmole, N-glycolyl neuraminic acid, acetyl-N-acetyl neuraminic acid, and 4-0-acetyl-N-acetyl neuraminic acid.
Generation of glycoproteins containing Asn-linked GlcNAc-->G1cNAc--> In a second embodiment, the oligosaccharide chains of the glycoprotein, whether natural or produced in the presence of a processing inhibitor or in a mutant cell line, are trimmed back to the two, rather than one, innermost cere GlcNAc residues by the use of appropriate exoglycosidases. For example, a and B-mannosidase would be used to trim a high-mannose oligosaccharide. The product of this treatment, GlcNAcB1-->4GlcNAc-->Asn(protein), is then converted to the tetrasaccharide SAa2-->6(3)GalBl-->4(3)GlcNAc81-->4IGcNAc-->Asn- (protein) by sequential treatment with galactosyl- and sialyltransferases.
Attachment of oligosaccharides to non-glycosylated amino acid residues of proteins.
In a third embodiment, an oligosaccharide such as the trisaccharide SA-->Gal-->GlcNAc--> or disaccharide is attached at non-glycosylated amino acid residues of a protein expressed either in a eukarykotic system or in a bacterial system. For example, to attach the trisaccharide SA-->Gal-->GlcNAc, the protein is treated with a chemically reactive glycoside derivative of GlcNAc-->, Gal- >GicNAc-->, or In the first two cases, the mono- or disaccharide is then extended to the trisaccharide by the appropriate glycosyltransferase(s). The initial carbohydrate moieties can be attached to the protein by a chemical reaction between a suitable amino acid and a glycoside derivative of the carbohydrate containing an appropriately activated chemical group. Depending on the activation group present in the glycoside, the carbohydrate will be attached to amino acids with free amino groups, carboxyl groups, 0 i 'i i I w
I
0 87/05330 PCT/US86/00495 -11sulfhydryl groups, or hydroxyl groups or to aromatic amino acids.
Generation of other oligosaccharides by elongation of protein-linked GlcNAc residues.
Variations of the disclosed procedures can be used to produce glycoproteins with oligosaccharides other than the tri- or tetrasaccharides described above. For example, extended oligosaccharide chains consisting of SAa2-->6(3)GalBl-->4(GcNAcB1-->3GalB-->4) GlcNAc--> n or SAa2-->6(3)GalBl-->4(G1cNAcB1-->3GalBl-->4)nGlcNAcBl-->4GlcNAc-->, where n is 1-10, can be constructed by subjecting a glycoprotein carrying one or two core GlcNAc residues to alternate rounds of B1-->4 galactosyltransferase and B1-->3 N-acetylglucosaminyltransferase treatments. The resulting extended oligosaccharide chain can be useful for increasing solubility or masking protease-sensitive or antigenic sites of the ooiyoeptide.
Many other useful oligosaccharide structures can be constructed by elongation of protein-linked monosaccharides or disaccharides with the use of appropriate glycosyltransferases. An example is the branched fucosylated trisaccharide GasB1-->4(Fucal-->3)G1cNAc-->.
These and other structures could be useful in preferentially "targeting" a glycoprotein to a specific tissue known to contain receptors for a specific mono- or oligosaccharide.
r WO 87/05330 PCT/US86/00495 -12- BRIEF DESCRIPTION OF THE DRAWINGS Fig. I shows the structures of the lipid-linked precursor oligosaccharide, Glc 3 MangGlcNAc 2 a high-mannose Asn-linked oligosaccharide, Man 9 GlcNAc 2 and a typical triantennary complex Asn-linked oligosaccharide. The anomeric configurations and linkage positions of the sugar residues are indicated, and dotted lines enclose the invariant pentasaccharide core shared by all known eukaryotic Asn-linked oligosaccharides.
Fig. 2 is a simplified biosynthetic pathway for Asn-linked oligosaccharide biosynthesis in yeast and higher organisms. For clarity, anomeric configurations and linkage positions are not shown, but the arrangement of the branches is tne same as in Fig. 1.
Fig. 3 is a Coomassie blue-stained gel prepared by sodium dodecylsulfate-polyacrylamide gel electrophoresis (SDS-PAGE) of yeast external invertase before and after treatment with glycosidases. The acrylamide concentration was untreated invertase; invertase after treatment with Endo H under non-denaturing conditions; (C) invertase after Endo H treatment under denaturing conditions (0.7% SDS); and an aliquot of i sample first treated with Endo H under non-denaturing conditions and subseauently treated with jack bean amannosidase.
Fig. 4 is a fluorogram of a 6% SDS-PAGE gel of samples of yeast external invertase removed at intervals (5 min, 1 hr, 2 hr, 3 hr, hr, 9 hr and 19 hr) during galactosylation of Endo H-treated, SOSdenatured invertase (Fig. 3B) with UDP-[ 3 H]Gal and bovine milk 81-->4 galactosyltransferase.
SFig. 5 shows the rate of incorporation of acid-precipitable radioactivity into Endo H-treated, SDS-denatured yeast external invertase during treatment with UDP-[ 3 HJGal and bovine milk B1-->4 galactosyltransferase.
Fig. 6 is an autoradiogram of a 6% SDS-PAGE gel of various yeast external invertase derivatives that have been sialylated using CMP- 14 C]NeuAc and bovine -colostrum a2-->6 sialyltransferase. Sialylation product derived from galactosylated, Endo H-treated, SDSdenatured invertase; sialylation product derived from a galactosylated sample of Endo H- and jack bean a-mannosidase-treated, non- 1 1 1 1 i 1 1 .1 V'1 lla In accordance with a further aspect of this invention there is provided a method for modifying proteins comprising: derivatizing amino acids on the protein with a glycoside or thioglycoside S-X, wherein S is a first saccharide selected from the group consisting of Nacetylglucosamine and galactose and X is an aglycone, and enzymically attaching a second saccharide selected from the group consisting of galactose, N-acetylglucosamine, fucose, and sialic acid.
The invention also extends to a method for targeting a protein to a cell having a specific surface receptor for a saccharide, said method comprising: attaching to the protein an oligosaccharide chain, said oligosaccharide chain having an exposed saccharide and a Gal-->GlcNAc sequence, wherein said exposed saccharide is recognized by the cell surface receptor and said oligosaccharide chain is attached to the protein with a Gal-->GlcNAc sequence.
1 i
I
i i r
S
4 4*0455
S
S
SE
S
S
*r S 04 a
S
S y r WO 87/05330 PCT/US86/00495 -13denatured invertase; sialylation product derived from untreated invertase.
Fig. 7 is a Coomassie blue-stained 6% SDS-PAGE gel of untreated bovine serum albumin (BSA); BSA converted to GlcNAc-BSA containing approximately 48 GlcNAc residues per molecule of protein by incubation with 2-imino-2-methoxyethyl-l-thio-N-acetylglucosaminide in 0.25 M sodium borate pH 8.5 for 24 hr at room temperature; galactosylated BSA formed by treatment of GlcNAc-BSA with UDP-C 3 H]Gal and bovine milk 81-->4 galactosyltransferase; and sialylated BSA formed by treatment of Gal-->GicNAc-BSA with CMP-C14C]NeuAc and bovine colostrum a2--> 6 sialyltransferase.
Fig. 8 is a graph of specific uptake (ng/2 x 105 cells) of Gal-->GlcNAc-[ 1 2 5 1]BSA and GlcNAc-[ 125 I]BSA by thu Man/GlcNAc receptor of thioglycollate-elicited mouse peritoneal macrophages as a function of the concentration of glycosylated BSA (ug/ml), where specific uptake is equal to total uptake (uptake in the absence of mannan) minus non-specific uptake (value obtained in the presence of mannan).
Fig. 9 is a graph of specific uptake (ng/mg cellular protein) of Gal-->GlcNAc[125I]BSA and NeuAc-->Gal-->GlcNAc-r 1 2 5 1]SA by the Gal/GalNAc receptor of HepG2 cells vs. protein concentration to 7.5 pg protein/ml), where specific uptake is equal to total uptake (uptake in the absence of asialo-orosomucoid) minus non-specific uptake (value obtained in the presence of asialo-orosomucoid).
Fig. 10 Analysis of 3 H]Gal-->GlcNAc-RNase by fast protein liquid chromatography (FPLC) on a Mono S column before and after a sialylation with CMP-NeuAc and rat liver a2-->6 sialyltransferase, where the column was eluted with a linear gradient as described below.
Ji Z- LI WO 87/05330 PCT/US86/00495 -14- DETAILED DESCRIPTION OF THE INVENTION The present invention is a method for modifying proteins wherein oligosaccharide chains are bound to the protein to enhance in vivo stability or to target the protein to cells having specific receptors for an exposed saccharide in the attached oligosaccharide chain(s).
The method has two principal embodiments. The first is to cleave the existing Asn-linked oligosaccharide chains on a glycoprotein to leave one or two GlcNAc residues attached to the protein at Asn and then enzymatically extend the terminal GlcNAc to attach Gal and SA. The second is to chemically or enzymatically attach a GlcNAc or Gal residue to the protein at any of a number of different amino acids and then enzymatically extend the terminal GlcNAc or Gal to form an oligosaccharide chain capped with sialic acid. There are a number of variations of the methods and enzymes used at each step of the methods, depending on the substrate and desired oligosaccharide structure.
A. Generation of glycoproteins containing SA-->Gal-->G1cNAc-->Asn- (protein) Step 1. Generation of GlcNAc-->Asn(protein). There are several methods for preparing glycoproteins containing a single GlcNAc residue attached to glycosylated asparagine residues. Six methods are as follows.
a. Cleavage by Endo H. To anerate GlcNAc-->Asn(protein) enzymatically on glycoproteins having one or more oligosaccharides of the high-mannose or mannan type, the glycoprotein is incubated with an endo-B-N-acetylglucosaminidase capable of cleaving these oligosaccharide structures. The enzyme hydrolyzes the bond between the two core GlcNAc residues of susceptible N-linked oligosaccharides, leaving behind a single GlcNAc residue attached to the glycosylated Asn residues. The preferred enzyme for this purpose is Endo H, which has been isolated from Streptomyces plicatus. The enzyme is available either as the naturally occurring protein or as the recombinant DNA product expressed in E. coli or Streptomyces lividans.
Endo H cleaves all susceptible oligosaccharide structures of p4 f j WO 87/05330 PCT/US86/00495 denatured glycoproteins and many of those on native glycoproteins.
However, in native glycoproteins the GlcNAc 2 cores of some highmannose glycans may be protected from cleavage by Endo H due to steric factors such as polypeptide folding. This can frequently be overcome by the use of one of several mild denaturing agents that promote partial polypeptide unfolding. Examples of such mild denaturants include detergent such as Triton X-100, NP-40, octyl glucoside, deoxycholate and dilute sodium dodecyl sulfate; disulfide bond reducing agents such as dithiothreitol and B-mercaptoethanol; chaotropic agents such as urea, guanidinium hydrochloride and sodium isothiocyanate; and low concentrations of organic solvents such as alcohols (methanol, ethanol, propanol or butanol), DMSO or acetone. Endo H is a very stable enzyme, active over a pH range of about 5 to 6.5, in low- or highionic strength buffers, and in the presence of the above-mentioned denaturing agents or protease inhibitors such as phenylmethanesulfonyl fluoride, EDTA, aprotinin, leupeptide and pepstatin. Protocols for the use of Endo H have been published by Trimble and Maley in Anal.
Biochem. 141, 515-522 (1984). The precise set of reaction conditions which will optimize the cleavage of oligosaccharides by Endo H while preserving biological activity will most likely vary depending on the glycoprotein being modified and can be determined routinely by someone of ordinary skill in this field.
In situations where one or more intact high-mannose glycans persist even after incubation under the most stringent Endo H reaction conditions judged safe to use, exposed mannose residues can be irimmed away by the use of an a-mannosidase such as the commercially available a-mannosidase from jack bean. While high-mannose oligosaccharides modified in this way will not serve as substrates for the further modification reactions described below, this treatment should reduce the possibility that mannose-specific receptors on macrophages or other cells might bind to residual high-mannose glycan(s) on the glycoprotein and cause its premature clearance from the circulation.
As mentioned earlier, yeast glycoproteins sometimes contain O-linked oligosaccharides consisting of one to four a-linked mannose residues. Because these could bind to a mannose-speci.,ic receptor and shorten the serum lifetime of a glycoprotein, it is advisable to treat Co any protein found to contain such oligosaccharides with an a-mannosilj t 'lw F 0 I I WO 87/05330 PCT/US86/00495 -16dase such as the enzyme from jack bean. This would remove all but the innermost, protein-linked mannose residue from the 0-linked chains.
Because a-mannosidase treatment could interfere with subsequent cleavage by Endo H or Endo CII, it should be performed after digestion with these enzymes.
A common 0-linked oligosaccharide in animal cells is Gai-->GalNAc- >Ser/Thr(protein). These glycans can be removed with the enzyme endo-a-N-acetylgalactosaminidase, which is commercially available from Genzyme Corp., Boston MA. Many other mammalian 0-linked oligosaccharides can be converted to Gal-->GalNAc-->Ser/Thr(protein) by treatment with exoglycosidases such as sialidase, 8-hexosaminidase and a-fucosidase. The resulting protein-linked disaccharides could then be removed from the polypeptide with endo-a-N-acetylgalactosaminidase.
b. Cleavage by other endo-8-N-acetylglucosaminidases. Several other endo-6-N-acetylglucosaminidases are also capable of cleaving between the two innermost GlcNAc residues of various N-linked oligosaccharides. The oligosaccharide specificities of these enzymes vary and are summarized in Table I. Two of these endoglycosidases, Endo CII and Endo F, can be used in place of Endo H to cleave high-mannose glycans. Unlike Endo H, however, Endo F is also active with bianternary complex N-linked oligosaccharides. Although the N-linked oligosaccharides of vertebrates are not substrates for Endo D, this enzyme would be active with glycoproteins produced by insect cells, which produce significant quantities of N-linked Man 3 GlcNAc 2 in addition to high-mannose oligosaccharides, as reported by Hsieh and Robbins in J. Biol. Chem. 259, 2375-82 (1984). in situations where the target glycoprotein contains multiple oligosaccharides sensitive to different endo-B-N-acetylglucosaminidases, the glycoprotein can be incubated with the enzymes either sequentially or in combination to maximize cleavage.
c. Cleavage by Endo H after incubation of cultured cells with oligosaccharide processing inhibitors. Mammalian cells often synthesize glycoproteins carrying oligosaccharides with structures that are resistant to all of the above-mentioned endo-8-N-acetylglucosaminidases, tri- or tetraantennary complex oligosaccharides. If such a glycoprotein is being produced in a cultured cell system, it is possible to block the later stages of oligosaccharide processing by l 1 1 i SWO 87/05330 PCT/US86/00495 -17adding oligosaccharide processing inhibitors to the culture medium.
Two preferred processing inhibitors are deoxymannojirimycin and swainsonine. Cells treated with one of these inhibitors will preferen- Stially synthesize N-linked oligosaccharides with Endo H-sensitive structures. Deoxymannojirimycin inhibits Mannosidase I, thereby blocking further modification of high-mannose N-linked oligosaccharides. Swainsonine is a Mannosidase II inhibitor, blocking the removal of the two a-linked mannose residues on the al-->6-linked mannose residue of the Man 2 GlcNAc 2 core conversion of structure M-d to structure M-e in Fig. As a result, glycosylated Asn residues which would normall2y carry Endo H-resistant complex type glycans will carry Endo H-sensitive "hybrid" oligosaccharides instead. Swainsonine and deoxymannojirimycin are both commercially available, for example from Genzyme Corp., Boston MA, or Boehringer Mannheim, Indianapolis IN In most cases, the altered glycoproteins produced in the presence of deoxymannojirimycin or swainsonine will still be secreted in biologically active form. The use and properties of swainsonine and deoxymannojirimycin, as well as those of other oligosaccnaride processing inhibitors, have been reviewed by Schwartz and Datema, Adv.
Carbohyd. Chem. Biochem. 40, 287-379 (1982) and by Fuhrmann et ail.
Biochim. Bioohys. Acta 825, 95-110 (1985).
Oligosaccharide processing inhibitors that block Glucosidases I or II, such as deoxynojirimycin or castanospermine, which are both available from Genzyme Corp., Boston MA, will also generate Endo Hsensitive structures, but these inhibitors are less preferred because they sometimes block secretion. Many other oliaosaccharide processing inhibitors, described in the two reviews cited in the previous oaragraph, will also serve the same purpose.
d. Cleavage by endo-B-N-acetylglucosami nidases after production of a glycoprotein in a mutant cell line. Another approach for manipulating the structures of the N-linked oligosaccharides of a glycoprotein is to express it in cells with one or more mutations in the oligosaccharide processing pathways. Such mutations are readily selected for in mammalian cells. A number of techniques have been used to generate processing mutants, but selection for resistance or hypersensitivity to one or more of a variety of lectins, as an indicator of the presence of a processing mutation, has been one useful
A
I
WO 87/05330 PCT/US86/00495 -18approach. DNA coding for a glycoprotein(s) can be introduced into such a mutant cell line using conventional methods transformation with an expression vector containing the DNA). Alternatively, a mutant subline with defective processing can be selected from a line already capable of producing a desired glycoprotein.
Depending on the desired phenotype, any of a wide variety of mutant cell lines can be used. For example, there are perfectly viable, fat-growing GlcNAc transferase I mutants of both CHO cells (an established Chinese hamster ovary cell line long used for mutational studies and mammalian protein expression) and BHK-21 cells (an established line of baby hamster kidney origin). Both CHO and BHK-21 cells are available from the American Type Culture Collection, Rockville MD. Because of the missing enzyme activity, the mutant cells are unable to synthesize any complex or hybrid N-linked oligosaccharides; glycosylated Asn residues which would normally carry sucn glycans carry Man 5 GlcNAc 2 instead. Thus, glycosylated Asn residues carry only Man 5 _GlcNAc 2 all structures which are sensitive to Endo H. Many other mutant cell lines have also been characterized, examples of which include lines with various defects in fucosylation, a defect in galactosylation resulting in failure to extend the outer branches past the GlcNAc residues, an inability to add extra branches to produce tri- and tetraantennary complex oligosaccharides, and various defects in Ser/Thr-linked glycan synthesis. The subject of processing-defective animal cell mutants has been reviewed by Stanley, in The Biochemistry of Glycoproteins and Proteoglycans, edited by Lennarz, Plenum Press, New York, 1980.
A series of yeast mutants with various defects in mannan synthesis has al.so been produced, as described by Ballou, in The Molecular Biology of the Yeast Saccharomyces, edited by Strathern et al., Cold Spring Harbor Laboratory, 1982. Thus, it is possible to produce a 'glycoprotein in a mutant S. cerevisiae strain which cannot elongate high-mannose oligosaccharides into large mannans.
e. Sequential exoglycosidase digestion with or without subsequent cleavage by Endo L or Endo D. An alternative, but less preferred method for generating GlcNAc-->Asn(protein) in cases where the glycoprotein contains high-mannose or mannan-type oligosaccharides is to remove monosaccharide units by exoglycosidase digestion with or WO 87/05330 PCT/US86/00495 -19without subsequent use of Endo L. The first step is digestion with an a-mannosidase to remove all a-linked mannose residues. In the case of mannans from some yeast strains, it may be desirable to include other exoglycosidases or phosphatases if other sugars or phosphate residues are present in the outer portion of the mannan structure. In the second digestion step, the last mannose residue is removed with a Bmannosidase. The product, GlcNAc 2 >Asn(protein), is then subjected to the third digestion step, which is carried out with B-hexosaminidase. This enzyme removes the terminal GlcNAc residue to generate GlcNAc-->Asn(protein); since the last GlcNAc is linked to the protein by an amide rather than a glycosidic bond, the hexosaminidase cannot remove the innermost GlcNAc residue from the asparagine.
Alternatively, a-mannosidase treatment of high- 1 ,annose or mannantype oligosaccharides can be followed by incubation wiTh Endo L, which can be purified from Streptomyces plicatus. This enzyme can cleave between the GlcNAc residues of Man8I-->4GicNAcB1-->4GlcNAc.
In the case of a glycoprotein containing complex or hybrid-type oligosaccharides, sequential (or, when the requirements of the enzymes make it possible, simultaneous) incubation with the appropriate exoglycosidases, such as sialidase, 6- and/or a-galactosidase, hexosaminidase, and a-fucosidase, will trim the oigcosaccharides back to Man3GlcNAc 2 This oligosaccharide can be cleaved by Endo D or Endo F. Alternatively, it can be treated with a-mannosidase to generate protein-linked ManS1-->4GlcNAc81-->4GlcNAc;. This can be cleaved either with Endo L or with digestions with a-mannosidase, B-mannosidase, and B-hexosaminidase.
Sialidase can be purified from a variety of sources, including col i, Clostridi.um perfringens, Vibrio cnolerae, and Arthrobacter urefaciens, and is commercially available from a number of sources such as Calbiochem-Behring, San Diego CA, or Sigma Chemical Corp., St.
Louis MO. B-Galactosidase can be purified from Asperoillus nier, C.
perfringens, jack bean, or other suitable sources and is commercially available from Sigma Chemical Corp., St. Louis MO. a-Galactosidase from E. coli or green coffee beans is available from Boehringer Mannheim, Indianapolis IN. B-Hexosaminidase can be purifed from jack bean, bovine liver or testis, or other suitable sources and is also commercially available from Sigma Chemical Corp., St. Louis MO. B- I 0 1 1 'I I 4 -I i; WO 87/05330 PCT/US86/00495 Mannosidase has been purified from the snail Achatina fulica, as described by Sugahara and Yamashima in Meth. Enzymol. 28, 769-772 (1972), and from hen oviduct, as described by Sukeno et al. in Meth.
Enzymol. 28, 777-782 (1972). a-Mannosidase from jack bean is preferred and is commercially available from Sigma Chem. Corp., St. Louis MO. Endo H, Endo D, and Endo F are commercially available from Genzyme Corp., Boston MA; from New England Nuclear, Boston MA; from Miles Scientific, Naperville IL; or from Boehringer Mannheim, Indianapolis IN. Conditions for the use of these and the other endo-B-N-acetylglucosaminidases Endo CII and Endo L are described in the publications cited in Table I.
f. Chemical removal of all sugars except N-linked GlcNAc. It is also possible to generate protein-linked GlcNJAc chemically. For example, as described by Kalyan and Bahl in J. Biol. Chem. 258, 67-74 (1983), hydrolysis with trifluoromethane sulfonic acid (TFMS) has been used to remove all sugars except the N-linked GlcNAc residues while leaving the protein backbone intact. Similar results have been obtained using hydrofluoric acid, as described by Mort and Lamport in Anal. Biochem. 82, 289-309 (1977).
Step 2. Attachment of galactose to G1cNAc-->Asn(protein).
In Step 2, the terminal GlcNAc residue generated in Step 1 serves as a site for tne attachment of galactose. Either of two galactosyltransferases may be used: UDP-Gal:GlcNAc-R B1-->4 galactosyltransferase or UDP-Gal:GlcNAc-R 81-->3 galactosyltransferase. In the first variation of tnis step, a B1-->4-linked galactose residue is added to G1cNAc-->Asn(protein). UDP-Gal:GlcNAc-R 81-->4 galactosyltransferase can be obtained from a variety of sources, the most common and costeffective one being bovine milk. Enzyme from this source is commercially available from Sigma Chem. Corp., St. Louis MO. The reaction conditions for using the bovine milk galactosyltransferase to transfer galactose from UDP-Gal to GlcNAc-->Asn(protein) are similar to those described by Trayer and Hill in J. Biol. Chem. 246, 6666-75 (1971) for natural substrates. The preferred reaction pH is 6.0 to 6.5. Most buffers can be used with the exception of phosphate, which inhibits enyzme activity, and a broad range of salt concentrations can be used.
WO 87/05330 PCT/US86/00495 -21- It is preferable to have 5-20 mM Mn 2 or Mg 2 present. Peptidase inhibitors such as phenylmethanesulfonyl fluoride, TPCK, aprotinin, leupeptin, and pepstatin and exoglycosidase inhibitors such as galactono-1,4-lactone can be added without interfering with the activity of the galactosyltransferase.
Since the removal of the carbohydrate from the protein can cause solubility problems, it is sometimes necessary to use relatively high concentrations of a non-ionic detergent such as 2-3% Triton X-100, other suitable solubilizers such as DMSO, or denaturing agents such as 2-3 M urea to keep the protein in solution. We have found that this does not interfere with the galactosylation step, the bovine milk 81-->4 galactosyltransferase apparently remaining sufficiently active under these conditions.
In the second variation of this step, a S1-->3-linked galactose residue is transferred to GlcNAc-->Asn(protein). UDP-Gal:GlcNAc-R 01-->3 galactosyltransferase has been purified from pig trachea.
Conditions for the use of this enzyme to transfer galactose from UDP-Gal to GlcNAc-R have been described by Sheares and Carlson in J.
Biol. Chem. 258, 9893-98 (1983).
Step 3. Attachment of sialic acid to Ga81-->4(3)GlcNAc-->Asn- (protein) The term "sialic acid" (SA) includes any naturally occurring or chemically synthesized sialic acid or sialic acid derivative. The preferred naturally occurring sialic acid is -acetylneuraminic acid (NeuAc). As discussed by Schauer in Adv. Carb. Chem. Biochem. 131-234 (1982), other sialic acids can also be transferred from CMP-SA to galactose, for example, N-glycolyl neuraminic acid, neuraminic acid, and 4-0-acetyl-N-acetyl neuraminic acid. Many other sialic acids such as those described in Sialic Acids: Chemistry, Metabolism and Function, edited by R. Schauer (Springer-Verlag, New York, (1982), are potential substrates. There are two variations of the method for attaching sialic acid to the substrate generated in Steps 1 and 2, Gal8i-->4(3)GlcNAc-->Asn(prolein).
In the first of the two variations, the sialic acid is attached to GalB1-->4GlcNAc-->Asn(protein) in an a2-->6 linkage. The CMP-SA:- WO 87/05330 PCT/US86/00495 -22- GalB1-->4GlcNAc-R a2-->6 sialyltransferase used in this step can be obtained from a variety of sources, the more usual ones being bovine colostrum and rat liver. The rat liver enzyme has recently become commercially available from Genzyme Corp., Boston MA.
The reaction conditions for using the bovine colostrum and rat liver a2--> 6 sialyltransferases to transfer sialic acid from CMP-SA to GalS1-->4GlcNAc-->Asn(protein) are similar to those described by Paulson et al. in J. Biol. Chem. 252, 2356-62 (1977) for natural substrates, except that it may be desirable to add additional enzyme to accelerate the rate of the reaction. The preferred pH is 6.5-7.0.
Although most buffers, with the exception of phosphate, can be employed, preferred buffers are Tris-maleate or cacodylate. The enzyme is functional in the presence of mild detergents such as NP-40 and Triton X-100; peptidase inhibitors such as phenylmethanesulfonyl fluoride, TPCK, aprotinin, leupeptin and pepstatin; and exoglycosidase inhibitors such as galactono-1,4-lactone.
In the second variation of this step, the sialic acid is attached to the GalB1-->(3)GicNAc-->Asn(protein) by an linkage. Two sialyltransferases producing this linkage have been described. The first, CMP-SA:GalB1-->4GlcNAc a2-->3 sialyltransferase, has been identified in human placenta by van den Eijnden and Schiphorst as described in J. Biol. Chem. 256, 3159-3162 (1981). This enzyme, although not yet purified, can be purified using conventional methods.
The second enzyme, CMP-SA:GalB1-->3(4)GIcNAc c2-->3 sialyltransferase, has been purified from rat liver by Weinstein et al. as described in J. Biol. Chem. 257, 13835-44 (1982). The rat liver enzyme has a somewhat relaxed specificity and is able to transfer sialic acid from CMP-sialic acid to.the C-3 position of galactose in both GalB!-->4- GlcNAc and GalS1-->3GlcNAc sequences. Conditions for the use of the a2-->3 sialyltransferases are described in the two publications just cited.
B. Method for preparing glycoproteins containing GlcNAc-->G1cNAc-->Asn(protein) The method used to generate SA-->Gal-->GlcNAc-->GlcNAc-->Asni -W is functional in the prsec of m i; deeget suh s P- n
I
WO 87/05330 PCT/US86/00495 -23- (protein) is similar to the method described above for generating modified glycoproteins containing the trisaccharide sequence SA-->Gal-->GlcNAc-->Asn(protein). In the preferred embodiment, both core GlcNAc residues of the original N-linked oligosaccharide are left attached to the protein and a tetrasaccharide sequence, GlcNAc-->GlcNAc--> is constructed enzymatically.
Step 1. Generation of G1cNAcB1-->4G1cNAc-->Asn(protein) The intact N-linked oligosaccharide chain is treated with exoglycosidases selected to remove all carbohydrate exterior to the two innermost GlcNAc residues. In tne case of high-mannose or mannan-type oligosaccharides, a- and B-mannosidase are used. In the case of complex or hybrid-type oligosaccharides, additional exoglycosidases are required, the specific enzymes used depending on the structures of the carbohydrate chains being modified. In most cases, treatments with sialidase, 8- and/or a-aalactosidase, 8-hexosaminidase, and if necessary, a-fucosidase, are carried out in addition to treatment with a- and B-mannosidase. Tne 8-hexosaminidase treatment is intended to remove GlcNAc residues only from the outer branches of the oligosaccharides, not from the core, and care should be taken that no B-hexosaminidase is present during or after B-mannosidase treatment. The reaction conditions and sources of the exoglycosidases are identical to those described above for Step 1 in the generation of SA-->Gal-->GlcNAc-->Asn(protein).
The methods used to attach galactose to GlcNAcl1-->4GlcNAc-->Asn- (protein) and sialic acid to GalB1-->4(3)GIcNAcB1-->4GlcNAc-->Asn- (protein) are the same as those described earlier for the preparation of modified glycoproteins containing N-linked SAa2-->3(6)Gal81-->4(3)- GlcNAc-->Asn(protein).
C. Method for attaching oligosaccharides to nonglycosylated amino acid residues of proteins The principal method for attaching oligosaccharides such as .I S W ik^ t WO 87/05330 PCT/US86/00495 -24- SA-->Gal-->G1cNAc--> to non-glycosylated amino acid residues is to react an activated glycoside derivative of what is to be the innermost sugar residue, in this case GlcNAc, with the protein and then to use glycosyltransferases to extend the oligosaccharide chain. Chemical and/or enzymatic coupling of glycosides to proteins can be accomplished using a variety of activated groups, for example, as described by Aplin and Wriston in CRC Crit. Rev. Biochem., pp. 259-306 (1981).
The advantages of the chemical coupling techniques are that they are relatively simple and do not need the complicated enzymatic machinery required for natural N-linked glycosylation. Depending on the coupling mode used, the sugar(s) can be attach arginine, histidine, or the amino-terminal amino acid of the polyoeptide; free carboxyl groups, such as those of glutamic acid or aspartic acid or the carboxyterminal amino acid of the polypeptice; free sulfhydryl groups, such as those of cysteine; free hydroxyl groups, such as those of serine, threonine, or hydroxyproline; aromatic residues such as those of phenyla'ianine, tyrosine, or tryptophan; or the amiae group of glutamine.
As shown below, the aglycone, R, is the chemical moiety that combines with the sugar to form a glycoside and which is reacted with the amino acid to bind tne sugar to the protein.
0 ,R 0 R1 -S(CH2, OH H, where R is: 0 S Ni'rCC3 0 0 R2 -0(CH 2 R3 -S(CH 2 )nNH 2 R4 -0(CH 2 )nNH2
SR.
5
-S(CH
2 )nNH-CH=CH 2 R -(CH2NH-CH=CH R -S(CH2)n-- N2+C1 and R 8
-(CH
2 N2+C1" GicNAc residues can be attached to the e-amino groups of lysine residues of a nonglycosylated protein by-treating the protein with 2imino-2-methyoxyethyl-l-thio-B-N-acetylglucosaminide as described by ~:l~il lll~i Uli -i i WO 87/05330 PCT/US86/00495 Stowell and Lee in Meth. Enzymol. 83, 278-288 (1982). Other coupling procedures can be used as well, such as treatment of the protein with a glycoside or thioglycoside derivative of GlcNAc in which the aglycone contains an activated carboxylic acid, for example R 1 or R 2 GlcNAc residues can be attached to the carboxyl groups of aspartic acid and glutamic acid residues of a nonglycosylated protein by treatment of the protein with a glycoside or thioglycoside derivative of GlcNAc in which the aglycone contains a free amino group, for example R 3 or R 4 in the presence of a coupling reagent such as a carbodiimide.
Compounds which contain free amino groups, for example GlcNAc derivatives containing the aglycones R 3 or R 4 can also be used to derivatize the amide groups of glutamine through the action of transglutaminase as described by Yan and Wold in Biochemistry 23, 3759-3765 (1984).
Attachment of GlcNAc residues to the thiol groups of the cysteine residues of a nonglycosylated protein can be accomplished by treating the protein with a GlcNAc glycoside or thioglycoside in which the aglycone contains an electrophilic site such as an acrylate unit, for example the aglycones R 5 or Rg.
The glycosylation of aromatic amino acid residues of a protein with a monosaccharide such as GlcNAc can be accomplished by treatment with a glycoside or thioglycoside in which the aglycone contains a diazo group, for example aglycones R 7 or R 8 A large number of other coupling methods and aglycone structures can be employed to derivatize a protein with a GlcNAc derivative.
After chemical derivatization of the protein with GlcNAc residues, the trisaccharide sequence SAa2-->3(6)GalB1-->4(3)GlcNAc--> is constructed by seauential enzymatic attachment of galactose and sialic acid residues, as described for Asn-linked GlcNAc residues.
In other variations, the protein is derivatized with: Gal81-->4(3)GcNAc-X, GalBl-->4(3)GlcNAc81-->4G1cNAc-X, SAa2-->3(6)Ga1l-->4(3)GlcNAc-X, or SAa2-->3(6)GalI1-->4(3)GlcNAcB1-->4GlcNAc-X, where X is an aglycone containing a free amino group, an activated ester of a carboxylic acid, a diazo group, or other groups described 1 J017;i ,7 ii-~ i- WO 87/05330 PCT/US86/00495 -26above.
The same procedures may be used to chemically attach galactose, rather than GlcNAc, directly to an amino acid. The galactose may then be enzymatically extended or capped with sialic acid, as previously described.
D. Generation of additional protein-linked oligosaccharides by elongation of GlcNAc-protein or GlcNAc-->GlcNAc-protein.
Procedures similar to those used to extend GlcNAc-protein or GlcNAc-->GlcNAc-protein to a protein-linked oligosaccharide resembling tne outer branch of a complex oligosaccharide can be employed to construct other carbohydrate structures found on GlcNAc residues attached to the terminal mannose units of the core pentasaccharide.
Example 1. Generation of proteins containing repeating units of (GicNAcS1-->3GalS1-->4). After preparation of either GlcNAc-protein or GicNAcS1-->4GcNAc-protein using the methods described above, a long carbohydrate chain may be generated by several rounds of alternating UDP-Gal:GlcNAc-R 81-->4 galactosyitransferase and UDP-GicNAc:GalS21-->4GlcNAc-R 81-->3 N-acetylglucosaminyltransferase incubations. This will generate a poiylactosaminyl-tyoe structure of the type (GlcNAcSi-->3GalB1-->4) attached to the GlcNAc-protein or G1cNAcSl-->4GicNAc-orotein starting material. Kaur, Turco and Laine reported in Biochemistry International 345-351 (1982) that bovine milk UDP-Gal :GicNAc 81-->4 galactosyltransferase can transfer the 81-->4-linked galactosyl residues to polylactosaminyl cligosaccharides, and a 61-->3 N-acetylglucosaminyvitransferase has been identified in Novikoff ascites tumor cells by van den Eijnoen et al., J.
Biol. Chem. 258, 3435-37 (1983). The number of repeating GieAc-->Gal units in the structure can be varied depending on the desired length, 1-10 such units should suffice for most applications. The essential element is that, after attachment of the disaccharide units, an exposed galactose residue is present so that the carbohydrate chain can be capped with or 2-->6-linked sialic acid, as described above.- Thus, the final structure would be SAa2-->6(3)Gal81-->4[GlcNAc1--3GalB1-->4J l cNAc-protein, or SAM2-->6(3)GalS1-->4[Gl1cNAcS1-->3Gal1-->4j GlcNAc1-->4GlcNAcn
C.
I I
U
4..
AK...
I".
36/00495 y ir :0 ;o ?s nf n a'.
,.IF 049 ill NIIIIIIIIIEFF- -Oqw
I
WO 87/05330 PCT/US86/00495 -27protein, where n is 1-10.
The advantages of introducing such a polylactosaminyl structure would be to, increase solubility or to better mask the protein backbone to protect it from recognition by the immune system or from degradation by proteases.
Example 2. Generation of glycoproteins containing terminal GalSI-->4(3)[Fuca1-->3(4)GlcNAc or structures. After preparation of GalSI-->4(3)GlcNAc-protein, Galel-->4(3)GlcNAc~l-->4GlcNAc-protein, Galol->'.(4)[GcN~c.1-->Gal~ -4In cNSI>4cN-poen SAaL2-->3Gal~1-->3GulcNJAc-protein, SAa2-->3Galoi-->3EGlcNAc~1-->3Gal81-->41 nGlcNAc-protein, SAaL2-->3Gale1-->3GlcNAc81-->4GlcNAc-protein or SMc2-->3Gal~l-->3EGlcWAcB1-->3GalI-->4.1 rGlcNAc~l-->4Gulcl'4Ac-protLein where n is between 1 and 10, using the methods described above, a fucose can be attached to any of the acceptor GlcNAc residues by treatment with O-u and a GDP-Fu.-:GicNAc fucosyltransferase. The purification of this f ucosyl transf erase, its substrate Specificity and preferred reaction conditions have been reported by Prieels et al in J. Biol. Chem. 256, 104456-634 (1981). The activity of this enzyme with sialylated substrates has been described by Johnson and Watkins in Proc. VIIIth Int. Sym~p. Glycoconjugates (1985), elds.
E.A. Davidson, UI.C. Williams and Di Ferrante. if it is desired to attach fucose only in an al-->3 linkage to the appropriate acceptor GlcNAc residues, the GDP-Fuc:GlcNAc a1-->3 f ucosyl transf erase can be used. This enzyme has been described by Johnson and Watkins in Proc.
VIIth Int. Symp. Glycoconjugates (1985), eds. E.A. Davidson, J.C.
Williams and N.M. Di Ferrante.
E. Targeting of glycosylated proteins to specific cells Cells with sugar-specific cell surface receptors are able to recognize and internalize glycoproteins bearing appropriate carbohydrate structures. The best characterized sugar-specific cell WO 8 7 j surfac recept on hep sugar- As hwe 1 by Le p rec ep sugar to ex i nvol as de Anoth as de 1403 ol i go repor glyco cant attac
G
where whe r whe r cyte enz
I
k -1 40 oligosaccharide chains are cleaved by digesting the tiI-h ~n n nvmpA selected from the group i i-I WO 87/05330 PCT/US86/00495 -28surface receptors are the Gal receptor of hepatocytes, the Man/GlcNAc receptor of reticulo-endothelial cells and the fucose receptor found on hepatocytes, lymphocytes and teratocarcinoma cells. The subject of sugar-specific cell surface receptors has been reviewed by Neufeld and Ashwell in The Biochemistry of Glycoproteins and Proteoglycans, edited by Lennarz, Plenum Press, New York (1980), pp. 241-266.
Proteins can be targeted to cells with sugar-specific cell surface receptors by generating glycoproteins that contain the appropriate sugar at nonreducing terminal positions. Several procedures are used to expose the desired terminal sugars. One orocedure, in general, involves the treatment of a native glycoprotein with exoglycosidases, as described by Ashwell and Morell in Adv. Enzvmol. 41, 99-128 (1974).
Another procedure is the attachment of monosaccharides to the protein, as described by Stahl et al. in Proc. Nati. Acad. Sci. USA 75, 1399- 1403 (1978). A third approach is the attachment of derivatives of oligosaccharides isolated from natural sources such as ovalbumin, as reported by Yan and Wold in Biochemistry 23, 3759-3765 (1984). The glycosylated proteins tnat are the subject of the present invention can be targeted to specific cells, depending on the specific sugars attached.
Gal-->GlcNAc-protein, Gal-->GlcNAc-->GicNAc-protein, (Gal-->GlcNAc) >Gal-->GcNAc-protein and (Gal-->GlcNAc) Gal-->GcNAc-->GcNAc-protein, where n is 1-10, are directed to hepatocytes.
GlcNAc-protein, GlcNAc-->GlcNAc-protein, (GlcNAc-->Gal) >GlcNAc-protein and (GlcNAc-->Gal),-->GlcNAc-->G1lcNAc-protein, Swhere n is 1-10, are targeted to macrophages. Finally, Gal-->(Fuc-->)GlcNAc-protein, Gal-->(Fuc-->)GlcNAc-->GlcNA-protein, GlcNAc -protein, and Gal-->(Fuc-->)GlcNAc-->[Gal-->(Fuc-->)mGlcNAc]n-->GlcNAc-protein, m n where n is 1-10 and m is 0 or 1, are targeted to hepatocytes, lymphocytes and teratocarcinoma cells. One application of targeting is for enzyme replacement therapy. For example, glucocerebrosidase can be II ~C' -r: WO 87/05330
'I
PCT/US86/00495 -29targeted to macrophages for the treatment of Gaucher's disease. A second application is to target drugs or toxint to teratocarcinoma cellIs.
WO 87/05330 PCT/US86/00495 The following non-limiting example demonstrates the method of the present invention on a yeast glycoprotein possessing multiple highmannose and mannan oligosaccharides.
Step 1. Endo H treatment of yeast external invertase.
Yeast external invertase is a glycoprotein containing approximately two high mannose and seven mannan oligosaccharides. External invertase of a commercial preparation from Saccharomyces cerevisiae, obtained from Sigma Chem. Corp, St. Louis MO, was purified as described by Trimble and Maley in J. Biol. Chem. 252, 4409-12 (1977), and treated with Endo H essentially as described by Trimble et al. in J.
Biol. Chem. 258, 2562-67 (1983). The purified invertase was denatured by placing a 1% SDS solution of the glycoprotein in a boiling water bath for 5 minutes. The denatured invertase (250 pg) was then incubated with Endo H (0.3 pg, from Miles Scientific, Naperville IL) for hours at 37*C in 175 1l of 0.1 M sodium citrate buffer, pH After Endo H treatment, the reaction mixture was desalted on a Bio-Gel P-4 column (1 x 10 cm) eauilibrated and eluted with 50 mM ammonium acetate, pH 6.5. The method of desalting is not critical. Dialysis or protein precipitation can also be used. The material eluting in the void volume of the column was pooled and lyophilized.
Analysis of the Enao H-treated preparation of SDS-aenatured invertase by SOS-PAGE, shown in Fig. 3c, indicated that the glycoprotein had been converted to a form consistent with an invertase possessing only a single GlcNAc residue at each glycosylation site.
In a parallel experiment, native invertase was treated with Endo H in the same manner as the SDS-denatured invertase. Analysis of the desalted reaction product by SOS-PAGE, shown in Fig. 3b, indicated .C that 2-3 oligosaccharide chains of native invertase were resistant to cleavage by Endo H. To remove exposed mannose residues on the resistant chains, 250 ug Endo H-treated invertase was desalted, lyophilized, and incubated in 100 pl of 50 mM sodium acetate, pH containing 50 mM NaCI, 4 mM ZnCl 2 and 20 mU of jack bean a-mannosidase (a gift from Dr. R. Trimble at State University of New York, Albany NY) for 17 hours at 37 C. Analysis of the reaction mixture by SDS-PAGE, shown in Fig. 3d, demonstrated through a shift to lower molecular weight that the a-mannosidase treatment removed additional i M WO 87/05330 PCT/US86/00495 -31mannose residues.
Step 2. Galactosylation of the Endo H-treated samples of native and denatured yeast external invertase.
An Endo H-treated sample of denatured yeast external invertase Sg, containing approximately 15 nmol of GlcNAc-->Asn(protein) sites] was incubated at 37'C in 180 il of 50 mM 2-(N-morpholino)ethanesulfonic acid (MES), pH 6.3, containing 0.8% Triton X-100, 25 mM MnCl 2 1.25 mM UDP- 3 H]Gal (specific activity, 8 Ci/mol) and bovine milk UDP-Gal:GlcNAc B1-->4 galactosyltransferase (100 mU, Sigma Chem.
Corp., St. Louis MO). Aliquots were removed at selected times and analyzed by SDS-PAGE, as shown in Fig. 4. A gradual increase in apparent molecular weight was apparent up to a reaction time of one hour.
This result was confirmed by measuring the incorporation of tritium into material precipitable by 0.5 M HC1/1% phosphotungstic acid, which gave the result shown in Fig. Nonradiolabeled galactosylated samples of native and denatured yeast external invertase were prepared as substrates for the sialylation reaction. Endo H-treated denatured invertase and Endo H plus a-mannosidase-treated native invertase were galactosylated with nonradioactive UDP..Gal using the procedures described above.
Step 3. Sialylation of the galactosylated samples of native and denatured yeast external invertase.
The native and denatured samples of nonradioactive galactosylated yeast external invertase (50 ug of orotein) were incubated at 37"C for 17 hours in 70 il of 0.1 M Tris-maleate, pH 6.7, containing 0.7 Triton X-100, 2 mM CMP-[ 14 C]NeuAc (specific activity, 1.1 Ci/mmol) and bovine colostrum CMP-SA:Gal1-->4GlcNAc-R a2-->6 sialyltransferase [1.1 mU, purified according to Paulson et al. in J. Biol. Chem. 252, 2356-2362 (1977)]. The reaction mixtures were analyzed by SDS-PAGE and autoradiography, as shown in Fig. 6. The radioactivity associated with the invertase band demonstrates that sialic acid has been attached to the galactose residues of the invertase by the sialyltransferase.
The following non-limiting example demonstrates the method of the
O
i WO 87/05330 PCT/US86/00495 -32present invention using chemical and enzymatic techniques on a protein that is not glycosylated in its native form.
Step 1. Chemical attachment of a thioglycoside derivative of GlcNAc to bovine serum albumin (BSA).
BSA was derivatized by treatment with 2-imino-2-methoxyethyl-1thio-N-acetylglucosaminide by Dr. R. Schnaar at Johns Hopkins University according to the procedure described by Lee et al. in Biochemistry 15, 3956-63 (1976). The glycosylated BSA contained, on the average, 48 lysine-linked GlcNAc residues per molecule.
Step 2. Galactosylation of GlcNAc 48
-BSA.
GlcNAc 48 -BSA (0.9 mg) was incubated at 37'C for 17 hours in 60Q. l of 0.12 M MFS, pH 6.3, containing 0.6% Triton X-100, 20 mM MnCl 2 5 mM
UDP-[
3 H]Gal (specific activity, 1 Ci/mol), 1 mM galactono-1,4-lactone, 1 mM phenylmethanesulfonyl fluoride, TPCK (21 ug), aprotinin (12 uTIU), leupeptin (0.6 vg), Depstatin (0.6 ug) and bovine milk UDP- Gal:GlcNAc-R 81-->4 galactosyltransferase. The glycosylated BSA was partially purified from other reaction components by Bio-Gel P-4 gel filtration. After measurement of the amount of radioactivity incorporated into the BSA, it was calculated that 46% of the available GlcNAc residues were galactosylated. A second incubation of tne galactosylated BSA under identical conditions increased the extent of reaction from 46 to 51%. The galactosylated BSA was ourified with an anti-BSA antibody column obtained from Cooper Biomedical, Malvern PA.
Step 3. Sialylation of galactosylated BSA.
The galactosylated SSA (240 ug) was incubated for 16 hours at 37'C in 120 pl of 0.1 M Tris-maleate, pH 6.7, containing 3 mM CMP- F 14 C]NeuAc (specific activity 0.55 Ci/mol) and bovine colostrum CMP-SA:GalBl-->4GcNAc-R a2-->6 sialyltransferase (2.1 mU). The glycosylated BSA was partially purified from other reaction components by gel filtration. After measurement of the ratio of 14C to radioactivity incorporated into the samples, it was calculated that 42% of the Gal-->GlcNAc-->protein residues were sialylated. A second incubation of the sialylated BSA with 25 mU of sialyltransferase increased the extent of sialylation to 51%. The glycoprotein was isolated by immunoaffinity chromatography on an anti-BSA antibody column.
ax WO 87/05330 PCT/US86/00495 -33- Analysis of the three glycosylated forms of BSA by SDS-PAGE demonstrated a significant increase in apparent molecular weight after each step of the procedure, as shown in Fig. 7. This evidence confirms that SA-->Gal-->GlcNAc--> moieties have been constructed on the protein.
The following nonlimiting example demonstrates the differential uptake of GlcNAc-BSA and GalB1-->4GlcNAc-BSA by GlcNlAc/Man-specific reccptors of macrophages.
iouse peritoneal macrophages, which possess cell surface receptors that recognize terminal GlcNAc and Man residues, were obtained from mice 4-5 days after intraperitoneal injection of thioglycollate broth ml per mouse). The peritoneal cells were washed with Dulbecco's modified minimal essential medium (DME) containing 10% fetal calf serum (FCS) and plated in 96-well tissue culture trays at a density of 2 x 105 cells per well. After 4 hours the wells were washed twice with phosphate-buffered saline (PBS) to remove nonadherent cells. The adherent cells remaining in the wells were used for uptake experiments with GlcNAc-[ 1 2 BSA and GalB8-->4GicNAc-7 12 I]BSA which had been radiolabeled with 12 by the chloramine T method. The radiolabeled protein preparations were added at a concentration of C.1-1.2 Pg/ml to 100 il of DME containing 10% FCS and 10 mM HEPES [4-(2-hydroxyethyl)- 1-piperazine ethanesulfonic acid], pH 7.4. Parallel exoeriments were run in the presence of yeast mannan (1 mg/mi) to measure nonspecific uptake of the glycosylated BSA samples. The cells were incubated with the samples for 30 min at 37'C and then washed five times with PBS to remove residual protein not taken up by the cells. The washed cells were dissolved in 200 ul of 1% SDS and the radioactivity determined.
Nonspecific uptake (CPM in the presence of yeast mannan) was subtracted from the total uptake (CPM in the absence of yeast mannan) to determine Man/GlcNAc receptor-specific uptake by the mouse peritoneal macrophages.
The specific uptake of GlcNAc-[125 ]BSA and GalB1-->4GlcNAc- 12 5 I]BSA is presented as a function of ESA concentration in Fia. 8.
The results demonstrate that GlcNAc-BSA, but not GalB1-->4G1cNAc-BSA, is recognized and endocytosed by mouse peritoneal macrophages.
i WO 87/05330 PCT/US86/00495 -34- The following non-limiting example demonstrates the differential uptake of GalBl-->4GIcNAc-BSA and SAa2-->6GalB1-->4GlcNAc-BSA by galactose-specific receptors of hepatoma cell line HepG2.
Samples of GlcNAc-BSA and Gal-->GlcNAc-BSA were radiolabeled with 1251 by the chloramine T method. HepG2 cells were cultured in DME containing 10% fetal calf serum. Uptake experiments were performed on cells plated in 35 mm tissue culture dishes at approximately confluency. The cells were washed with protein-free medium and incubated with 1 ml of DME containing 20 mM HEPES, pH 7.3, containing 125 cytochrome c (0.2 mg/ml) and 0.5-7.5 pg of GalB1-->4GlcNAc-[ I]BSA or SAa2-->6GalBl-->4GlcNAc-[1 25 I]BSA. Parallel experiments were performed in the presence of nonradioactive asialo-orosomucoid (0.2 mg/ml) to determine nonspecific uptake. The cells were incubated with the radiolabeled protein solutions for 2.5 hours at 37"C in a 5% CO 2 atmosphere, and then rinsed five times with chilled PBS containing 1.7 mM Ca. The washed cells were solubilized with 1 ml of 1 M SDS. Separate aliquots were used to measure radioactivity and the amount of protein per culture dish. It is assumed that the amount of protein in each dish is proportional to the number of cells. Nonspecific uptake (CPM in tne presence of asialo-orosomucoid) was subtracted from tne total uptake (CPM in the absence of asialoorosomucoid) to determine the galactose receptor-specific uptake by the HepG2 cells.
The galactose receptor-specific uptake is shown as a function of glycosylated BSA concentration in Fic. 9. The results demonstrate that Gal1-->4GlcNAc-BSA, but not SAa2-->6Gal81-->4GcNAc-BSA, is recognized and endocytosed by HepG2 cells.
The following non-limiting example demonstrates the method of the present invention on a mammalian glycoprotein having one oligosaccharide chain of the high-mannose type.
Step 1 Deglycosylation of ribonuclease B, a glycoprotein having a single high-mannose oligosaccharide.
Native ribonuclease B (490 ug), obtained from Sigma Chem. Corp., St. Louis MO, and further purified by concanavalin A affinity chromatography as described by Baynes and Wold in J. Biol. Chem. 251, 6016- 24 (1976) was incubated with Endo H (50 mU, obtained from Genzyme i'i77 S 1 1 *f "-yt1T. i WO 87/05330 PCT/US86/00495 Corp., Boston MA) in 100 ul of 50 mM sodium acetate, pH 5.5, for 24 hours at 37°C. SDS-PAGE indicated complete conversion of the glycoprotein to a form containing a single GlcNAc residue. The modified ribonuclease B was desalted on a Bio-Gel P6DG column and the ribonuclease fractions were freeze-dried.
Step 2. Galactosylation of Endo H-treated ribonuclease B.
Endo H-treated ribonuclease B (400 Vg) was incubated for 3 hours at 37° in 250 Pl of 0.1 M MES, pH 6.3, containing 0.1% Triton X-100, 0.01-M MnCl 2 100 mU bovine milk UDP-Gal:GlcNAc-R 81-->4 galactosyltransferase and 300 nmol UDP-F 3 H]Gal (specific activity 17.3 Ci/mmol).
The galactosylated ribonuclease was analyzed by FPLC on a Mono S column. A linear gradient from 20 mM sodium phosphate, pH 7.95 to mM sodium phosphate containing 1 M NaCl was run. The galactosylated ribonuclease eluted at a NaCl concentration of 0.13 M. The protein peak measured by UV absorbance (A 280 coincided with a peak of radioactivity, as shown in Fig. 10 The protein peak eluting at 0.13 M NaCl was collected and analyzed by SDS-PAGE. The only protein band detected after staining with Coomassie blue co-migrated with Endo H-treated ribonuclease B (not shown).
Step 3. Sialylation of galactosylated ribonuciease.
A 40 al aliquot of the reaction mixture from Step 2 was mixed with pl of 6.5 mM CMP-NeuAc and 10 ul of rat liver CMP-NeuAc:Gai-R a2-->6 sialyltransferase (1.6 mU, obtained from Genzyme Corp., Boston MA) and incubated at 37°C for 18 hours. The sialyiated ribonuclease was analyzed by FPLC on a Mono S column using the conditions described in Step 2. The sialylated ribonuclease eluted at a NaCl concentration of 0.18 M, as judged by the profiles of both A 280 and radioactivity.
I -The profile of radioactivity is shown in Fig 10, The conversion of Gal-->GlcNAc-RNAse to SA-->Gal-->RNAse appeared to be quantitative.
OF-
N 1t r WO 87/05330 PCT/US86/00495 -36- Although this invention has been described with reference to specific embodiments, it is understood that modifications and variations of the methods for modifying or glycosylating proteins, and the glycosylated proteins, may occur to those skilled in the art. It is intended that all such modifications and variations be included within the scope of the appended claims.
T77 i' WO 87/05330 PCT/US86/00495 -37- Table I. Oligosaccharide Specificities of Endo-O-N-acetyl gl ucosarninidases Enzyme and Source Susceptible N-i inked ol igosacchari des (Ref.) Endo H (Srpoye Iicatui Endo
C
11 (C'lostridium per-fri ngens) Endo D (Dipiococcus oneunioni ae) Endo L (S..Dlicatus) Endo F (Fl avobacteri urn meni ngoseoti curn) Yeast mannans, all high-mannose oligosaccharides, and hybrid oligosaccharides.
(The enzyme requires an al-->3-linked mannose residue att'ached to the al-->6 mannose residue of the Man 3 GlcNAc 2 core and we have found that the enzyme is not inhibited by an al-->6 linked fucose attached to the innermost GicNAc residue.) Certain hich-mannose ol iaosaccharides.
(Similar to Endo H except that it will not cleave substrates ifthe mannose linked to the B-linked rnannose is substitute at C-4 with another sugar or if the E-linked mannose residue is substituted with a 81-->4linked GIcNAc residue.) Man~ GlcNAc ,with or without* a fucose res~a~e link d to the i nnermost GlcNAc residue ManGi cNAc 2 High-mannose and biantennary complex ol igosaccliarides (1,2) (3) (4) (6) References: 1. Tarentino et al., Meth. Enzymol. 50, 574-880 (1978).
2. Tai and KO DtfiT BiTiim. Biooinys-7es. Commun. 78, 434-441 (i19717).
3. Kobata, Meth. Enz--7 5,56-7 ,7) 4. MuramatsW,-et-TnymoIT 50, 555-559 (1.978).
Trimble et -a17 uJ71"oT. Oe~m. .254, 9708-13 (1979).
1I

Claims (26)

1. A method for modifying a glycoprotein comprising: attaching a galactose residue to a core N-acetylglucosamine to form a Gal-GlcNAc sequence; and attaching a sialic acid residue to the galactose to form a SA- Gal-GlcNAc sequence.
2. The method of claim 1 further comprising first cleaving asparagine-linked oligosaccharide chains of the glycoprotein to remove all sugars other than core N-acetylglucosamine residues bound to the glycoprotein. The method of claim 2 wherein the oligosaccharide chains are cleaved by an endoglycosidase.
4. The method of claim 3 wherein the endoglycosidase is selected from the group consisting of endo--N-acetylglucosaminidase H, endo-B-. N-acetylglucosaminidase F, endo--N-acetyl glucosaminidase CIT, endo-0- N-acetylglucosaminidase D, endo-B-N-acetylglucosaminidase L, and combinations thereof. The method of claim oligosaccharide chains with consisting of a-mannosidase, combinations thereof.
6. The method of claim oligosaccharide chains with consisting of a-mannosidase, 4 further comprising cleaving 0-linked an enzyme selected from the group endo-a--acetylgalactosaminidase, and 1 further comprising cleaving 0-linked an enzyme selected from the group endo-o-N-acetylgalactosaminidase, and ~0 WO 87/05330 3 9 PCT/US86/00495 combinations thereof.
7. The method of claim 2 wherein the oligosaccharide chain is cleaved by digestion with an exoglycosidase.
8. The method of claim 7 wherein the exoglycosidase is selected from the group consisting of sialidase, a-mannosidase, B-mannosidase, a-galactosidase, B-galactosidase, a-fucosidase, B-hexosaminidase, and combinations-thereof.
9. The method of claim 8 wherein the oligosaccharide chains are sequentially cleaved by: 1) digesting the glycoprotein with a-mannosidase to remove a-mannose residues; 2) digesting the product of step 1) with B-mannosidase to remove B-mannose residues. The method of claim 9 further comprising digesting the glycoprotein with an additional enzyme selected from the group consisting of exoglycosidases and phosphatases.
11. The method of claim 10 wherein the product of step 2 is digested with B-hexosaminidase.
12. The method of claim 2 wherein the oligosaccharide chains are cleaved by sequentially digesting the glycoprotein first with an exoglycosidase and secondly with an endoglycosidase.
13. The method of claim 12 wherein the oligosaccharide chains are cleaved by S 1) digesting the glycoprotein with a-mannosidase to remove a-mannose residues; and 2) digesting the product of step 1) with an enaoglycosidase selected from the group consisting of endo-B-N-acetylglucosaminidase L and endo-8-N-acetylglucosaminidase D.
14. The method of claim 12 for modifying glycoproteins wherein the, 'SA paia I I 40 oligosaccharide chains are cleaved by digesting the glycoprotein with an enzyme selected from the group consisting of sialidase, a-galactosidase, p-galactosidase, 0-hexosaminidase, a-fucosidase, and combinations thereof followed by digestion with an enzyme selected from the group consisting of endo-P-N-acetyl- glucosaminidase D and endo-3-N-acetylglucosaminidase F. The method of claim 2 further comprising cleaving high-mannose oligosaccharide chains with a-mannosidase to remove mannose residues.
16. The method of claim 2 wherein the oligosaccharide chains are cleaved by chemical treatment. *S.
17. The method of claim 16 wherein the oligosaccharide chains are cleaved with a compound selected from the group consisting of trifluoromethane sulfonic acid and hydrofluoric acid.
18. The method of claim 1 further comprising first producing the glycoproteins in cells in the presence of a glycosidase inhibitor.
19. The method of claim 18 wherein the glycosidase inhibitor is selected from the group consisting of deoxymannojirimycin, swainsonine, castanospermine and 0o, deoxynojirimycin. S 20. The method of claim 1 further comprising first producing the glycoprotein in cells with one or more mutations in the oligosaccharide processing pathway.
21. A method for modifying proteins comprising: derivatizing amino acids on the protein with a glycoside or thioglycoside S-X, wherein S is N- acetylglucosamine and X is an aglycone, enzymatically' 1 V44 n/ Af n -41- attaching galactose to the N-acetylglucosamine to form a Gal-GlcNAc sequence; and thereafter attaching a sialic acid residue to the galactose to form a SA-Gal-GlcNAc sequence.
22. The method of claim 1 or 21 wherein the galactose residue is attached to the N-acetylglucosamine ressidue by a galactosyltransferase.
23. The method of claim 22 wherein the galactosyltransferase is selected from the group consisting of UDP-Gal:GlcNAc-R 01-->4 galactosyltransferase and UDP-Gal:GlcNAc-R 31-->3 galactosyltransferase.
24. The method of claim 1 or 21 wherein the galactose is attached to the N-acetylglucosamine by: 1) incubating UDP-Gal:GlcNAc-R 01-->4 galacto- syltransferase with the derivatized protein to attach a terminal galactose to the GlcNAc; 2) incubating the product of step 1 with UDP- GlcNAc:Galpl-->4GlcNAc-R 31-->3 N-acetylglucosaminyl- *o transferase to attach a terminal GlcNAc to the terminal galactose; 3) incubating the product to step 2 with UDP- Gal:GlcNAc-R galactosyltransferase to attach a terminal galactose to the terminal GlcNAc; and 4) repeating steps 2 and 3 until an oligosaccharide a chain (Galpl-->4GlcNAcpl-->3)n units, wherein n is between 1 and 10, is produced. The method of claim 1 or 21 or 24 further comprising attaching fucose to a Gal-->GlcNAc--> sequence.
26. The method of claim 25 wherein the fucose is attached to the Gal-GlcNAc--> sequence with CDP- Fuc:GlcNAc al-->3 fucosyltransferase. 11-12 I I S42
27. The method of claim 1 or 21 further comprising attaching the galactose residues to the N-acetyl- glucosamine in a solution containing a non-ionic detergent, a chaotropic agent, an organic solvent, urea, a protease inhibitor, an exoglycosidase inhibitor, a disulfide bond reducing agent, or a combination thereof.
28. The method of claim 1 wherein the sialic acid residue is attached to the Gal-GlcNAc sequence in an a linkage by a sialyltransferase.
29. The method of claim 28 wherein the sialyltransferase is selected from the group consisting of CMP-SA:Gall-- >4GlcNAc-R 02-->6 sialyltransferase, CMP-SA:Gal3l-->3- (4)GlcNAc a2-->3 sialyltransferase, and CMP--SA:Gal-pl--> 4GlcNAc a2-->3 sialyltransferase.
30. A method for targeting a protein to a cell having a specific surface receptor for a saccharide, said method comprising: attaching to the protein an oligosaccharide chain, seeo said oligosaccharide chain having an exposed saccharide which is SAa2--> and a Gal-->GlcNAc sequence, wherein said exposed SAa2 saccharide is recognized by the cell surface receptor and said oligosaccharide chain is attached to the protein with the Gal-->GlcNAc I sequence.
31. The method of claim 30 wherein the oligosaccharide Schain is a branched oligosaccharide selected from the group of SAa2-->3Galpl-->3(Fucal-->4)GlcNAc and SAa2--> 3GalS1-->3(Fucal-->)GlcNAcl-->4GlcNAc.
32. A protein produced according to the method of any one of claims 1 to 29, said protein comprising an oligosaccharide sequence consisting of: 9 00314 i;L_7- ,jsF i -43- SAa2-->3Ga1J31-->4GlcNAc-->; SAa2-->3Galfll-->4GlcNAc-->; SAa2-->6Galol-->3GlcNAc1-4cc-->; SAa2-->3GalI31-->4GlcNAcI31-->4GlcNAc-->; SAa2-->3Galt31-->3GlcNAc/31-->4GlcNAc-->; SAc2-->3Galfol-->3(Fucal-->4)GlcNAcf31-->4GlcNAc-->; wherein n is betweel n 1 ->3ad 10; ,lc~c-> wherein n is between 1 and C SAa2-->3Galf31-->3[GlcNAc31-->3GaJ431-->4],GlcNAc-->, ***wherein n is between 1 and e~g SAa2-->3Ga.q31-->3(Fucal-->4)GcNAc31-->3Gal31-->4- .:..*GlcNAcf3-->3],GalJ1-->4GlcNAc-->, wherein n is between see l and wherein n is between 1 and SAc2-->3Galfpl-->4(GlcNAcf31-->3Galol-->4),GlcNAC0l-->3Gal, wherein n is between 1 and *33. A glycosylated protein comprising SA-Gal-GlcNAc- protein, wherein the SA is attached to the Gal by an enzyme selected from the group consisting of CMP- SAGll04Gc@- illtaseSe;adCP SA:Galfol-->4GlcNAc-R a2-->3 sialyltransferase; n CP DATED this'l4th day of March, 1990 MASSACHUSETTS INSTITUTE OF TECHNOLOGY by its Patent Attorneys DAVIES COLLISON
AU56271/86A 1986-03-07 1986-03-07 Method for enhancing glycoprotein stability Ceased AU597574B2 (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US1986/000495 WO1987005330A1 (en) 1986-03-07 1986-03-07 Method for enhancing glycoprotein stability

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU51336/90A Division AU624487B2 (en) 1986-03-07 1990-03-14 Method for enhancing glycoprotein stability

Publications (2)

Publication Number Publication Date
AU5627186A AU5627186A (en) 1987-09-28
AU597574B2 true AU597574B2 (en) 1990-06-07

Family

ID=22195409

Family Applications (2)

Application Number Title Priority Date Filing Date
AU56271/86A Ceased AU597574B2 (en) 1986-03-07 1986-03-07 Method for enhancing glycoprotein stability
AU51336/90A Ceased AU624487B2 (en) 1986-03-07 1990-03-14 Method for enhancing glycoprotein stability

Family Applications After (1)

Application Number Title Priority Date Filing Date
AU51336/90A Ceased AU624487B2 (en) 1986-03-07 1990-03-14 Method for enhancing glycoprotein stability

Country Status (5)

Country Link
EP (1) EP0272253A4 (en)
JP (1) JPS63502716A (en)
AU (2) AU597574B2 (en)
DK (1) DK583087A (en)
WO (1) WO1987005330A1 (en)

Families Citing this family (555)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IL80973A (en) * 1985-12-20 1992-08-18 Sanofi Sa Modified ribosome-inactivating glycoproteins,their preparation,immunotoxins containing them and pharmaceutical compositions containing such immunotoxins
EP0400075A4 (en) * 1988-02-10 1991-07-03 Genzyme Corporation Enhancement of the therapeutic properties of glycoprotein
DE3927801A1 (en) * 1989-08-23 1991-02-28 Hoechst Ag PROCESS FOR ENZYMATIC SYNTHESIS OF GALACTOSYLATED GLYCOPROTEIN COMPONENTS
US6319695B1 (en) * 1991-10-15 2001-11-20 The Scripps Research Insitute Production of fucosylated carbohydrates by enzymatic fucosylation synthesis of sugar nucleotides; and in situ regeneration of GDP-fucose
CA2121365C (en) 1991-10-15 2000-11-28 Chi-Huey Wong Production of fucosylated carbohydrates by enzymatic fucosylation synthesis of sugar nucleotides; and in situ regeneration of gdp-fucose
US5877016A (en) 1994-03-18 1999-03-02 Genentech, Inc. Human trk receptors and neurotrophic factor inhibitors
US5708142A (en) 1994-05-27 1998-01-13 Genentech, Inc. Tumor necrosis factor receptor-associated factors
DE4423131A1 (en) * 1994-07-01 1996-01-04 Bayer Ag New hIL-4 mutant proteins as antagonists or partial agonists of human interleukin 4
SK284191B6 (en) 1995-02-24 2004-10-05 Genentech, Inc. Human DNASE I variants
US6020473A (en) * 1995-08-25 2000-02-01 Genentech, Inc. Nucleic acids encoding variants of vascular endothelial cell growth factor
US7005505B1 (en) 1995-08-25 2006-02-28 Genentech, Inc. Variants of vascular endothelial cell growth factor
US6030945A (en) * 1996-01-09 2000-02-29 Genentech, Inc. Apo-2 ligand
US6998116B1 (en) 1996-01-09 2006-02-14 Genentech, Inc. Apo-2 ligand
US6469144B1 (en) 1996-04-01 2002-10-22 Genentech, Inc. Apo-2LI and Apo-3 polypeptides
US6159462A (en) * 1996-08-16 2000-12-12 Genentech, Inc. Uses of Wnt polypeptides
US5851984A (en) * 1996-08-16 1998-12-22 Genentech, Inc. Method of enhancing proliferation or differentiation of hematopoietic stem cells using Wnt polypeptides
US6462176B1 (en) 1996-09-23 2002-10-08 Genentech, Inc. Apo-3 polypeptide
US5990281A (en) * 1996-09-30 1999-11-23 Genentech, Inc. Vertebrate smoothened proteins
US6136958A (en) 1996-09-30 2000-10-24 Genentech, Inc. Antibodies to vertebrate smoothened proteins
DK0942992T3 (en) 1997-01-31 2007-07-02 Genentech Inc O-fucosyltransferase
US6342369B1 (en) 1997-05-15 2002-01-29 Genentech, Inc. Apo-2-receptor
CA2293724C (en) 1997-06-05 2010-02-02 Xiaodong Wang Apaf-1, the ced-4 human homolog, an activator of caspase-3
JP2002508663A (en) 1997-06-18 2002-03-19 ジェネンテク,インコーポレイテッド Apo-2DcR
CA2382376A1 (en) 1997-09-17 1999-03-25 Genentech, Inc. Novel polypeptides and nucleic acids encoding pro287 which are useful for treating disorders of the pancreas
DE69839401T2 (en) 1997-09-18 2009-05-07 Genentech Inc., San Francisco DCR3 POLYPEPTIDE, A TNFR HOMOLOG
IL135051A0 (en) 1997-10-10 2001-05-20 Genentech Inc Apo-3 ligand polypeptide
IL135607A0 (en) 1997-10-29 2001-05-20 Genentech Inc Wnt-1 inducible genes
CA2306183A1 (en) 1997-10-29 1999-05-06 Genentech, Inc. Wnt-1 induced secreted polypeptides: wisp-1, -2 and -3
ATE410512T1 (en) 1997-11-21 2008-10-15 Genentech Inc PLATELE-SPECIFIC ANTIGENS AND THEIR PHARMACEUTICAL USE
US7192589B2 (en) 1998-09-16 2007-03-20 Genentech, Inc. Treatment of inflammatory disorders with STIgMA immunoadhesins
WO1999036535A1 (en) 1998-01-15 1999-07-22 Genentech, Inc. Apo-2 ligand
US6727079B1 (en) 1998-02-25 2004-04-27 The United States Of America As Represented By The Department Of Health And Human Services cDNA encoding a gene BOG (B5T Over-expressed Gene) and its protein product
NZ525914A (en) 1998-03-10 2004-03-26 Genentech Inc Novel polypeptides and nucleic acids encoding the same
DK1064382T3 (en) 1998-03-17 2008-12-08 Genentech Inc Homologous polypeptides for VEGF and BMP1
EP1865061A3 (en) 1998-05-15 2007-12-19 Genentech, Inc. IL-17 homologous polypeptides and therapeutic uses thereof
PT1076703E (en) 1998-05-15 2007-10-10 Genentech Inc Therapeutic uses of il-17 homologous polypeptides
EP3112468A1 (en) 1998-05-15 2017-01-04 Genentech, Inc. Il-17 homologous polypeptides and therapeutic uses thereof
US20020172678A1 (en) 2000-06-23 2002-11-21 Napoleone Ferrara EG-VEGF nucleic acids and polypeptides and methods of use
CA2450402A1 (en) 1998-12-22 2000-06-29 Genentech, Inc. Methods and compositions for inhibiting cancer cell growth comprising pro224
EP2330198A1 (en) 1998-12-23 2011-06-08 Genentech, Inc. IL-1 related polypeptides
EP1978029A3 (en) 1999-06-15 2008-10-15 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids endoding the same
AU2055401A (en) 1999-12-01 2001-06-12 Genentech Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
EP1897947B1 (en) 1999-12-23 2012-01-18 Genentech, Inc. IL-17 homologous polypeptides and therapeutic uses thereof
JP2003519478A (en) 2000-01-10 2003-06-24 マキシゲン・ホールディングズ・リミテッド G-CSF conjugate
ATE424457T1 (en) 2000-01-13 2009-03-15 Genentech Inc HUMAN STRA6 POLYPEPTIDES
DE60138364D1 (en) 2000-02-11 2009-05-28 Bayer Healthcare Llc CLEANING FACTOR VII OR VIIA CONJUGATE
US7101974B2 (en) 2000-03-02 2006-09-05 Xencor TNF-αvariants
WO2004043361A2 (en) 2002-11-08 2004-05-27 Genentech, Inc. Compositions and methods for the treatment of natural killer cell related diseases
EP2168980A1 (en) 2000-06-23 2010-03-31 Genentech, Inc. Compositions and methods for the diagnosis and treatment of disorders involving angiogensis
EP2792747A1 (en) 2000-06-23 2014-10-22 Genentech, Inc. Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
DE60136281D1 (en) 2000-08-24 2008-12-04 Genentech Inc METHOD FOR INHIBITING IL-22-INDUCED PAP1
EP1944317A3 (en) 2000-09-01 2008-09-17 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US6576452B1 (en) 2000-10-04 2003-06-10 Genencor International, Inc. 2,5-diketo-L-gluconic acid reductases and methods of use
US6673580B2 (en) 2000-10-27 2004-01-06 Genentech, Inc. Identification and modification of immunodominant epitopes in polypeptides
US7892730B2 (en) 2000-12-22 2011-02-22 Sagres Discovery, Inc. Compositions and methods for cancer
US7700274B2 (en) 2000-12-22 2010-04-20 Sagres Discovery, Inc. Compositions and methods in cancer associated with altered expression of KCNJ9
US20030232334A1 (en) 2000-12-22 2003-12-18 Morris David W. Novel compositions and methods for cancer
US7820447B2 (en) 2000-12-22 2010-10-26 Sagres Discovery Inc. Compositions and methods for cancer
US7645441B2 (en) 2000-12-22 2010-01-12 Sagres Discovery Inc. Compositions and methods in cancer associated with altered expression of PRLR
MXPA03007619A (en) 2001-02-27 2003-12-04 Maxygen Aps New interferon beta-like molecules.
US20070160576A1 (en) 2001-06-05 2007-07-12 Genentech, Inc. IL-17A/F heterologous polypeptides and therapeutic uses thereof
KR100607612B1 (en) 2001-06-20 2006-08-02 제넨테크, 인크. Compositions and Methods for the Diagnosis and Treatment of Tumor
PL368972A1 (en) 2001-08-29 2005-04-04 Genentech, Inc. Bv8 nucleic acids and polypeptides with mitogenic activity
EP2153843B1 (en) 2001-09-18 2012-08-15 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
US7320789B2 (en) 2001-09-26 2008-01-22 Wyeth Antibody inhibitors of GDF-8 and uses thereof
US7214660B2 (en) 2001-10-10 2007-05-08 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
US7265085B2 (en) 2001-10-10 2007-09-04 Neose Technologies, Inc. Glycoconjugation methods and proteins/peptides produced by the methods
EP2298354B1 (en) 2001-10-10 2014-03-19 ratiopharm GmbH Remodelling and glycoconjugation of interferon-beta
US7439043B2 (en) 2001-10-10 2008-10-21 Neose Technologies, Inc. Galactosyl nucleotide sugars
US7173003B2 (en) 2001-10-10 2007-02-06 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US7297511B2 (en) 2001-10-10 2007-11-20 Neose Technologies, Inc. Interferon alpha: remodeling and glycoconjugation of interferon alpha
US7696163B2 (en) 2001-10-10 2010-04-13 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
US7125843B2 (en) 2001-10-19 2006-10-24 Neose Technologies, Inc. Glycoconjugates including more than one peptide
US7179617B2 (en) 2001-10-10 2007-02-20 Neose Technologies, Inc. Factor IX: remolding and glycoconjugation of Factor IX
US8008252B2 (en) 2001-10-10 2011-08-30 Novo Nordisk A/S Factor VII: remodeling and glycoconjugation of Factor VII
US7265084B2 (en) 2001-10-10 2007-09-04 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
US7795210B2 (en) * 2001-10-10 2010-09-14 Novo Nordisk A/S Protein remodeling methods and proteins/peptides produced by the methods
SG159381A1 (en) 2001-10-10 2010-03-30 Novo Nordisk As Remodeling and glycoconjugation of peptides
US7157277B2 (en) 2001-11-28 2007-01-02 Neose Technologies, Inc. Factor VIII remodeling and glycoconjugation of Factor VIII
US7399613B2 (en) 2001-10-10 2008-07-15 Neose Technologies, Inc. Sialic acid nucleotide sugars
US7226903B2 (en) 2001-10-10 2007-06-05 Neose Technologies, Inc. Interferon beta: remodeling and glycoconjugation of interferon beta
US7473680B2 (en) 2001-11-28 2009-01-06 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
WO2003046150A2 (en) 2001-11-28 2003-06-05 Neose Technologies, Inc. Glycoprotein remodeling using endoglycanases
US20030228305A1 (en) 2002-01-02 2003-12-11 Genentech, Inc. Compositions and methods for the diagnosis and treatment of tumor
JP4429728B2 (en) 2002-02-21 2010-03-10 ワイス エルエルシー Follistatin domain-containing protein
EP2388265A1 (en) 2002-02-22 2011-11-23 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
CA2477249A1 (en) 2002-02-25 2003-09-04 Genentech, Inc. Novel type-1 cytokine receptor glm-r
US20040023267A1 (en) 2002-03-21 2004-02-05 Morris David W. Novel compositions and methods in cancer
EP1571968A4 (en) 2002-04-16 2007-10-17 Genentech Inc Compositions and methods for the diagnosis and treatment of tumor
EP2305710A3 (en) 2002-06-03 2013-05-29 Genentech, Inc. Synthetic antibody phage libraries
JP2005528905A (en) 2002-06-07 2005-09-29 ジェネンテック・インコーポレーテッド Compositions and methods for tumor diagnosis and treatment
EP2383278A1 (en) 2002-07-08 2011-11-02 Genentech, Inc. Method to determine B cell mediated diseases
US7364870B2 (en) 2002-08-02 2008-04-29 Wyeth MK2 interacting proteins
CN100594239C (en) 2002-08-12 2010-03-17 金克克国际有限公司 Mutant E. coli APPA phytase enzymes
JP5401001B2 (en) 2002-09-11 2014-01-29 ジェネンテック, インコーポレイテッド Novel compositions and methods for the treatment of immune related diseases
WO2004024072A2 (en) 2002-09-11 2004-03-25 Genentech, Inc. Novel compositions and methods for the treatment of immune related diseases
EP2444409A2 (en) 2002-09-16 2012-04-25 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
US20070042945A1 (en) 2002-09-25 2007-02-22 Genentech, Inc. Nouvelles compositions et methods de traitement du psoriasis
EP1576137A4 (en) 2002-10-29 2010-06-30 Genentech Inc Compositions and methods for the treatment of immune related diseases
US20070048301A1 (en) 2002-11-26 2007-03-01 Bodary-Winter Sarah C Compositions and methods for the treatment of immune related diseases
TWI335920B (en) 2002-12-24 2011-01-11 Yasuhiro Kajihara Sugar chain asparagine derivatives, sugar chain asparagine and sugar chain and manufacture thereof
US7553930B2 (en) 2003-01-06 2009-06-30 Xencor, Inc. BAFF variants and methods thereof
US20040170982A1 (en) 2003-02-14 2004-09-02 Morris David W. Novel therapeutic targets in cancer
US7767387B2 (en) 2003-06-13 2010-08-03 Sagres Discovery, Inc. Therapeutic targets in cancer
JP2007524362A (en) 2003-02-14 2007-08-30 サイグレス ディスカバリー, インコーポレイテッド Therapeutic GPCR targets in cancer
FR2851471B1 (en) * 2003-02-24 2006-07-28 Synt Em COMPOUNDS COMPRISING AT LEAST ONE ACTIVE SUBSTANCE AND AT LEAST ONE VECTOR CONNECTED BY A BINDING AGENT, USES THEREOF AND THE BINDING AGENTS
JP4912144B2 (en) 2003-03-12 2012-04-11 ジェネンテック, インコーポレイテッド Use of BV8 and / or EG-VEGF to promote hematopoiesis
NZ542306A (en) 2003-03-14 2008-04-30 Wyeth Corp Antibodies against human IL-21 receptor and uses therefor
CA2519092C (en) 2003-03-14 2014-08-05 Neose Technologies, Inc. Branched water-soluble polymers and their conjugates
ATE549359T1 (en) 2003-04-02 2012-03-15 Hoffmann La Roche ANTIBODIES TO INSULIN-LIKE GROWTH FACTOR I RECEPTOR AND THEIR USES
US8791070B2 (en) 2003-04-09 2014-07-29 Novo Nordisk A/S Glycopegylated factor IX
WO2006127896A2 (en) 2005-05-25 2006-11-30 Neose Technologies, Inc. Glycopegylated factor ix
EP1615945B1 (en) 2003-04-09 2011-09-28 BioGeneriX AG Glycopegylation methods and proteins/peptides produced by the methods
WO2004091499A2 (en) 2003-04-09 2004-10-28 Neose Technologies, Inc. Intracellular formation of peptide conjugates
US20050025763A1 (en) 2003-05-08 2005-02-03 Protein Design Laboratories, Inc. Therapeutic use of anti-CS1 antibodies
US7709610B2 (en) 2003-05-08 2010-05-04 Facet Biotech Corporation Therapeutic use of anti-CS1 antibodies
ATE540055T1 (en) 2003-05-09 2012-01-15 Biogenerix Ag COMPOSITIONS AND METHODS FOR PRODUCING HUMAN GROWTH HORMONE GLYCOSYLATION MUTANTS
KR20180014881A (en) 2003-05-30 2018-02-09 제넨테크, 인크. Treatment with anti-VEGF antibodies
PL2784084T3 (en) 2003-07-08 2020-03-31 Genentech, Inc. Antagonist antibodies to IL-17A/F heterologous polypeptides
US7579157B2 (en) 2003-07-10 2009-08-25 Hoffmann-La Roche Inc. Antibody selection method against IGF-IR
US9005625B2 (en) 2003-07-25 2015-04-14 Novo Nordisk A/S Antibody toxin conjugates
WO2005019258A2 (en) 2003-08-11 2005-03-03 Genentech, Inc. Compositions and methods for the treatment of immune related diseases
US8399618B2 (en) 2004-10-21 2013-03-19 Xencor, Inc. Immunoglobulin insertions, deletions, and substitutions
US8883147B2 (en) 2004-10-21 2014-11-11 Xencor, Inc. Immunoglobulins insertions, deletions, and substitutions
EP1675871A2 (en) 2003-10-10 2006-07-05 Xencor Inc. Protein based tnf-alpha variants for the treatment of tnf-alpha related disorders
SI2161283T1 (en) 2003-11-17 2014-10-30 Genentech, Inc. Compositions comprising antibodies against CD79b conjugated to a growth inhibitory agent or cytotoxic agent and methods for the treatment of tumor of hematopoietic origin
US20080305992A1 (en) 2003-11-24 2008-12-11 Neose Technologies, Inc. Glycopegylated erythropoietin
US8633157B2 (en) 2003-11-24 2014-01-21 Novo Nordisk A/S Glycopegylated erythropoietin
CA2547140A1 (en) 2003-11-24 2005-06-09 Neose Technologies, Inc. Glycopegylated erythropoietin
US7956032B2 (en) 2003-12-03 2011-06-07 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
TR201815885T4 (en) 2004-01-07 2018-11-21 Novartis Vaccines & Diagnostics Inc M-csf specific monoclonal antibodies and their use.
CN101072789B (en) 2004-01-08 2013-05-15 生物种属学股份公司 O-linked glycosylation of peptides
PT1737891E (en) 2004-04-13 2013-04-16 Hoffmann La Roche Anti-p-selectin antibodies
EP1745141B2 (en) 2004-05-04 2019-09-25 Novo Nordisk Health Care AG O-linked glycoforms of faktor vii and method to manufacture them
US20080300173A1 (en) 2004-07-13 2008-12-04 Defrees Shawn Branched Peg Remodeling and Glycosylation of Glucagon-Like Peptides-1 [Glp-1]
EP2361931B1 (en) 2004-07-20 2017-12-06 Genentech, Inc. Inhibitors of angiopoietin-like 4 protein, combinations, and their use
US20060024677A1 (en) 2004-07-20 2006-02-02 Morris David W Novel therapeutic targets in cancer
EP2332977B1 (en) 2004-07-23 2015-11-25 Acceleron Pharma Inc. ActRII receptor polypeptides
WO2006031811A2 (en) 2004-09-10 2006-03-23 Neose Technologies, Inc. Glycopegylated interferon alpha
HUE026826T2 (en) 2004-10-29 2016-07-28 Ratiopharm Gmbh Remodeling and glycopegylation of fibroblast growth factor (FGF)
EP1817340B1 (en) 2004-11-12 2012-05-16 Xencor, Inc. Fc variants with altered binding to fcrn
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
CA2589374C (en) 2004-11-30 2016-05-03 Curagen Corporation Antibodies directed to gpnmb and uses thereof
ES2449195T3 (en) 2005-01-10 2014-03-18 Ratiopharm Gmbh Glycopegylated granulocyte colony stimulating factor
KR101335798B1 (en) 2005-02-15 2013-12-02 듀크 유니버시티 Anti-cd19 antibodies and uses in oncology
US9820986B2 (en) * 2005-03-04 2017-11-21 Taiwan Hopaz Chems, Mfg. Co., Ltd. Glycopeptide compositions
TW200720289A (en) 2005-04-01 2007-06-01 Hoffmann La Roche Antibodies against CCR5 and uses thereof
CA2604885A1 (en) 2005-04-07 2006-10-19 Guoying Yu Cacna1e in cancer diagnosis, detection and treatment
AU2006235258A1 (en) 2005-04-07 2006-10-19 Novartis Vaccines And Diagnostics Inc. Cancer-related genes
WO2006121569A2 (en) 2005-04-08 2006-11-16 Neose Technologies, Inc. Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
JP5047947B2 (en) 2005-05-05 2012-10-10 デューク ユニバーシティ Anti-CD19 antibody treatment for autoimmune disease
EP1888098A2 (en) 2005-05-25 2008-02-20 Neose Technologies, Inc. Glycopegylated erythropoietin formulations
PL2460831T3 (en) 2005-05-27 2017-05-31 Biogen Ma Inc. Tweak binding antibodies
WO2008018854A2 (en) 2005-06-06 2008-02-14 The Rockefeller University Bactiophage lysins for bacillus anthracis
AU2006255686A1 (en) 2005-06-06 2006-12-14 Genentech, Inc. Transgenic models for different genes and their use for gene characterization
US7582291B2 (en) 2005-06-30 2009-09-01 The Rockefeller University Bacteriophage lysins for Enterococcus faecalis, Enterococcus faecium and other bacteria
CA2619577A1 (en) 2005-08-15 2007-02-22 Genentech, Inc. Gene disruptions, compositions and methods relating thereto
US20070105755A1 (en) 2005-10-26 2007-05-10 Neose Technologies, Inc. One pot desialylation and glycopegylation of therapeutic peptides
EP1917355B1 (en) 2005-08-24 2010-12-08 The Rockefeller University Ply-gbs mutant lysins
US20090048440A1 (en) 2005-11-03 2009-02-19 Neose Technologies, Inc. Nucleotide Sugar Purification Using Membranes
SI1948798T1 (en) 2005-11-18 2015-09-30 Glenmark Pharmaceuticals S.A. Anti-alpha2 integrin antibodies and their uses
US20090293137A1 (en) 2005-11-21 2009-11-26 Genentech, Inc. Novel Gene Disruptions, Compositions and Methods Relating Thereto
DK1973559T3 (en) 2005-11-23 2013-03-25 Acceleron Pharma Inc Activin-ActRIIa antagonists as well as applications for bone growth
US20080311107A1 (en) 2006-02-17 2008-12-18 Genetech, Inc. Novel Gene Disruptions, Compositions and Methods Relating Thereto
TWI417301B (en) 2006-02-21 2013-12-01 Wyeth Corp Antibodies against human il-22 and uses therefor
TW200744634A (en) 2006-02-21 2007-12-16 Wyeth Corp Methods of using antibodies against human IL-22
US8389688B2 (en) 2006-03-06 2013-03-05 Aeres Biomedical, Ltd. Humanized anti-CD22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
JP2009529915A (en) 2006-03-20 2009-08-27 ゾーマ テクノロジー リミテッド Human antibodies and methods specific for gastrin substances
US7846724B2 (en) 2006-04-11 2010-12-07 Hoffmann-La Roche Inc. Method for selecting CHO cell for production of glycosylated antibodies
EP2082645A1 (en) 2006-04-19 2009-07-29 Genentech, Inc. Novel gene disruptions, compositions and methods relating thereto
TWI395754B (en) 2006-04-24 2013-05-11 Amgen Inc Humanized c-kit antibody
ES2516694T3 (en) 2006-07-21 2014-10-31 Ratiopharm Gmbh Glycosylation of peptides through glycosylation sequences with O-binding
EP2420252A1 (en) 2006-08-04 2012-02-22 Novartis AG EPHB3-specific antibody and uses thereof
CA2661023C (en) 2006-08-18 2017-08-15 Novartis Ag Prlr-specific antibody and uses thereof
EP2492278A1 (en) 2006-09-18 2012-08-29 Compugen Ltd. Antibodies against an agonist of G-protein coupled receptors and its use indiagnosis and therapy
ATE550355T1 (en) 2006-09-29 2012-04-15 Hoffmann La Roche ANTIBODIES TO CCR5 AND THEIR USE
US7767206B2 (en) 2006-10-02 2010-08-03 Amgen Inc. Neutralizing determinants of IL-17 Receptor A and antibodies that bind thereto
US8969532B2 (en) 2006-10-03 2015-03-03 Novo Nordisk A/S Methods for the purification of polypeptide conjugates comprising polyalkylene oxide using hydrophobic interaction chromatography
KR101556248B1 (en) 2006-10-04 2015-09-30 노보 노르디스크 에이/에스 Glycerol linked pegylated sugars and glycopeptides
MX2009004718A (en) 2006-11-02 2009-06-19 Acceleron Pharma Inc Alk1 receptor and ligand antagonists and uses thereof.
US20080254512A1 (en) 2006-11-02 2008-10-16 Capon Daniel J Hybrid immunoglobulins with moving parts
EA200900767A1 (en) 2006-12-07 2009-12-30 Новартис Аг ANTAGONISTIC ANTIBODIES AGAINST EPHB3
TWI782836B (en) 2007-02-02 2022-11-01 美商艾瑟勒朗法瑪公司 Variants derived from actriib and uses therefor
TWI459963B (en) 2007-02-09 2014-11-11 Acceleron Pharma Inc Pharmaceutical composition comprising an actriia-fc fusion protein; use of an actriia-fc fusion protein for treatment or prevention of cancer-related bone loss;use of an actriia-fc fusion protein for the treatment or prevention of multiple myeloma
WO2008103962A2 (en) 2007-02-22 2008-08-28 Genentech, Inc. Methods for detecting inflammatory bowel disease
PL2144923T3 (en) 2007-04-03 2013-12-31 Biogenerix Ag Methods of treatment using glycopegylated g-csf
EP2737907A3 (en) 2007-05-07 2014-11-05 MedImmune, LLC Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
US9493499B2 (en) 2007-06-12 2016-11-15 Novo Nordisk A/S Process for the production of purified cytidinemonophosphate-sialic acid-polyalkylene oxide (CMP-SA-PEG) as modified nucleotide sugars via anion exchange chromatography
JP5355561B2 (en) 2007-07-12 2013-11-27 コンピュゲン エルティーディー. Biologically active peptides and methods of use thereof
CN111499748A (en) 2007-07-16 2020-08-07 健泰科生物技术公司 anti-CD 79B antibodies and immunoconjugates and methods of use
PE20090481A1 (en) 2007-07-16 2009-05-18 Genentech Inc ANTI-CD79B ANTIBODIES AND HUMANIZED IMMUNOCONJUGATES AND METHODS OF USE
CN101361968B (en) 2007-08-06 2011-08-03 健能隆医药技术(上海)有限公司 Use of interleukin-22 in treating fatty liver
NO2188313T3 (en) 2007-08-21 2018-03-31
US8207112B2 (en) 2007-08-29 2012-06-26 Biogenerix Ag Liquid formulation of G-CSF conjugate
UY31309A1 (en) 2007-08-29 2009-03-31 ANTI-CXCR5 HUMANIZED ANTIBODIES, DERIVED FROM THE SAME AND ITS USE
EP2769728A1 (en) 2007-09-04 2014-08-27 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
TW200918553A (en) 2007-09-18 2009-05-01 Amgen Inc Human GM-CSF antigen binding proteins
CN107412734A (en) 2007-09-18 2017-12-01 阿塞勒隆制药公司 Activin A CTRIIA antagonists and the purposes for reducing or suppressing FSH secretions
EP2050764A1 (en) 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
JP5771005B2 (en) 2007-10-30 2015-08-26 インディアナ ユニバーシティー リサーチ アンド テクノロジー コーポレーションIndiana University Research And Technology Corporation Glucagon antagonist and compound showing GLP-1 agonist activity
EP2261254A3 (en) 2007-12-21 2011-04-13 Amgen, Inc Anti-amyloid antibodies and uses thereof
KR101616758B1 (en) 2007-12-26 2016-04-29 젠코어 인코포레이티드 Fc VARIANTS WITH ALTERED BINDING TO FcRn
US9181327B2 (en) 2008-01-07 2015-11-10 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Anti-HIV domain antibodies and method of making and using same
US9175078B2 (en) 2008-01-25 2015-11-03 Amgen Inc. Ferroportin antibodies and methods of use
ES2848323T3 (en) 2008-01-31 2021-08-06 Inst Nat Sante Rech Med Antibodies against human CD39 and their use to inhibit the activity of regulatory T cells
EP3269737B1 (en) 2008-01-31 2024-06-19 Genentech, Inc. Anti-cd79b antibodies and immunoconjugates and methods of use
WO2009108806A1 (en) 2008-02-27 2009-09-03 Novo Nordisk A/S Conjugated factor viii molecules
KR101855381B1 (en) 2008-04-09 2018-05-09 제넨테크, 인크. Novel compositions and methods for the treatment of immune related diseases
ES2487846T3 (en) 2008-05-01 2014-08-25 Amgen, Inc. Anti-hepcindin antibodies and methods of use
AU2009241755B2 (en) 2008-05-02 2015-10-01 Acceleron Pharma Inc. Methods and compositions based on ALK1 antagonists for modulating angiogenesis and pericyte coverage
US8093018B2 (en) 2008-05-20 2012-01-10 Otsuka Pharmaceutical Co., Ltd. Antibody identifying an antigen-bound antibody and an antigen-unbound antibody, and method for preparing the same
PE20100056A1 (en) 2008-06-17 2010-01-26 Univ Indiana Res & Tech Corp GLUCAGON ANALOGS AS GIP AGONISTS
EP2300037B1 (en) 2008-06-17 2016-03-30 Indiana University Research and Technology Corporation Glucagon/glp-1 receptor co-agonists
CN104447980A (en) 2008-06-17 2015-03-25 印第安纳大学研究及科技有限公司 Glucagon analogs exhibiting enhanced solubility and stability in physiological pH buffers
AU2009262199B2 (en) 2008-06-27 2012-08-09 Amgen Inc. Ang-2 inhibition to treat multiple sclerosis
TWI626945B (en) 2008-08-14 2018-06-21 艾瑟勒朗法瑪公司 Use of gdf traps to increase red blood cell levels
AU2008361352B2 (en) 2008-09-07 2013-05-09 Glyconex Inc. Anti-extended Type I glycosphingolipid antibody, derivatives thereof and use
WO2010040766A1 (en) 2008-10-07 2010-04-15 INSERM (Institut National de la Santé et de la Recherche Médicale) Neutralizing antibodies and fragments thereof directed against platelet factor-4 variant 1 (pf4v1)
AU2009312532B2 (en) 2008-11-06 2013-05-16 Ichnos Sciences SA Treatment with anti-alpha2 integrin antibodies
CN102438652B (en) 2008-11-12 2014-08-13 米迪缪尼有限公司 Antibody formulation
EP2865689A1 (en) 2008-12-08 2015-04-29 Compugen Ltd. FAM26F polypeptides and polynucleotides, and uses thereof as a drug target for producing drugs and biologics
RU2550696C2 (en) 2008-12-19 2015-05-10 Индиана Юниверсити Рисерч Энд Текнолоджи Корпорейшн Amide-based prodrugs of glucagon superfamily peptides
CN102245624B (en) 2008-12-19 2016-08-10 印第安纳大学研究及科技有限公司 Insulin prodrug based on amide
JO3382B1 (en) 2008-12-23 2019-03-13 Amgen Inc Human cgrp receptor binding antibodies
CA2752647A1 (en) 2009-02-24 2010-09-02 The Salk Institute For Biological Studies Designer ligands of tgf-beta superfamily
US20120064096A1 (en) 2009-03-17 2012-03-15 Universite De La Mediterranee BTLA Antibodies and Uses Thereof
WO2010108153A2 (en) 2009-03-20 2010-09-23 Amgen Inc. Carrier immunoglobulins and uses thereof
EP2233500A1 (en) 2009-03-20 2010-09-29 LFB Biotechnologies Optimized Fc variants
US8563513B2 (en) 2009-03-27 2013-10-22 Van Andel Research Institute Parathyroid hormone peptides and parathyroid hormone-related protein peptides and methods of use
CN107375919B (en) 2009-03-27 2022-07-29 台湾地区“中央研究院” Methods and compositions for antiviral immunization
EP3702001A1 (en) 2009-03-30 2020-09-02 Acceleron Pharma Inc. Bmp-alk3 antagonists and uses for promoting bone growth
JP6132548B2 (en) 2009-04-01 2017-05-24 ジェネンテック, インコーポレイテッド Anti-FcRH5 antibodies and immunoconjugates and methods of use
US8580732B2 (en) 2009-04-07 2013-11-12 Duke University Peptide therapy for hyperglycemia
EP2248903A1 (en) 2009-04-29 2010-11-10 Universitat Autònoma De Barcelona Methods and reagents for efficient and targeted gene transfer to monocytes and macrophages
EP2440576A4 (en) 2009-06-08 2013-11-20 Acceleron Pharma Inc Methods for increasing thermogenic adipocytes
KR20210034684A (en) 2009-06-12 2021-03-30 악셀레론 파마 인코포레이티드 TRUNCATED ActRIIB-FC FUSION PROTEINS
SG176858A1 (en) 2009-06-16 2012-02-28 Univ Indiana Res & Tech Corp Gip receptor-active glucagon compounds
EP3838919A1 (en) 2009-08-13 2021-06-23 Acceleron Pharma Inc. Combined use of gdf traps and erythropoietin receptor activators to increase red blood cell levels
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
KR20120062874A (en) 2009-09-09 2012-06-14 악셀레론 파마 인코포레이티드 Actriib antagonists and dosing and uses thereof
WO2011032099A1 (en) 2009-09-11 2011-03-17 The Board Of Trustees Of The University Of Illinois Methods of treating diastolic dysfunction and related conditions
US9885711B2 (en) 2009-09-25 2018-02-06 Xoma Technology Ltd. Screening methods
US8926976B2 (en) 2009-09-25 2015-01-06 Xoma Technology Ltd. Modulators
TW201117824A (en) 2009-10-12 2011-06-01 Amgen Inc Use of IL-17 receptor a antigen binding proteins
EP2488643A4 (en) 2009-10-15 2013-07-03 Hoffmann La Roche Chimeric fibroblast growth factors with altered receptor specificity
CN102711826B (en) 2009-10-22 2017-03-29 霍夫曼-拉罗奇有限公司 For the method and composition that the HEPSIN for regulating and controlling macrophage-stimulating albumen is activated
JP5711751B2 (en) 2009-10-26 2015-05-07 エフ.ホフマン−ラ ロシュ アーゲーF. Hoffmann−La Roche Aktiengesellschaft Process for the preparation of glycosylated immunoglobulins
JO3244B1 (en) 2009-10-26 2018-03-08 Amgen Inc Human il-23 antigen binding proteins
WO2011056494A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor-like kinase-1 antagonist and vegfr3 antagonist combinations
WO2011056502A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Bone morphogenetic protein receptor type ii compositions and methods of use
WO2011056497A1 (en) 2009-10-26 2011-05-12 Genentech, Inc. Activin receptor type iib compositions and methods of use
WO2011056572A1 (en) 2009-10-27 2011-05-12 The Board Of Trustees Of The University Of Illinois Methods of diagnosing diastolic dysfunction
US20110129469A1 (en) 2009-11-03 2011-06-02 Acceleron Pharma Inc. Methods for treating fatty liver disease
EP2501400B1 (en) 2009-11-17 2017-11-01 Acceleron Pharma, Inc. Actriib proteins and variants and uses therefore relating to utrophin induction for muscular dystrophy therapy
AU2010321832B2 (en) 2009-11-20 2014-08-14 Amgen Inc. Anti-Orai1 antigen binding proteins and uses thereof
PE20121584A1 (en) 2009-11-30 2012-11-29 Genentech Inc COMPOSITIONS AND METHODS FOR THE DIAGNOSIS AND TREATMENT OF TUMORS
EP2507265B1 (en) 2009-12-01 2016-05-11 Compugen Ltd. Antibody specific for heparanase splice variant T5 and its use.
BR112012013330A2 (en) 2009-12-02 2017-03-28 Acceleron Pharma Inc compositions and methods for increasing fc fusion protein serum half life
UA109888C2 (en) 2009-12-07 2015-10-26 ANTIBODY OR ANTIBODILITY ANTIBODY OR ITS BINDING TO THE β-CLOTE, FGF RECEPTORS AND THEIR COMPLEXES
WO2011075393A2 (en) 2009-12-18 2011-06-23 Indiana University Research And Technology Corporation Glucagon/glp-1 receptor co-agonists
US8551946B2 (en) 2010-01-27 2013-10-08 Indiana University Research And Technology Corporation Glucagon antagonist-GIP agonist conjugates and compositions for the treatment of metabolic disorders and obesity
WO2011097527A2 (en) 2010-02-04 2011-08-11 Xencor, Inc. Immunoprotection of therapeutic moieties using enhanced fc regions
JP5972176B2 (en) 2010-02-23 2016-08-17 サノフイ Anti-alpha2 integrin antibodies and uses thereof
AR080243A1 (en) 2010-02-23 2012-03-21 Genentech Inc COMPOSITIONS AND METHODS FOR DIAGNOSIS AND TUMOR TREATMENT
NZ602634A (en) 2010-03-26 2015-06-26 Dartmouth College Vista regulatory t cell mediator protein, vista binding agents and use thereof
US20150231215A1 (en) 2012-06-22 2015-08-20 Randolph J. Noelle VISTA Antagonist and Methods of Use
US10745467B2 (en) 2010-03-26 2020-08-18 The Trustees Of Dartmouth College VISTA-Ig for treatment of autoimmune, allergic and inflammatory disorders
AU2011239689A1 (en) 2010-04-15 2012-11-08 Amgen Inc. Human FGF receptor and beta-Klotho binding proteins
MA34291B1 (en) 2010-05-03 2013-06-01 Genentech Inc COMPOSITIONS AND METHODS FOR DIAGNOSING AND TREATING A TUMOR
JP2013528374A (en) 2010-05-10 2013-07-11 パーシード セラピューティクス リミテッド ライアビリティ カンパニー Polypeptide inhibitors of VLA4
CN103079587B (en) 2010-05-13 2016-05-11 印第安纳大学研究及科技有限公司 Present the hyperglycemic factor superfamily peptide of nuclear hormone receptor activity
AU2011268146A1 (en) 2010-06-17 2013-01-10 Actogenix Nv Compositions and methods for treating inflammatory conditions
JP5912112B2 (en) 2010-06-24 2016-04-27 インディアナ ユニバーシティー リサーチ アンド テクノロジー コーポレーションIndiana University Research And Technology Corporation Amide insulin prodrug
CA2796894A1 (en) 2010-06-24 2011-12-29 Indiana University Research And Technology Corporation Amide based glucagon superfamily peptide prodrugs
EP2588123A2 (en) 2010-06-30 2013-05-08 Compugen Ltd. C1orf32 for the treatment of multiple sclerosis, rheumatoid arthritis and other autoimmune disorders
EP2596025B1 (en) 2010-07-23 2018-06-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for cancer management targeting co-029
EP2598530A2 (en) 2010-07-29 2013-06-05 Xencor, Inc. Antibodies with modified isoelectric points
US8999335B2 (en) 2010-09-17 2015-04-07 Compugen Ltd. Compositions and methods for treatment of drug resistant multiple myeloma
CN103339145A (en) 2010-09-22 2013-10-02 安姆根有限公司 Carrier immunoglobulins and uses thereof
EP3828205A1 (en) 2010-10-01 2021-06-02 Oxford BioTherapeutics Ltd Anti-ror1 antibodies
TWI537385B (en) * 2010-11-04 2016-06-11 中央研究院 Methods for producing virus particles with simplified glycosylation of surface proteins
WO2012080769A1 (en) 2010-12-15 2012-06-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Anti-cd277 antibodies and uses thereof
ES2684602T3 (en) 2010-12-22 2018-10-03 Orega Biotech Antibodies against human CD39 and use thereof
ES2713952T3 (en) 2010-12-22 2019-05-24 Univ Indiana Res & Tech Corp Glucagon analogs showing GIP receptor activity
US20140056811A1 (en) 2010-12-27 2014-02-27 Compugen Ltd. New cell-penetrating peptides and uses thereof
JOP20210044A1 (en) 2010-12-30 2017-06-16 Takeda Pharmaceuticals Co Anti-cd38 antibodies
WO2012101125A1 (en) 2011-01-24 2012-08-02 INSERM (Institut National de la Santé et de la Recherche Médicale) Specific antibodies against human cxcl4 and uses thereof
TWI700093B (en) 2011-03-16 2020-08-01 法商賽諾菲公司 Uses of a dual v region antibody-like protein
SG193428A1 (en) 2011-03-31 2013-10-30 Inst Nat Sante Rech Med Antibodies directed against icos and uses thereof
CN103596974B (en) 2011-04-15 2016-08-31 卡姆普根有限公司 Polypeptide and polynucleotide and for treating immunity related disorder and the purposes of cancer
CN108178789B (en) 2011-04-20 2021-11-02 阿塞勒隆制药公司 Endoglin polypeptides and uses thereof
KR102101806B1 (en) 2011-05-19 2020-04-20 인쎄름 (엥스띠뛰 나씨오날 드 라 쌍떼 에 드 라 흐쉐르슈 메디깔) Anti-human-her3 antibodies and uses thereof
EP2714735B1 (en) 2011-06-03 2021-07-21 XOMA Technology Ltd. Antibodies specific for tgf-beta
US9574002B2 (en) 2011-06-06 2017-02-21 Amgen Inc. Human antigen binding proteins that bind to a complex comprising β-Klotho and an FGF receptor
GEP20176629B (en) 2011-06-22 2017-02-27 Indiana Unversity Research And Tech Corporation Glucagon/glp-1 receptor co-agonists
WO2013001517A1 (en) 2011-06-30 2013-01-03 Compugen Ltd. Polypeptides and uses thereof for treatment of autoimmune disorders and infection
CN113683705A (en) 2011-07-01 2021-11-23 恩格姆生物制药公司 Compositions, uses and methods for the treatment of metabolic disorders and diseases
EP2543677A1 (en) 2011-07-08 2013-01-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies for the treatment and prevention of thrombosis
EP2543679A1 (en) 2011-07-08 2013-01-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies for the treatment and prevention of thrombosis
EP2543678A1 (en) 2011-07-08 2013-01-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies for the treatment and prevention of thrombosis
US20140234330A1 (en) 2011-07-22 2014-08-21 Amgen Inc. Il-17 receptor a is required for il-17c biology
WO2013022855A1 (en) 2011-08-05 2013-02-14 Xencor, Inc. Antibodies with modified isoelectric points and immunofiltering
UA118332C2 (en) 2011-09-22 2019-01-10 Емджен Інк. Cs27l antigen binding proteins
JP2014531605A (en) 2011-10-06 2014-11-27 ザ ボード オブ トラスティーズ オブ ザ ユニバーシティ オブ イリノイ Myosin binding protein-C for use in methods associated with diastolic heart failure
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
DK2766392T3 (en) 2011-10-10 2019-10-07 Xencor Inc PROCEDURE FOR CLEANING ANTIBODIES
EA034563B1 (en) 2011-10-17 2020-02-20 Акселерон Фарма Инк. Method for treating or preventing iron overload in a patient having thalassemia
KR20140097151A (en) 2011-11-17 2014-08-06 인디애나 유니버시티 리서치 앤드 테크놀로지 코퍼레이션 Glucagon superfamily peptides exhibiting glucocorticoid receptor activity
CN104220460A (en) 2011-12-08 2014-12-17 安姆根有限公司 Agonistic human lcat antigen binding proteins and uses thereof in therapy
CN104039341A (en) 2011-12-16 2014-09-10 卡乐斯治疗公司 Methods and uses of anp (atrial natriuretic peptide), bnp (brain natriuretic peptide) and cnp (c-type natriuretic peptide)-related peptides and derivatives thereof for treatment of retinal disorders and diseases
WO2013096386A1 (en) 2011-12-20 2013-06-27 Indiana University Research And Technology Corporation Ctp-based insulin analogs for treatment of diabetes
IN2014KN00848A (en) 2012-02-01 2015-10-02 Compugen Ltd
KR20140123558A (en) 2012-02-02 2014-10-22 악셀레론 파마 인코포레이티드 Alk1 antagonists and their uses in treating renal cell carcinoma
WO2013136193A2 (en) 2012-03-16 2013-09-19 University Health Network Methods and compositions for modulating toso activity
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
US9809636B2 (en) 2012-04-06 2017-11-07 Acceleron Pharma Inc. Methods for increasing red blood cell levels comprising administering BMP9
WO2013155346A1 (en) 2012-04-11 2013-10-17 The Regents Of The University Of California Diagnostic tools for response to 6-thiopurine therapy
KR101932697B1 (en) 2012-04-27 2019-03-21 노보 노르디스크 에이/에스 Human cd30 ligand antigen binding proteins
EP2847219A1 (en) 2012-05-07 2015-03-18 Amgen Inc. Anti-erythropoietin antibodies
JO3623B1 (en) 2012-05-18 2020-08-27 Amgen Inc St2 antigen binding proteins
WO2013188740A1 (en) 2012-06-14 2013-12-19 Ambrx, Inc. Anti-psma antibodies conjugated to nuclear receptor ligand polypeptides
TWI599575B (en) 2012-06-21 2017-09-21 印第安納大學科技研究公司 Glucagon analogs exhibiting gip receptor activity
TR201808818T4 (en) 2012-06-21 2018-07-23 Novo Nordisk As Glucagon analogs exhibiting gip receptor activity.
US9890215B2 (en) 2012-06-22 2018-02-13 King's College London Vista modulators for diagnosis and treatment of cancer
EP2864355B1 (en) 2012-06-25 2016-10-12 Orega Biotech Il-17 antagonist antibodies
US20140004121A1 (en) 2012-06-27 2014-01-02 Amgen Inc. Anti-mesothelin binding proteins
EP3613765A1 (en) 2012-08-03 2020-02-26 Dana-Farber Cancer Institute, Inc. Antibody against repulsive guidance molecule b (rgmb)
US9695247B2 (en) 2012-09-03 2017-07-04 Inserm (Institut National De La Sante Et De La Recherche Medicale) Antibodies directed against ICOS for treating graft-versus-host disease
EP2892558B1 (en) 2012-09-07 2019-04-10 The Trustees Of Dartmouth College Vista modulators for diagnosis and treatment of cancer
CN113604550A (en) 2012-10-24 2021-11-05 细胞基因公司 Biomarkers for treating anemia
KR102279522B1 (en) 2012-11-02 2021-07-19 셀진 코포레이션 Activin-actrii antagonists and uses for treating bone and other disorders
MY171100A (en) 2012-11-20 2019-09-25 Sanofi Sa Anti-ceacam5 antibodies and uses thereof
CA2892152A1 (en) 2012-11-28 2014-06-05 Ngm Biopharmaceuticals, Inc. Compositions and methods for treatment of metabolic disorders and diseases
TW201425336A (en) 2012-12-07 2014-07-01 Amgen Inc BCMA antigen binding proteins
MX363407B (en) 2012-12-10 2019-03-22 Biogen Ma Inc Anti-blood dendritic cell antigen 2 antibodies and uses thereof.
CN108888757A (en) 2012-12-27 2018-11-27 恩格姆生物制药公司 Method for adjusting bile acid homeostasis and treating bile acid disorder and disease
EA201591219A1 (en) 2012-12-27 2015-12-30 Санофи ANTIBODIES AGAINST LAMP1 AND CONJUGATES ANTIBODIES AND MEDICINES AND THEIR APPLICATION
US10717965B2 (en) 2013-01-10 2020-07-21 Gloriana Therapeutics, Inc. Mammalian cell culture-produced neublastin antibodies
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
CN105051069B (en) 2013-01-14 2019-12-10 Xencor股份有限公司 Novel heterodimeric proteins
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
CA2897987A1 (en) 2013-01-15 2014-07-24 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
WO2014114801A1 (en) 2013-01-25 2014-07-31 Amgen Inc. Antibodies targeting cdh19 for melanoma
JO3519B1 (en) 2013-01-25 2020-07-05 Amgen Inc Antibody constructs for CDH19 and CD3
WO2014164301A2 (en) 2013-03-11 2014-10-09 Amgen Inc. Protein formulations
US9546203B2 (en) 2013-03-14 2017-01-17 Amgen Inc. Aglycosylated Fc-containing polypeptides with cysteine substitutions
CA2904337A1 (en) 2013-03-14 2014-10-02 Amgen Inc. Chrdl-1 antigen binding proteins and methods of treatment
US20160017041A1 (en) 2013-03-15 2016-01-21 Biogen Ma Inc. Treatment and prevention of acute kidney injury using anti-alpha v beta 5 antibodies
DK2968503T3 (en) 2013-03-15 2018-12-03 Intrinsic Lifesciences Llc ANTI-HEPCIDIN ANTIBODIES AND APPLICATIONS THEREOF
US10035860B2 (en) 2013-03-15 2018-07-31 Biogen Ma Inc. Anti-alpha V beta 6 antibodies and uses thereof
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
JP6594855B2 (en) 2013-03-15 2019-10-23 ゼンコア インコーポレイテッド Heterodimeric protein
DK3611180T3 (en) 2013-03-15 2022-02-28 Biomolecular Holdings Llc HYBRID IMMUNOGLOBULIN CONTAINING NON-PEPTIDYL BINDING
WO2014145174A1 (en) 2013-03-15 2014-09-18 Biological Mimetics, Inc. Immunogenic human rhinovirus (hrv) compositions
WO2014140368A1 (en) 2013-03-15 2014-09-18 Amgen Research (Munich) Gmbh Antibody constructs for influenza m2 and cd3
AU2014232416B2 (en) 2013-03-15 2017-09-28 Xencor, Inc. Modulation of T Cells with Bispecific Antibodies and FC Fusions
ES2770976T3 (en) 2013-03-15 2020-07-06 Amgen Inc Human PAC1 antibodies
WO2014144553A1 (en) 2013-03-15 2014-09-18 Amgen Inc. Secreted frizzle-related protein 5 (sfrp5) binding proteins and methods of treatment
US10035859B2 (en) 2013-03-15 2018-07-31 Biogen Ma Inc. Anti-alpha V beta 6 antibodies and uses thereof
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
UY35484A (en) 2013-03-15 2014-10-31 Amgen Res Munich Gmbh Single chain binding molecule comprising N-end ABP
SG11201507429TA (en) 2013-03-15 2015-10-29 Genentech Inc Il-22 polypeptides and il-22 fc fusion proteins and methods of use
WO2014144292A2 (en) 2013-03-15 2014-09-18 Sanofi Pasteur Biologics , Llc Antibodies against clostridium difficile toxins and methods of using the same
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
US9260527B2 (en) 2013-03-15 2016-02-16 Sdix, Llc Anti-human CXCR4 antibodies and methods of making same
WO2014183885A1 (en) 2013-05-17 2014-11-20 INSERM (Institut National de la Santé et de la Recherche Médicale) Antagonist of the btla/hvem interaction for use in therapy
NZ713636A (en) 2013-05-30 2022-07-01 Kiniksa Pharmaceuticals Ltd Oncostatin m receptor antigen binding proteins
US20160237399A1 (en) 2015-02-18 2016-08-18 Biogen Ma Inc. Control of Protein Glycosylation by Culture Medium Supplementation and Cell Culture Process Parameters
EP3916081A3 (en) 2013-08-19 2022-03-23 Biogen MA Inc. Control of protein glycosylation by culture medium supplementation and cell culture process parameters
CN105658672A (en) 2013-08-22 2016-06-08 阿塞勒隆制药公司 TGF-beta receptor type II variants and uses thereof
TW201605896A (en) 2013-08-30 2016-02-16 安美基股份有限公司 GITR antigen binding proteins
AR097648A1 (en) 2013-09-13 2016-04-06 Amgen Inc COMBINATION OF EPIGENETIC FACTORS AND BIESPECTIVE COMPOUNDS THAT HAVE LIKE DIANA CD33 AND CD3 IN THE TREATMENT OF MYELOID LEUKEMIA
EP3757130A1 (en) 2013-09-26 2020-12-30 Costim Pharmaceuticals Inc. Methods for treating hematologic cancers
US10081682B2 (en) 2013-10-11 2018-09-25 Oxford Bio Therapeutics Ltd. Conjugated antibodies against LY75 for the treatment of cancer
WO2015057939A1 (en) 2013-10-18 2015-04-23 Biogen Idec Ma Inc. Anti-s1p4 antibodies and uses thereof
EA035481B1 (en) 2013-10-25 2020-06-23 Акселерон Фарма, Инк. Endoglin peptides to treat fibrotic diseases
ES2759252T3 (en) 2013-10-31 2020-05-08 Resolve Therapeutics Llc Nuclease-albumin fusions and therapeutic methods
CN104623637A (en) 2013-11-07 2015-05-20 健能隆医药技术(上海)有限公司 Application of IL-22 dimer in preparation of intravenous injection drugs
JP2017500017A (en) 2013-12-20 2017-01-05 バイオジェン・エムエイ・インコーポレイテッドBiogen MA Inc. Use of perfusion seed cultures to improve biopharmaceutical fed-batch production capacity and product quality
JP6590810B2 (en) 2013-12-24 2019-10-16 ヤンセン ファーマシューティカ エヌブイ Anti-VISTA antibodies and fragments
US11014987B2 (en) 2013-12-24 2021-05-25 Janssen Pharmaceutics Nv Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
RU2019119862A (en) 2014-02-20 2019-07-15 Аллерган, Инк. ANTIBODY TO C5 COMPLETE COMPONENT
SG11201606983SA (en) 2014-02-27 2016-09-29 Allergan Inc COMPLEMENT FACTOR Bb ANTIBODIES
US20150252434A1 (en) 2014-03-07 2015-09-10 University Health Network Methods and compositions for detection of targets involved in cancer metastasis
SG10201807877TA (en) 2014-03-14 2018-10-30 Daniel Capon Hybrid immunoglobulin containing non-peptidyl linkage
DK3122869T3 (en) 2014-03-24 2019-09-09 Biogen Ma Inc PROCEDURES FOR REDUCING GLUTAMINE DEPRESSION IN MAMMAL CULTURE CULTURE
EA201691925A1 (en) 2014-03-28 2017-06-30 Ксенкор, Инк. BISPECIFIC ANTIBODIES THAT BIND WITH CD38 AND CD3
FR3020063A1 (en) 2014-04-16 2015-10-23 Gamamabs Pharma ANTI-HER4 HUMAN ANTIBODY
US10544231B2 (en) 2014-04-16 2020-01-28 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies for the prevention or the treatment of bleeding episodes
BR112016025450A2 (en) 2014-05-02 2017-12-12 Medimmune Ltd ion channel modulators and uses thereof
KR102132144B1 (en) 2014-06-04 2020-07-09 악셀레론 파마 인코포레이티드 Methods and compositions for treatment of disorders with follistatin polypeptides
MX2016016310A (en) 2014-06-11 2017-10-20 A Green Kathy Use of vista agonists and antagonists to suppress or enhance humoral immunity.
CA3228982A1 (en) 2014-07-09 2016-01-14 Dsm Nutritional Products, Llc Oligosaccharide compositions and methods for producing thereof
MX2017001403A (en) 2014-07-31 2017-07-07 Amgen Res (Munich) Gmbh Bispecific single chain antibody construct with enhanced tissue distribution.
TW201609812A (en) 2014-07-31 2016-03-16 安美基研究(慕尼黑)公司 Optimized cross-species specific bispecific single chain antibody constructs
UY36245A (en) 2014-07-31 2016-01-29 Amgen Res Munich Gmbh ANTIBODY CONSTRUCTS FOR CDH19 AND CD3
US20170275344A1 (en) 2014-08-26 2017-09-28 Compugen Ltd. Polypeptides and uses thereof as a drug for treatment of autoimmune disorders
JP6672257B2 (en) 2014-08-27 2020-03-25 アムジエン・インコーポレーテツド Tissue metalloproteinase inhibitor type 3 (TIMP-3) variants, compositions and methods
WO2016040767A2 (en) 2014-09-12 2016-03-17 Amgen Inc. Chrdl-1 epitopes and antibodies
NZ730186A (en) 2014-09-22 2020-04-24 Intrinsic Lifesciences Llc Humanized anti-hepcidin antibodies and uses thereof
NZ729808A (en) 2014-09-30 2022-02-25 Neurimmune Holding Ag Human-derived anti-dipeptide repeats (dprs) antibody
MA41685A (en) 2014-10-17 2017-08-22 Biogen Ma Inc COPPER SUPPLEMENT FOR THE REGULATION OF GLYCOSYLATION IN A MAMMAL CELL CULTURE PROCESS
CA2965034C (en) 2014-10-24 2023-05-02 Astrazeneca Ab Combination of an immunomodulatory agent and an antisense oligonucleotide
AU2015339134B2 (en) 2014-10-30 2021-06-17 Acceleron Pharma Inc. Methods and compositions using GDF15 polypeptides for increasing red blood cells
MA40864A (en) 2014-10-31 2017-09-05 Biogen Ma Inc HYPOTAURINE, GABA, BETA-ALANINE AND CHOLINE FOR THE REGULATION OF THE ACCUMULATION OF RESIDUAL BY-PRODUCTS IN MAMMAL CELL CULTURE PROCESSES
WO2016069889A1 (en) 2014-10-31 2016-05-06 Resolve Therapeutics, Llc Therapeutic nuclease-transferrin fusions and methods
WO2016081835A2 (en) 2014-11-21 2016-05-26 University Of Maryland, Baltimore Targeted structure-specific particulate delivery systems
BR112017011092A2 (en) 2014-11-26 2017-12-26 Xencor Inc cd3-binding heterodimeric antibodies and tumor antigens
US10259887B2 (en) 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
PE20171103A1 (en) 2014-11-26 2017-08-07 Xencor Inc HETERODIMERIC ANTIBODIES THAT BIND TO CD3 AND CD38
HUE062189T2 (en) 2014-12-03 2023-09-28 Celgene Corp Activin-actrii antagonists and uses for treating myelodysplastic syndrome
MX2017007136A (en) 2014-12-05 2017-12-04 Immunext Inc Identification of vsig8 as the putative vista receptor and its use thereof to produce vista/vsig8 modulators.
WO2016094273A1 (en) 2014-12-08 2016-06-16 Dana-Farber Cancer Institute, Inc. Methods for upregulating immune responses using combinations of anti-rgmb and anti-pd-1 agents
WO2016105450A2 (en) 2014-12-22 2016-06-30 Xencor, Inc. Trispecific antibodies
TW202130662A (en) 2015-01-23 2021-08-16 法商賽諾菲公司 Anti-cd3 antibodies, anti-cd123 antibodies and bispecific antibodies specifically binding to cd3 and/or cd123
WO2016122887A1 (en) 2015-01-26 2016-08-04 Midori Usa, Inc. Oligosaccharide compositions for use animal feed and methods of producing thereof
WO2016141387A1 (en) 2015-03-05 2016-09-09 Xencor, Inc. Modulation of t cells with bispecific antibodies and fc fusions
WO2016161410A2 (en) 2015-04-03 2016-10-06 Xoma Technology Ltd. Treatment of cancer using inhibitors of tgf-beta and pd-1
US10851144B2 (en) 2015-04-10 2020-12-01 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
IL255076B2 (en) 2015-04-17 2023-10-01 Amgen Res Munich Gmbh Bispecific antibody constructs for cdh3 and cd3
US20160347848A1 (en) 2015-05-28 2016-12-01 Medimmune Limited Therapeutic combinations and methods for treating neoplasia
JP7026509B2 (en) 2015-06-24 2022-02-28 ヤンセン ファーマシューティカ エヌブイ Anti-VISTA antibody and fragment
DK3322733T3 (en) 2015-07-13 2021-10-18 Compugen Ltd HIDE1 COMPOSITIONS AND METHODS
TWI744242B (en) 2015-07-31 2021-11-01 德商安美基研究(慕尼黑)公司 Antibody constructs for egfrviii and cd3
TWI793062B (en) 2015-07-31 2023-02-21 德商安美基研究(慕尼黑)公司 Antibody constructs for dll3 and cd3
TWI717375B (en) 2015-07-31 2021-02-01 德商安美基研究(慕尼黑)公司 Antibody constructs for cd70 and cd3
TWI796283B (en) 2015-07-31 2023-03-21 德商安美基研究(慕尼黑)公司 Antibody constructs for msln and cd3
TWI829617B (en) 2015-07-31 2024-01-21 德商安美基研究(慕尼黑)公司 Antibody constructs for flt3 and cd3
AU2016301380B2 (en) 2015-08-04 2021-07-01 Acceleron Pharma Inc. Methods for treating myeloproliferative disorders
US10358497B2 (en) 2015-09-29 2019-07-23 Amgen Inc. Methods of treating cardiovascular disease with an ASGR inhibitor
WO2017066561A2 (en) 2015-10-16 2017-04-20 President And Fellows Of Harvard College Regulatory t cell pd-1 modulation for regulating t cell effector immune responses
US10227410B2 (en) 2015-12-07 2019-03-12 Xencor, Inc. Heterodimeric antibodies that bind CD3 and PSMA
TWI797073B (en) 2016-01-25 2023-04-01 德商安美基研究(慕尼黑)公司 Pharmaceutical composition comprising bispecific antibody constructs
TN2018000266A1 (en) 2016-02-03 2020-01-16 Amgen Res Munich Gmbh Psma and cd3 bispecific t cell engaging antibody constructs.
EA039859B1 (en) 2016-02-03 2022-03-21 Эмджен Рисерч (Мюник) Гмбх Bispecific antibody constructs binding egfrviii and cd3
MD3411402T2 (en) 2016-02-03 2022-05-31 Amgen Res Munich Gmbh BCMA and CD3 bispecific T cell engaging antibody constructs
TW202216201A (en) 2016-02-12 2022-05-01 比利時商楊森製藥公司 Anti-vista antibodies and fragments, uses thereof, and methods of identifying same
CN109328069B (en) 2016-04-15 2023-09-01 亿一生物医药开发(上海)有限公司 Use of IL-22 in the treatment of necrotizing enterocolitis
RU2021111187A (en) 2016-04-15 2021-04-29 Янссен Фармасьютикалз, Инк. ANTIBODIES AGAINST HUMAN VISTA AND THEIR APPLICATION
JOP20170091B1 (en) 2016-04-19 2021-08-17 Amgen Res Munich Gmbh Administration of a bispecific construct binding to CD33 and CD3 for use in a method for the treatment of myeloid leukemia
US11016085B2 (en) 2016-04-25 2021-05-25 The Johns Hopkins University ZNT8 assays for drug development and pharmaceutical compositions
EP3808764A1 (en) 2016-05-04 2021-04-21 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
WO2017209519A1 (en) 2016-05-31 2017-12-07 Mogam Institute For Biomedical Research Ab6 family designer ligands of tgf-beta superfamily
MX2018015592A (en) 2016-06-14 2019-04-24 Xencor Inc Bispecific checkpoint inhibitor antibodies.
JP7021127B2 (en) 2016-06-28 2022-02-16 ゼンコア インコーポレイテッド Heterodimer antibody that binds to somatostatin receptor 2
JP7308034B2 (en) 2016-07-01 2023-07-13 リゾルブ セラピューティクス, エルエルシー Optimized double nuclease fusions and methods
CA3030834A1 (en) 2016-07-15 2018-01-18 Takeda Pharmaceutical Company Limited Methods and materials for assessing response to plasmablast- and plasma cell-depleting therapies
TWI790206B (en) 2016-07-18 2023-01-21 法商賽諾菲公司 Bispecific antibody-like binding proteins specifically binding to cd3 and cd123
MA45715A (en) 2016-07-25 2019-05-29 Biogen Ma Inc ANTI-HSPA5 ANTIBODIES (GRP78) AND THEIR USES
JP7148493B2 (en) 2016-08-01 2022-10-05 ゾーマ (ユーエス) リミテッド ライアビリティ カンパニー Parathyroid hormone receptor 1 (PTH1R) antibodies and uses thereof
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
CN109996817A (en) 2016-09-15 2019-07-09 艾科赛扬制药股份有限公司 Reverse primitive gut embryogenesis polypeptide and application thereof
BR112019005944A2 (en) 2016-09-28 2019-06-11 Musc Foudation For Res Development antibodies that bind to interleukin 2 and their uses
WO2018064098A1 (en) 2016-09-28 2018-04-05 Cohbar, Inc. Therapeutic mots-c related peptides
AU2017338916B2 (en) 2016-10-05 2022-03-24 Acceleron Pharma Inc. Variant ActRIIB proteins and uses thereof
MX2019004327A (en) 2016-10-14 2019-10-14 Xencor Inc Bispecific heterodimeric fusion proteins containing il-15/il-15ralpha fc-fusion proteins and pd-1 antibody fragments.
TWI778985B (en) 2016-10-20 2022-10-01 法商賽諾菲公司 Anti-chikv antibodies and uses thereof
SI3565828T1 (en) 2017-01-05 2022-04-29 Kahr Medical Ltd. A sirp1 alpha-41bbl fusion protein and methods of use thereof
WO2018127917A1 (en) 2017-01-05 2018-07-12 Kahr Medical Ltd. A pd1-41bbl fusion protein and methods of use thereof
JOP20190189A1 (en) 2017-02-02 2019-08-01 Amgen Res Munich Gmbh Low ph pharmaceutical composition comprising t cell engaging antibody constructs
WO2018152496A1 (en) 2017-02-17 2018-08-23 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Compositions and methods for the diagnosis and treatment of zika virus infection
US11117930B2 (en) 2017-02-23 2021-09-14 Adrx, Inc. Peptide inhibitors of transcription factor aggregation
CA3051640A1 (en) 2017-03-16 2018-09-20 Innate Pharma Compositions and methods for treating cancer
EP3615569A1 (en) 2017-04-25 2020-03-04 The U.S.A. As Represented By The Secretary, Department Of Health And Human Services Antibodies and methods for the diagnosis and treatment of epstein barr virus infection
SI3628049T1 (en) 2017-05-04 2023-10-30 Acceleron Pharma Inc. Tgf-beta receptor type ii fusion proteins and uses thereof
BR112019022751A2 (en) 2017-05-05 2020-05-19 Amgen Inc pharmaceutical composition comprising bispecific antibody constructs for improved storage and administration
CN110997693A (en) 2017-06-07 2020-04-10 阿德克斯公司 Tau aggregation inhibitors
AU2018291497A1 (en) 2017-06-30 2020-01-16 Xencor, Inc. Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15Ra and antigen binding domains
WO2019014044A1 (en) 2017-07-12 2019-01-17 The Johns Hopkins University A proteoliposome-based znt8 self-antigen for type 1 diabetes diagnosis
WO2019018629A1 (en) 2017-07-19 2019-01-24 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Antibodies and methods for the diagnosis and treatment of hepatitis b virus infection
DK3661954T3 (en) 2017-08-03 2022-04-19 Amgen Inc INTERLEUKIN-21 MUTEINS AND METHODS OF TREATMENT
EP3668886A2 (en) 2017-08-18 2020-06-24 Adrx, Inc. Tau aggregation peptide inhibitors
CN111315767A (en) 2017-08-22 2020-06-19 萨纳生物有限责任公司 Soluble interferon receptors and uses thereof
EP3679040B1 (en) 2017-09-08 2022-08-03 Amgen Inc. Inhibitors of kras g12c and methods of using the same
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
AU2018366199A1 (en) 2017-11-08 2020-05-28 Xencor, Inc. Bispecific and monospecific antibodies using novel anti-PD-1 sequences
EA202091422A1 (en) 2017-12-11 2020-08-28 Эмджен Инк. METHOD FOR CONTINUOUS PRODUCTION OF PRODUCTS BASED ON BISPECIFIC ANTIBODIES
JP2021506291A (en) 2017-12-19 2021-02-22 ゼンコア インコーポレイテッド Modified IL-2 FC fusion protein
TW201940518A (en) 2017-12-29 2019-10-16 美商安進公司 Bispecific antibody construct directed to MUC17 and CD3
KR20200110358A (en) 2018-01-12 2020-09-23 암젠 인크 Anti-PD-1 Antibodies and Methods of Treatment
EP3743437A1 (en) 2018-01-26 2020-12-02 F. Hoffmann-La Roche AG Il-22 fc fusion proteins and methods of use
DK3743088T3 (en) 2018-01-26 2022-12-19 Hoffmann La Roche IL-22 Fc compositions and methods of use
KR20200135331A (en) 2018-02-14 2020-12-02 비엘라 바이오, 인크. Antibodies to Feline McDonough's Sarcoma (FMS)-Like Tyrosine Kinase 3 Receptor Ligand (FLT3L) and Its Use to Treat Autoimmune and Inflammatory Diseases
CA3091139A1 (en) 2018-02-21 2019-08-29 Genentech, Inc. Dosing for treatment with il-22 fc fusion proteins
US10982006B2 (en) 2018-04-04 2021-04-20 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
SG11202010163QA (en) 2018-04-18 2020-11-27 Xencor Inc Pd-1 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and pd-1 antigen binding domains and uses thereof
CA3097741A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Tim-3 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and tim-3 antigen binding domains
WO2019213416A1 (en) 2018-05-02 2019-11-07 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Antibodies and methods for the diagnosis, prevention, and treatment of epstein barr virus infection
EA202092518A1 (en) 2018-06-18 2021-08-23 Иннейт Фарма COMPOSITIONS AND METHODS FOR TREATMENT OF CANCER
US11530274B2 (en) 2018-07-02 2022-12-20 Amgen Inc. Anti-STEAP1 antigen-binding protein
CN112543773B (en) 2018-07-11 2024-05-31 卡尔医学有限公司 SIRP alpha-4-1 BBL variant fusion proteins and methods of use thereof
JP2021532140A (en) 2018-07-30 2021-11-25 アムジェン リサーチ (ミュニック) ゲゼルシャフト ミット ベシュレンクテル ハフツング Long-term administration of bispecific antibody constructs that bind to CD33 and CD3
AU2019314999A1 (en) 2018-08-03 2021-02-11 Amgen Inc. Antibody constructs for CLDN18.2 and CD3
US20210238279A1 (en) 2018-08-16 2021-08-05 The Johns Hopkins University Antibodies to human znt8
EP3861016A2 (en) 2018-10-03 2021-08-11 Xencor, Inc. Il-12 heterodimeric fc-fusion proteins
AU2019356564A1 (en) 2018-10-11 2021-04-29 Amgen Inc. Downstream processing of bispecific antibody constructs
BR112021013096A2 (en) 2019-01-04 2022-04-19 Resolve Therapeutics, Llc TREATMENT OF SJÖGREN'S DISEASE WITH NUCLEASE FUSION PROTEINS
MX2021008979A (en) 2019-01-28 2021-11-12 Cohbar Inc Therapeutic peptides.
EP3920972A1 (en) 2019-02-07 2021-12-15 Sanofi Use of anti-ceacam5 immunoconjugates for treating lung cancer
EP3693023A1 (en) 2019-02-11 2020-08-12 Sanofi Use of anti-ceacam5 immunoconjugates for treating lung cancer
AU2020223031A1 (en) 2019-02-12 2021-08-19 Ambrx, Inc. Compositions containing, methods and uses of antibody-TLR agonist conjugates
CN114173875A (en) 2019-03-01 2022-03-11 Xencor股份有限公司 Heterodimeric antibodies that bind ENPP3 and CD3
CA3131705A1 (en) 2019-03-27 2020-10-01 Umc Utrecht Holding B.V. Engineered iga antibodies and methods of use
AU2020272766A1 (en) 2019-04-08 2021-11-18 Biogen Ma Inc. Anti-integrin antibodies and uses thereof
US11180561B2 (en) 2019-04-09 2021-11-23 Abcuro, Inc. Killer cell lectin-like receptor subfamily G member 1 (KLRG1) depleting antibodies
US20220289854A1 (en) 2019-04-30 2022-09-15 Dana-Farber Cancer Institute, Inc. Methods for treating cancer using combinations of anti-cx3cr1 and immune checkpoint blockade agents
TW202045711A (en) 2019-06-13 2020-12-16 美商安進公司 Automated biomass-based perfusion control in the manufacturing of biologics
CA3146248A1 (en) 2019-07-11 2021-01-14 Kahr Medical Ltd. Heterodimers and methods of use thereof
WO2021023624A1 (en) 2019-08-02 2021-02-11 Orega Biotech Novel il-17b antibodies
US20210032370A1 (en) 2019-08-02 2021-02-04 Immatics Biotechnologies Gmbh Recruiting agent further binding an mhc molecule
DE102019121007A1 (en) 2019-08-02 2021-02-04 Immatics Biotechnologies Gmbh Antigen binding proteins that specifically bind to MAGE-A
EP4385573A2 (en) 2019-08-13 2024-06-19 Amgen Inc. Interleukin-2 muteins for the expansion of t-regulatory cells
CA3156683A1 (en) 2019-11-13 2021-05-20 Amgen Inc. Method for reduced aggregate formation in downstream processing of bispecific antigen-binding molecules
US20220395553A1 (en) 2019-11-14 2022-12-15 Cohbar, Inc. Cxcr4 antagonist peptides
CA3162705A1 (en) 2019-12-17 2021-06-24 Amgen Inc. Dual interleukin-2 /tnf receptor agonist for use in therapy
EP4090365A1 (en) 2020-01-15 2022-11-23 Immatics Biotechnologies GmbH Antigen binding proteins specifically binding prame
US20230093169A1 (en) 2020-01-22 2023-03-23 Amgen Research (Munch) Gmbh Combinations of antibody constructs and inhibitors of cytokine release syndrome and uses thereof
WO2021183861A1 (en) 2020-03-12 2021-09-16 Amgen Inc. Method for treatment and prophylaxis of crs in patients comprising a combination of bispecifc antibodies binding to cds x cancer cell and tnfalpha or il-6 inhibitor
WO2021207662A1 (en) 2020-04-10 2021-10-14 Genentech, Inc. Use of il-22fc for the treatment or prevention of pneumonia, acute respiratory distress syndrome, or cytokine release syndrome
WO2021231976A1 (en) 2020-05-14 2021-11-18 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (psma) and cd3
AU2021275049A1 (en) 2020-05-19 2022-12-22 Amgen Inc. MAGEB2 binding constructs
US20240209085A1 (en) 2020-05-29 2024-06-27 Amgen Inc. Adverse effects-mitigating administration of a bispecific construct binding to cd33 and cd3
CN116670285A (en) 2020-06-23 2023-08-29 江苏康缘药业股份有限公司 anti-CD 38 antibodies and uses thereof
CA3165342A1 (en) 2020-06-29 2022-01-06 James Arthur Posada Treatment of sjogren's syndrome with nuclease fusion proteins
TW202216778A (en) 2020-07-15 2022-05-01 美商安進公司 Tigit and cd112r blockade
CA3192204A1 (en) 2020-08-19 2022-02-24 Xencor, Inc. Anti-cd28 and/or anti-b7h3 compositions
EP4199968A1 (en) 2020-08-20 2023-06-28 Ambrx, Inc. Antibody-tlr agonist conjugates, methods and uses thereof
CN116113439A (en) 2020-09-04 2023-05-12 默克专利股份公司 anti-CEACAM 5 antibodies and conjugates and uses thereof
EP4240407A1 (en) 2020-11-06 2023-09-13 Amgen Inc. Antigen binding domain with reduced clipping rate
WO2022096700A1 (en) 2020-11-06 2022-05-12 Amgen Research (Munich) Gmbh Polypeptide constructs selectively binding to cldn6 and cd3
JP2023547661A (en) 2020-11-06 2023-11-13 アムジエン・インコーポレーテツド Polypeptide constructs that bind to CD3
WO2022096716A2 (en) 2020-11-06 2022-05-12 Amgen Inc. Multitargeting bispecific antigen-binding molecules of increased selectivity
JP2024504696A (en) 2021-01-20 2024-02-01 バイオアントレ エルエルシー CTLA4-binding proteins and methods of treating cancer
CA3212665A1 (en) 2021-03-09 2022-09-15 Xencor, Inc. Heterodimeric antibodies that bind cd3 and cldn6
KR20230154311A (en) 2021-03-10 2023-11-07 젠코어 인코포레이티드 Heterodimeric antibodies binding to CD3 and GPC3
CA3215594A1 (en) 2021-04-02 2022-10-06 Agnieszka KIELCZEWSKA Mageb2 binding constructs
MX2023011480A (en) 2021-04-03 2023-12-06 Ambrx Inc Anti-her2 antibody-drug conjugates and uses thereof.
CA3217180A1 (en) 2021-05-06 2022-11-10 Amgen Research (Munich) Gmbh Cd20 and cd22 targeting antigen-binding molecules for use in proliferative diseases
WO2022261183A2 (en) 2021-06-08 2022-12-15 Dana-Farber Cancer Institute, Inc. Compositions and methods for treating and/or identifying an agent for treating intestinal cancers
MX2024001277A (en) 2021-07-26 2024-02-15 Abcuro Inc Killer cell lectin-like receptor subfamily g member 1 (klrg1) depleting antibodies.
WO2023097119A2 (en) 2021-11-29 2023-06-01 Dana-Farber Cancer Institute, Inc. Methods and compositions to modulate riok2
CA3239856A1 (en) 2021-12-02 2023-06-08 Mustapha CHADJAA Cea assay for patient selection in cancer therapy
IL313223A (en) 2021-12-02 2024-07-01 Sanofi Sa Ceacam5 adc–anti-pd1/pd-l1 combination therapy
WO2023137161A1 (en) 2022-01-14 2023-07-20 Amgen Inc. Triple blockade of tigit, cd112r, and pd-l1
WO2023172968A1 (en) 2022-03-09 2023-09-14 Merck Patent Gmbh Anti-gd2 antibodies, immunoconjugates and therapeutic uses thereof
TW202346354A (en) 2022-03-09 2023-12-01 德商馬克專利公司 Anti-ceacam5 antibodies and conjugates and uses thereof
WO2023218027A1 (en) 2022-05-12 2023-11-16 Amgen Research (Munich) Gmbh Multichain multitargeting bispecific antigen-binding molecules of increased selectivity
WO2023240287A1 (en) 2022-06-10 2023-12-14 Bioentre Llc Combinations of ctla4 binding proteins and methods of treating cancer
WO2024059675A2 (en) 2022-09-14 2024-03-21 Amgen Inc. Bispecific molecule stabilizing composition
WO2024126431A1 (en) 2022-12-12 2024-06-20 Horizon Therapeutics Ireland Dac Anti-ilt7 binding agents for the treatment and prevention of myositis
WO2024147934A1 (en) 2023-01-06 2024-07-11 Takeda Pharmaceutical Company Limited Anti-cd38 antibodies for the treatment of autoimmune diseases

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3847890A (en) * 1971-11-01 1974-11-12 A Green Acidic monosaccharide-substituted proteins

Also Published As

Publication number Publication date
AU5627186A (en) 1987-09-28
EP0272253A1 (en) 1988-06-29
WO1987005330A1 (en) 1987-09-11
JPS63502716A (en) 1988-10-13
AU5133690A (en) 1990-08-23
DK583087D0 (en) 1987-11-06
EP0272253A4 (en) 1990-02-05
DK583087A (en) 1987-11-06
AU624487B2 (en) 1992-06-11

Similar Documents

Publication Publication Date Title
AU597574B2 (en) Method for enhancing glycoprotein stability
US4925796A (en) Method for enhancing glycoprotein stability
US5272066A (en) Synthetic method for enhancing glycoprotein stability
EP0799318B1 (en) Methods of modifying carbohydrate moieties
Schachter Glycoproteins: their structure, biosynthesis and possible clinical implications
EP2682473B1 (en) Method for producing sialic-acid-containing sugar chain
Staneloni et al. The biosynthetic pathway of the asparagine-linked oligosaccharides of glycoproteins
Watt et al. Enzyme-catalyzed formation of glycosidic linkages
Nakata et al. Structural study of the sugar chains of CD36 purified from bovine mammary epithelial cells: Occurrence of novel hybrid-type sugar chains containing the Neu5Ac. alpha. 2. fwdarw. 6GalNAc. beta. 1. fwdarw. 4GlcNAc and the Man. alpha. 1. fwdarw. 2Man. alpha. 1. fwdarw. 3Man. alpha. 1. fwdarw. 6Man groups
Schachter et al. Oligosaccharide branching of glycoproteins: biosynthetic mechanisms and possible biological functions
Endo et al. Structures of the asparagine-linked sugar chain of glucose transporter from human erythrocytes
Mendicino et al. Isolation and properties of. alpha.-D-mannose:. beta.-1, 2-N-acetylglucosaminyltransferase from trachea mucosa
Bahl et al. Characterization of glycoproteins: carbohydrate structures of glycoprotein hormones
Schachter et al. Glycosyltransferases involved in the biosynthesis of protein-bound oligosaccharides of the asparagine-N-acetyl-D-glucosamine and serine (threonine)-N-acetyl-D-galactosamine types
Thayer et al. Enzymatic synthesis of glycopeptides and glycoproteins
JP3811527B2 (en) Process for producing new glycoconjugates
JP3732871B2 (en) Method for producing complex carbohydrate
WO2023202991A2 (en) Cell-free enzymatic method for preparation of n-glycans
CA1340658C (en) Preparation of 1-amino-1-deoxyoligosaccharides and derivatives thereof
Paulsen et al. Structure, Metabolism and Function of Glycoconjugates
Eijnden et al. SIALYLTRANSFERASES; THEIR SPECIFICITY AND THEIR USE IN CARBOHYDRATE REMODELLING.
Rahman Evaluation of the enzymes in the N-glycosylation pathways of (native and recombinant) insect cells